<<

Karam Aboudehen, Ph.D.

UNIVERSITY OF MINNESOTA, MINNEAPOLIS, MN

Deregulation of Long Noncoding RNA in the Pathogenesis of Autosomal Dominant Polycystic Kidney

Polycystic kidney disease (PKD) is characterized by the formation of cysts, which originate from the epithelial tubules of the nephron. Progressive growth of the cysts causes damage and loss of functional nephrons, ultimately leading to end-stage renal failure. The of PKD is incompletely understood, and only one FDA-approved treatment (tolvaptan) exists today. Long noncoding RNAs (lncRNA) – defined by a length >200 and absence of a long open reading frame – are a class of non--coding RNAs implicated in a range of . The nature and extent of involvement of lncRNAs in PKD was not previously investigated. Utilizing two independent PKD mouse models, the applicant identified Hoxb3os, a highly conserved lncRNA, which was downregulated in mouse and human ADPKD. Deletion of Hoxb3os in kidney cells resulted in increased phosphorylation of mTOR and its downstream targets. Consistent with activation of mTOR signaling, Hoxb3os mutant cells displayed increased oxidative phosphorylation, increased proliferation, and decreased autophagy. The Hoxb3os mutant phenotype was partially rescued upon re- expressing wild-type Hoxb3os in knockout cells. Importantly, deletion of Hoxb3os in wild-type mice recapitulated the in vitro molecular phenotype and resulted in increased mTOR phosphorylation and subsequent increased cell proliferation, and defective autophagy. The overarching hypothesis for this K01 project is that downregulation of Hoxb3os exacerbates cyst formation and/or disease progression by dysregulating multiple pathways, including mTOR signaling. To test this hypothesis, the applicant will decipher the mechanism by which Hoxb3os inhibits mTOR signaling, identify novel Hoxb3os-regulated pathways, and determine the contribution of Hoxb3os to cyst formation in a mouse model of ADPKD. The applicant has assembled an interdisciplinary team of senior investigators to guide the proposed and provide mentorship during his transition to independence. He will have full access to the University's shared resources (mouse genetics laboratory, high-throughput sequencing, mass spectrometry and proteomics, bioinformatics). The applicant's training plan includes mentorship in core technique and concepts, generating and analyzing data, publishing and presenting results, completing coursework (in mouse genetics, biostatistics, RNA , computational methods, bioinformatics), and developing other requisite skills (in leadership, grant proposal development) needed to thrive as an independent investigator. Results from the proposed studies will form the basis for an R01 application to be submitted in the fourth year of this career development award. The applicant's long-term goal is to dedicate his career to advancing basic and translational research on cystic kidney disease as an independent investigator at an academic institution. As a kidney researcher with a deep interest and proven track record in molecular biology, he is uniquely positioned to answer the questions set forth in this proposal and to rapidly advance the mechanistic understanding of lncRNAs in PKD pathogenesis. Maisam Abu-El-Haija, M.D.

CINCINNATI CHILDRENS HOSP MED CTR, CINCINNATI, OH

Predicting Severity and Improving the Outcomes of Pediatric Pancreatitis

Project Summary/Abstract

The of pediatric Acute Pancreatitis (AP) has been rising to 1/10,000 cases, close to adult incidence. AP in pediatrics remains understudied with outcomes poorly defined. Previous studies in pediatric AP are mostly single-centered and retrospective in nature, and thereby insufficient for understanding the natural history. A high percentage (15-30%) of patients develop severe acute pancreatitis (SAP) with increased morbidity, increased length of hospital stay and cost. To date, there is no pediatric AP study that examines progression prospectively like we plan through our design, there is no validated pediatric prognostic severity system to improve outcomes of AP. Diabetes can result from AP in a subset of patients, and that leads to increased morbidity especially if underdiagnosed, given that there is no current method for screening post AP. The primary goal of my proposed career development is to acquire additional comprehensive training in biostatistics, study design, and outcome research through risk modeling, to acquire the skills necessary to build multicenter collaborations to study AP risk stratification and management that lead to the improved outcomes and decreased morbidity and mortality. I am an Assistant Professor of Pediatrics and a board certified Gastroenterologist in the Division of Gastroenterology, Hepatology and Nutrition in the Department of Pediatrics at Cincinnati Children’s Hospital Medical Center (CCHMC). CCHMC is committed to improving the health outcomes of children through innovative research. The exceptional environment at CCHMC facilitates the development of young investigators by having essential elements through core resources, expert mentorship, as well as the potential for internal collaboration. The pancreas Care Center at CCHMC is one of only a few pediatric pancreas centers in the United States and has an established referral pattern, with 70-90 new patients referred per year. My mentors Dr. Lee A. Denson- a nationally recognized leader in pediatric gastroenterology, Dr. Sohail Husain- a national expert and scientist in pediatric pancreatology, Dr. Woo- an expert in epidemiology and outcome research are invested in my training and guiding my path to be an independent investigators. The primary objective of this project is to improve outcomes of AP by decreasing SAP and improve screening for prediabetes. This will be accomplished through our prospective design. Specific Aim 1 to validate and optimize our previously generated SAP model by adding more factors, applying the new SAP definition, incorporating therapy effect, and incorporating MMPs and TIMPs as novel biomarkers. Specific Aim 2 to build a model to predict prediabetes post AP and understand the role of expanded testing in disease progression to prediabetes. Our proposal will help us better understand SAP and progression to prediabetes to fill the knowledge gap. Successful completion of this study has the potential to lead to improved outcomes by building clinical tools that facilitate management, and potentially lead to targeted therapies for AP in pediatrics. A. Lenore Ackerman, Ph.D., M.D.

CEDARS-SINAI MEDICAL CENTER, LOS ANGELES, CA

The Urinary Microbiota and Host Inflammation in Lower Urinary Tract Symptoms

PROJECT SUMMARY

Storage lower urinary tract symptoms (LUTS), which include urinary urgency, frequency, nocturia, painful urination, and bladder pressure/discomfort, refer to patient experiences when the bladder is unable hold urine appropriately. These highly prevalent symptoms are chronic and debilitating, substantially degrading physical activity and quality of life. Yet despite the heavy burden of storage LUTS on public health, little is understood of the pathophysiology of these symptoms, limiting diagnosis, treatment, and prevention options. Humans harbor diverse microbial communities that live in symbiosis with healthy hosts but are frequently altered in disease. The role of these alterations is unclear, but mounting research suggests that microbial components may interact with human tissue to alter organ function, tissue permeability, and even central nervous system responsivity. We and others have used novel, state-of-the-art DNA sequencing methods to identify bacteria and fungi residing within the urinary tract and describe global differences in urinary microbial communities in patients with storage LUTS. We have yet to understand how these differences impact bladder pathophysiology, but our preliminary data suggest that shifts in these microbial communities underlie or reflect storage LUTS symptoms and correlate with increased local and systemic inflammation. We postulate that interactions of these changed communities with the host alter local and systemic inflammation and increase immunologic activation of bladder urothelial cells, generating inflammatory signatures characteristic of specific urinary symptoms. Based on similarities to other inflammatory diseases, we hypothesize that this inflammation becomes pathogenic in susceptible hosts with dysregulated microbial recognition, possibly mediated by genetic differences in host responsiveness to microbial components. Using state- of-the-art microbial profiling techniques, we will identify changes in urinary bacterial and fungal communities linked to storage LUTS in patients. We will also identify disease-associated variations in inflammatory markers and urothelial activation and associate these findings with specific microbial signatures and symptom patterns. We will perform a targeted characterization of genetic polymorphisms associated with dysregulated inflammatory responses to microbes to explore the contribution of host susceptibility in these conditions. Few previous studies have examined the urinary microbiota; this proposal is the first to integrate multi-omic datasets with clinical metadata to allow the discovery of clinically useful disease markers, microbial and inflammatory, and place them into the context of disease mechanisms and host risk factors. This study may promote a more comprehensive understanding of storage LUTS pathogenesis, identifying the molecular pathways that could serve as targets of new therapeutic agents. Positive results from this study would have significant implications for IC/PBS diagnosis and treatment and provide an important starting point for further studies examining the pathophysiology of this challenging and refractory disease. Andres Acosta, Ph.D., M.D.

MAYO CLINIC ROCHESTER, ROCHESTER, MN

Mechanisms of Enteroendocrine L-Cell Dysfunction and Bile Adaptations to Weight Loss

ABSTRACT

In this Mentored Career Development Award (K23) proposal, this candidate proposes to solidify a basic science foundation and acquired during his PhD and post-doctoral work with a long-term goal of becoming an academic authority in enteroendocrine L-cell regulation of satiety in obesity. The candidate aims to acquire a strong foundation in clinical translation science and to acquire a set of novel complementary skills necessary for an independent research career in gastrointestinal regulation of food intake. Understanding pathophysiology of human obesity is limited by the heterogeneity of patients' phenotype and multiple etiological mechanisms. These factors contribute to the highly variable inter-individual weight loss response to all interventions. Recently, we identified a sub-population of obesity with significantly decreased satiety (defined as lack of sensation of feeling full or rapid return of hunger). Our preliminary data shows that, in addition to decreased perception of fullness after a meal, these individuals, compared to other obesity phenotypes, have very low postprandial levels of the GI satiety hormones GLP-1 and PYY which are secreted by enteroendocrine (EE) L-cells. The low levels of the satiety hormone PYY and possibly other EE cell products suggests that the increased calorie intake originates from a deficit in the gut hormones, and this phenotype is summarized as a “hungry gut”. The suboptimal EE cell function, which could be mediated by altered synthesis or secretion by the EE cells themselves, or a decrease in luminal concentrations of molecules that normally stimulate EE cells, such as amino acids, bile acids, or short-chain fatty acids. Our preliminary results also suggest that these individuals have lower levels of FGF-19, a surrogate of the luminal bile acid concentration. Based on these preliminary studies, it is essential to understand the mechanisms that are responsible for the deficient EE signals that lead to the “hungry gut” phenotype of obesity. To test this concept, we propose the following two aims: 1) to study the mechanism of the hungry gut phenotype down to the level of the EE cells, and compare these findings to other obesity phenotypes and normal weight, healthy controls. 2) To restore normal satiety by increasing enteroendocrine cell function and secretion in patients after Roux-in-y gastric bypass surgery or in response to treatment with luminal bile acids. These findings will provide the foundation for further studies of the “hungry gut” phenotype in a future R01 application. This work will be performed in an academically nurturing environment within Mayo Clinic and with full support of the Division of Gastroenterology. The candidate will be guided by a strong mentorship committee (Drs. Michael Camilleri, Nicholas LaRusso and Adrian Vella). As a result of this work, the candidate will significantly advance our understanding of EE cell function in human obesity and develop his career into an independent physician scientist. Shivani Agarwal, M.D.

ALBERT EINSTEIN COLLEGE OF MEDICINE, BRONX, NY

Identifying and Reducing Disparities in Outcomes Among Economically Vulnerable and Minority Young Adults with Type 1 Diabetes

There is a critical need to further understand disparities in care and improve outcomes in young adults (YA) with type 1 diabetes (T1D) of low socioeconomic (SES) and minority status. Only 13% of YA (ages 18-30) with T1D nationally achieve the American Diabetes Association-recommended HbA1c target of <7.0%, substantially increasing long-term risk exposure and mortality later in life. Young adults have unique challenges, including the developmental transition to adulthood and healthcare transition to adult care, making it a critical period to intervene early. Compared to their white peers, YA with T1D of low SES and minority race/ethnicity are at disproportionately higher risk for adverse outcomes, including worse glycemic control and increased rates of recurrent diabetic ketaoacidosis and psychiatric comorbidity. This may be in part due to unrecognized and unaddressed social determinants of health. Despite these stark discrepancies and poor overall outcomes in YA, very little research has focused on these high-risk YA with T1D. Thus, the overall objectives of this proposal are to 1) identify social determinants of health that contribute to poor outcomes in high-risk YA with T1D and 2) develop and implement innovative tailored solutions to address disparities that extend beyond the traditional healthcare environment. Shivani Agarwal, M.D., M.P.H. is an adult endocrinologist and early diabetes and health services researcher who has previously developed a successful adult-based healthcare transition intervention for YA with T1D, which will be enhanced to address high-risk YA needs based on information gleaned from this proposal. In Specific Aim 1, data from YA in the national T1D Exchange Clinic Network will be used to identify social determinants that impact glycemic control in low SES and minority compared to higher SES non-minority YA with T1D. In Specific Aim 2, qualitative interviews will be conducted locally to determine care-related needs and challenges for high-risk YA with T1D, and solicit potential solutions. In Specific Aim 3, based upon an improved understanding of how social determinants affect care engagement, care processes, and glycemic control in high-risk YA with T1D, Dr. Agarwal will use a multidisciplinary multiple stakeholder advisory panel to enhance her transition intervention model to address disparities. She will also test the enhanced intervention’s efficacy on retention in adult care in a pilot randomized controlled trial. When complete, these studies will offer new insight on meaningful, addressable determinants of health in high-risk YA with T1D, to inform a new model for healthcare transition to address disparities. This work will form the basis for an R01 application of a multi-site randomized controlled trial testing the efficacy of a comprehensive diabetes transition program against usual care. Dr. Agarwal seeks to acquire important skills in disparities and diabetes investigation in addition to quantitative, qualitative, and intervention methods through the proposed career development award. Her ultimate goal is to become an independent disparities and health services investigator focused on improving health in young adult diabetes populations.

Veeral Ajmera, M.D.

UNIVERSITY OF CALIFORNIA, SAN DIEGO, LA JOLLA, CA

A Novel Multimodal Approach to Characterize NAFLD Severity and Prognosis

PROJECT SUMMARY

Nonalcoholic fatty liver disease (NAFLD) is an increasingly common cause of cirrhosis and on pace to be the leading indication for liver transplantation in the United States.(1, 2) NAFLD presents as a spectrum of disease ranging from isolated steatosis, which portends little risk of significant morbidity, to nonalcoholic steatohepatitis (NASH), which is characterized by inflammation and cell death and has substantial risk of progression to cirrhosis and liver-related mortality.(3) Unfortunately, liver biopsy remains the only way to accurately discriminate between isolated steatosis and NASH; however the procedure is invasive and remains impractical to scale to the estimated affected population of 60 million adults in the United States. Attempts to use individual or small combinations of biomarkers to characterize risk in NAFLD have been largely unsuccessful leaving a tremendous need for non-invasive risk stratification. My central hypothesis is that distinct subtypes of NAFLD can be identified by combining multiple non-invasive biomarkers, genetic and clinical factors using advanced analytic techniques for high dimensional data. Through my collaboration with the NIH-funded, multicenter NASH Clinical Research Network (NASH CRN) I explored the association between 28 putative plasma biomarkers and NAFLD histology and found that small sets of biomarkers were limited in discriminating between clinically significant stages of histologic severity. However, by applying a novel statistical technique, latent class analysis (LCA), we generated preliminary data identifying distinct subgroups of patients with NAFLD that are strongly associated with histologic severity. The research goal of this application is to (1) combine clinical and dietary factors, genetic markers and an expanded set of plasma biomarkers to refine distinct phenotypes of NAFLD using LCA, (2) validate the association between LCA defined phenotypes and histologic severity in an independent cohort with biopsy proven NAFLD, (3) build on an existing longitudinal cohort and test the ability of these phenotypes to predict progression of fibrosis and inflammation. My long-term goal is to combine expertise in multimodal, non- invasive biomarkers of NAFLD with advanced analytic techniques to personalize the management and treatment of patients with NAFLD. In order to accomplish this goal, I have assembled an exceptional mentorship team including my primary mentor, Dr. Rohit Loomba, who is an internationally renowned expert in NAFLD and Director of the UCSD NAFLD Research Center. In addition, Dr. Ariel Feldstein, Chief of the Division of Pediatric Gastroenterology, and an expert in translating NAFLD pathophysiology into biomarker development will serve as a co-mentor. Professor Lily Xu, biostatistical director of the UCSD Clinical and Translational Research Institute, will serve as my biostatistical mentor. Together, we formed a four-fold career development plan to gain expertise in (1) cohort development, biobanking and advanced NAFLD phenotyping, (2) statistical analysis of genetic and high dimensional data, (3) NAFLD pathobiology and biomarker development, and (4) research dissemination and the development of national recognition in the non-invasive assessment of NAFLD. Oleh Akchurin, M.D.

WEILL MEDICAL COLL OF CORNELL UNIV, NEW YORK, NY

Iron and renal fibrosis in juvenile chronic kidney disease

ABSTRACT

This is an application for a K08 Mentored Clinical Scientist Research Career Development Award for Dr. Oleh Akchurin, a pediatric nephrologist at Weill Cornell Medical College, New York, NY. Dr. Akchurin is establishing himself as a young investigator and translational researcher in the field of pediatric chronic kidney disease (CKD). This award will provide him with the support necessary to accomplish the following goals: (1) to become an expert in translational research of juvenile CKD, with a focus on altered iron metabolism; (2) to conduct experimental investigations of the effects of iron therapy for renal anemia in juvenile CKD on renal fibrosis, as mediated by changes in autophagy; (3) to implement advanced laboratory techniques in these studies; (4) to advance skills in interpreting and analyzing scientific data; and (5) to develop an independent research career. To achieve these goals, Dr. Akchurin has assembled a team of accomplished investigators: his primary mentor Dr. Mary E. Choi, a nephrologist with expertise in renal fibrosis and autophagy; co-mentor Dr. Stefano Rivella, an expert in iron metabolism; and consultants Dr. Surya Seshan, a world-renowned renal pathologist, and Dr. Xuejun Jiang, an expert in iron-dependent autophagy and pathways, as well as members of his Advisory Committee. Although iron therapy is very common in children with CKD and the detrimental action of iron has been well established in acute kidney injury, the role of altered iron metabolism and iron therapy in renal fibrosis in the sphere of juvenile CKD remains unknown. Similarly, while we understand that iron's pro-oxidative activity has the potential to damage intracellular macromolecules which need to be recycled by autophagy, the role of iron in modulating pro-survival autophagic activity in renal fibrosis has not been examined. Recent studies by Dr. Akchurin strongly support the hypothesis that oral iron given at current therapeutic levels causes kidney iron accumulation and exacerbates renal fibrosis in juvenile CKD, as mediated in part by altered renal autophagy. During the period of this award, he will test this hypothesis using an adenine-based mouse model of juvenile CKD. In addition, Dr. Akchurin will investigate the mechanism of his findings, specifically targeting the role of autophagy in iron-mediated aggravation of renal fibrosis, as identified in his preliminary studies. This research will form the basis for identification of relevant molecular mechanisms underlying the impact of iron on renal fibrosis in juvenile CKD, and inform novel therapeutic targets, which will be proposed in an R01 grant application before the end of the K08 award. Sandra Albrecht, Ph.D., M.P.H.

COLUMBIA UNIVERSITY HEALTH SCIENCES, NEW YORK, NY

Diabetes disparities in Latino subpopulations: linking biology to social epidemiology

Abstract

The overall goal for this application for an NIDDK Mentored Research Scientist Development Award (K01) is to provide the candidate, Sandra Albrecht, PhD, MPH, with indispensable training and protected time to build an independent, transdisciplinary research program at the interface of the biomedical and socio-ecological domains to provide a more complete understanding of the factors underlying progression to and from type 2 diabetes (T2D) in Latino subpopulations. Responding to the staggering burden of T2D and poor glucose control in U.S. Latinos, to unanswered questions regarding variation in disease risk and burden within this broad population, and to the dearth of longitudinal studies in this area, the research aims are to 1) Characterize patterns of glucose dysregulation over time in non- diabetic Latino ethnic subgroups, and evaluate the role of social, environmental, and clinical factors in driving that variation; (2) Characterize patterns of T2D progression over time in diabetic Latino ethnic subgroups, and evaluate the role of social, environmental, and clinical factors in driving that variation; and (3) Characterize the effectiveness of a key social determinant – healthcare utilization - for successful T2D management within a specific Latino subpopulation, and evaluate how other social, environmental, and clinical factors modify that effectiveness. Data for Aims 1 and 2 will come from a large national longitudinal population-based study with oversamples of Latino subgroups: the Hispanic Community Health Study/Study of Latinos. Data for Aim 3 will come from electronic health record data from a large healthcare system in North Carolina, a region with a fast growing Latino population. As part of this aim, Latino diabetic subjects will be recruited for a pilot study evaluating self-reported barriers to achieving control of glucose as part of a long-term effort to integrate social factors into the clinical management of T2D. To match the scope of the proposed scientific work, the candidate will seek to integrate her expertise in the social epidemiology of obesity and diabetes with the following career development and training areas: 1) training in the physiology of glucose dysregulation; 2) understanding of clinical issues relevant to T2D prevention and treatment; and 3) use of electronic health records data and health informatics. This training will be augmented with career-building activities to acquire essential tools for leadership and professional growth, as well as dissemination and translation of findings. Experts have emphasized the need for a more complete understanding of subgroup-specific T2D physiology and of subgroup-specific risk factors underlying progression to and from T2D. This K01 project will impact public health by improving characterization of T2D outcomes in Latino subpopulations to inform tailored prevention and treatment strategies. The project builds upon exceptional resources and mentoring at the candidate's institution (University of North Carolina at Chapel Hill), to train her in key new areas, bolster applications for competitive funding, disseminate findings among researchers and the wider community, augment faculty diversity, and attain research independence. Jose Aleman, Ph.D., M.D.

NEW YORK UNIVERSITY SCHOOL OF MEDICINE, NEW YORK, NY

Metabolic Flux Analysis of Obesity-Associated Inflammation in Weight Loss

ABSTRACT

Obesity is the most important preventable cause of disease in the United States, and its major complications include insulin resistance/type 2 diabetes, coronary artery disease (CAD), and heart failure. The reasons for these complications are not totally understood and assessment of patients before and after weight loss provides the opportunity to dissect the factors that are improved with reduced body fat. Elevated white blood cell (WBC) levels are a well-established CAD risk factor, while adipose tissue inflammation is considered an emerging risk factor for the development of obesity complications. However, the beneficial effects of weight loss on these inflammatory markers are not universal. Our recently published manuscript demonstrates how rapid diet-induced weight loss improves insulin sensitivity while increasing adipose tissue inflammation in human subjects. The proposed study will explore this dissociation and bariatric weight loss to seek mechanistic insight. We will characterize human adipose tissue macrophage metabolism in the context of weight loss using an in-vitro adipose tissue inflammation model (Aim 1). We will further assess changes in circulating WBC numbers and phenotype with bariatric weight loss in metabolically healthy versus unhealthy obese subjects undergoing sleeve gastrectomy (Aim 2), and concomitantly measure changes in adipose tissue inflammation (Aim 3) within six weeks of this intervention. The central hypothesis of this research is that lipid signals from the adipocyte modulate adipose tissue macrophage metabolism to prevent adipose tissue inflammation in weight loss. Consequently, the nature of this metabolic queue will prevent or ameliorate the WAT inflammatory state. Antiinflammatory macrophages are thought to maintain the integrity of healthy adipose tissue, while proinflammatory macrophages are thought to be the hallmark cell in adipose tissue inflammation. The rationale that underlies this research is that better understanding of the metabolic and immune cues that lead to white adipose tissue inflammation will allow for the development of detection and therapeutic strategies for this cardiometabolic risk factor. To achieve these aims, I will be supported by my primary mentor Dr. Ira Goldberg (NYULMC), a scientific leader in the study of lipid metabolism and circulating inflammatory markers in metabolic disease, and my co-mentor with expertise in the execution of detailed translational studies in the metabolic ward setting, Dr. Jan Breslow (). Each mentor will help me complete the individual aims of this project and develop the skills to pursue my independent scientific program studying the immunometabolism of human adipose tissue during weight loss. Alina Allen, M.D.

MAYO CLINIC ROCHESTER, ROCHESTER, MN

Noninvasive detection of NASH by magnetic resonance elastography (MRE)

PROJECT SUMMARY

Nonalcoholic steatohepatitis (NASH) is an established precursor of end-stage liver disease and hepatocellular carcinoma. To date, liver biopsy remains the only test available to detect the two diagnostic features of NASH: hepatocyte ballooning and lobular inflammation, but it is limited by invasiveness and potential complications. The central hypothesis of this application is that magnetic resonance elastogram (MRE) can discriminate NASH from simple steatosis. The hypothesis is supported by strong preliminary data which show that with novel technological modifications, MRE can isolate imaging parameters that correlate with inflammation and ballooning. A single multiparametric test that combines inflammation and ballooning assessment by MRE, and steatosis measurement by MRI-proton density fat fraction has the potential to provide a comprehensive estimation of the 3 components of histologic NAFLD activity score (NAS) in one setting. The objective of this study is to determine and validate the diagnostic performance of this single multiparametric test (henceforth named Hepatogram) for the detection of NASH in human subjects. We will test the central hypothesis in 3 AIMs. In AIM 1 we will use a cohort of obese subjects scheduled for bariatric surgery to assess NASH histologically by liver biopsy obtained during bariatric surgery and noninvasively before surgery by Hepatogram. The imaging parameters of inflammation, ballooning and steatosis will be used to develop a statistical model to predict histologic NAS score. In AIM 2 we will validate this statistical model in an independent, non- bariatric NAFLD cohort, in whom NASH will be diagnosed by liver biopsy and assessed by Hepatogram. In AIM 3 we will examine the longitudinal diagnostic performance of Hepatogram in the detection of NASH regression after weight-loss interventions. We will reevaluate the bariatric cohort described in Aim 1, one year after bariatric surgery by assessing for NASH regression by Hepatogram and liver biopsy. The predictive performance of the statistical model developed in Aim 1 and validated in Aim 2 will be examined by comparing the predicted MRE-based change in NAS score to the change in histologic NAS. This proposal will apply technically innovative approaches to a large, rigorously characterized population with NAFLD to test the hypothesis that Hepatogram is an accurate for NASH diagnosis, monitoring and response to interventions. Obviating the impractical need for liver biopsy to diagnose NASH before onset of fibrosis would positively impact the care of NAFLD patients. This work will be performed in an academically nurturing environment within Mayo Clinic. The candidate will be guided by a strong mentorship committee of experts in the areas that the candidate identified for necessary training. The candidate acquired a strong foundation in statistical skills in assessment of disease risk, prognostication and outcomes evaluation during her post- doctoral training. She now proposes to solidify her foundation and acquire a set of new and complementary skills necessary for an independent career in clinical research in nonalcoholic fatty liver disease. Joana Almaca, Ph.D.

UNIVERSITY OF MIAMI SCHOOL OF MEDICINE, CORAL GABLES, FL

Role of pericytes in pancreatic islet fibrosis

Abstract

Fibrosis is the most frequent lesion in the islets of type 2 diabetics (T2D) and contributes to the age- dependent impairment of islet function. Defects in islet vasculature compromise exchanges between the endocrine cells and the blood, disrupt islet architecture and ultimately lead to endocrine cell death. An important component of the vasculature is the pericyte, a contractile smooth muscle-like cell that wraps small blood vessels. In different organs, pericytes have been shown to differentiate into myofibroblasts, leading to fibrosis and organ dysfunction. Whether islet pericytes also contribute to the profibrotic myofibroblast pool that causes islet fibrosis observed during aging and T2D has not been determined. The long-term goal of this proposal is to understand the role of vascular dysfunction in aging and diabetes. The objectives of this project are to determine how the islet pericyte phenotype changes during insulin resistant states (such as aging and T2D) and what causes the changes, using a combination of in vitro and approaches. The central hypothesis is that, during aging or T2D, the excessive exposure to insulin exacerbates signaling through the mammalian target of rapamycin (mTOR) in pericytes, which makes them differentiate into myofibroblasts. In our model, as hyperinsulinemia develops to compensate for insulin resistance, islet pericytes are exposed to higher levels of insulin. Insulin overactivates mTOR signaling in pericytes, which favors their differentiation into myofibroblasts and proliferation of these profibrotic cells. The rationale for the proposed research is that the results will make a lasting impact on our understanding of the role of the pericyte in islet biology. If the hypothesis is correct, it would demonstrate the fundamental contribution of pericytes to islet fibrosis and diabetes pathogenesis. The proposed research is therefore relevant to the mission of the NIH that pertains to the pursuit of fundamental knowledge about the nature and behavior of living systems. Guided by strong preliminary data, our central hypothesis will be tested by pursuing two specific aims: 1) Identify age- and diabetes-induced changes in the phenotype of the islet pericyte; 2) Determine the role of mTOR-dependent insulin signaling in pericyte transdifferentiation. Under the first aim, we will examine changes in the pericyte phenotype in aged and T2D mouse and human islets, and directly visualize the phenotypic transition from pericytes to myofibroblasts in vivo. Under the second aim, we will determine if direct in vitro and in vivo stimulation of islet pericytes with insulin triggers a pro-fibrotic myofibroblast-like phenotype. We will further manipulate mTOR signaling in pericytes in vivo and measure the effects on vascular function and glucose homeostasis. The proposed research is significant because pericytes can be targeted to limit the generation of myofibroblasts and interstitial collagen accumulation during islet fibrosis that accompanies aging and T2D. Importantly, these studies have the potential to impact the way diabetes is treated. Yu An, Ph.D.

UT SOUTHWESTERN MEDICAL CENTER, DALLAS, TX

Adipocyte mitochondrial distress drives the intercommunication between white and brown adipose tissues

Project Summary/Abstract

One third of American adults are suffering from obesity, which significantly contributes to the of many other life-threatening diseases, such as type 2 diabetes, cardiovascular diseases, cancers, etc. Although substantial resources have been deployed to resolve this major public health threat, the fact is that currently only limited ways of intervention have been developed. An in-depth understanding of the pathogenesis of obesity to facilitate finding more effective therapeutics is urgently needed. The overarching goal of this K01 proposal is to investigate the crosstalk between two major players in the onset of obesity, white and brown adipose tissues, and to identify novel factors mediating the white fat-brown fat communication. The applicant recently has identified that overexpression of amyloid precursor protein (APP) in white fat and its subsequent mistargeting into mitochondria induces dramatic mitochondrial dysfunction, thereby promoting obesity and insulin resistance. Preliminary data obtained from this unique APP-induced mitochondrial distress model show that mitochondrial distress in white fat induces a “whitening” phenotype in brown fat, and in contrast, brown fat- specific mitochondrial distress causes “browning” in white fat. Therefore, this proposal is set out to test the central hypotheses: 1) APP-induced mitochondrial distress is a central determinant of white/brown fat intercommunication; 2) white/brown fat intercommunication involves yet unrecognized signals; 3) white/brown fat intercommunication impacts systemic metabolism. In Aim 1, white or brown fat- specific overexpression or deletion of APP mouse models will be subject to metabolic characterizations and mechanistic investigations. In Aim 2, multiple layers of unbiased strategies will be conducted to discover neuronal or hormonal factors that act as communicating signals between white and brown fat, and these factors are predicted to exert important obesogenic or anti-obesogenic roles. Meanwhile, as a career development award, this proposal also outlines an integrated training and research plan for the applicant to complete further academic training under the mentorship of Dr. Philipp E. Scherer and Dr. Joel K. Elmquist, ensuing the transition to an independent investigator specializing in the field of metabolically active tissue crosstalk in the context of obesity. Furthermore, the outstanding resources provided by UT Southwestern Medical Center will maximize the potential for the applicant to fulfill the career objectives in identifying novel mechanisms underlying pathogenesis of obesity and new therapeutics treating obesity. Combined, together with longstanding interest and established ability of the applicant in studying adipose tissue biology, excellent mentorship from internationally recognized leaders in the metabolism field, and unparalleled environment at UT Southwestern, this K01 career development award will be essential for the applicant to receive additional training in brown adipose biology, neuronal regulation of browning activity, proteomics based secreting factor identification, and therapeutic discovery, thereby fully supporting a successful transition to independence for the applicant. Christina Astley, D.Sc.

BOSTON CHILDREN'S HOSPITAL, BOSTON, MA

Seasonal and Viral Determinants of Type 1 Diabetes Onset, Remission, and Glycemic Control

PROJECT SUMMARY/ABSTRACT

Glycemic control strongly predicts outcomes in type 1 diabetes (T1D), yet many children with T1D fail to achieve glycemic targets. This problem is magnified by rising T1D incidence and incomplete understanding of the etiology of T1D. Winter predominance of T1D onset, worsening of glycemic control, and loss of residual beta cell function suggest that there may be shared seasonal triggers that contribute to T1D pathogenesis. Epidemiologic studies using 3-month average glycemic control measures (i.e. glycated hemoglobin) obscure temporal trends. Investigators of potential viral triggers have examined the role of prespecified viruses, but have not conducted comprehensive testing for all human viruses. Wide use of continuous glucose monitoring (CGM) and the availability of systematic tests for all prior viral exposures can now be leveraged to dissect the causes of T1D and highlight relevant biology for T1D prevention and management. Dr. Christina Mills Astley, a pediatric endocrinologist and epidemiologist, will approach seasonality of incident and prevalent T1D using an innovative combination of translational phenotyping, time-series analyses, and integration of external datasets in her Mentored Patient-Oriented Research Career Development Award. She now seeks to build on her prior research experiences in infectious diseases modeling and causal inference for complex data, while acquiring new skills in time-series analysis and bioinformatics, and developing expertise in T1D pathophysiology, including the role of immune dysregulation, inflammation, and metabolic stress. Combined with the high-quality mentorship and supportive environment at Boston Children’s Hospital, these tools will broaden her expertise and enable her to establish an independent research program. In Aim 1, Dr. Astley will use pediatric T1D CGM data and time-series methods (e.g. change point, autoregression, temporal clustering) to define seasonal patterns, both acute and chronic, in clinically- relevant CGM-based glycemic control measures (e.g. coefficient of variation, time in range). In Aim 2, she will quantify the effect of individual, social, and environmental exposures on these glycemic control measures. Finally, in Aim 3, Dr. Astley will apply comprehensive antibody profiling (VirScan) to test which viral infection(s) drive T1D onset, end of remission, and seasonal dysglycemia. Together the results of these investigations will form the basis of a data-driven approach to understand seasonality in T1D, which in turn may be used to recognize optimal timing for anticipatory guidance to mitigate the risk of seasonal hyperglycemia and hypoglycemia, and to identify potential targets for intervention to slow the progression of T1D. Dr. Astley seeks to become an independent physician-scientist with unique expertise in computational epidemiology, translational methodologies, and T1D pathogenesis, so as to improve prevention and treatment. This K23 award will afford her the mentorship, training, expertise, and research to achieve these goals.

Ling Bai, Ph.D.

UNIVERSITY OF CALIFORNIA, SAN FRANCISCO, SAN FRANCISCO, CA

Sensory mechanisms underlying metabolic regulation

PROJECT SUMMARY

Animals have a remarkable ability to restore metabolic homeostasis after a challenge to their internal state. In order to maintain energy balance after a meal, the composition and quantity of ingested food needs to be closely monitored to coordinate the physiological responses of visceral organs. Dysregulation of this process leads to metabolic disease like obesity and diabetes. However, how sensory signals of nutritional state are detected and processed to generate metabolic commands is poorly understood. Vagal sensory neurons innervate the gut and are poised to detect diverse interoceptive cues. My postdoctoral work in Dr. Zachary Knight's lab has generated a cellular map of vagal sensory neurons that links their molecular identity to their target organ innervations and putative gut signals. This work provided a roadmap for the use of genetic tools to manipulate vagal subtypes with high specificity. In the mentored phase of this grant, I will build upon my postdoctoral discoveries by investigating how vagal sensory neurons regulate autonomic responses to achieve metabolic homeostasis (Aim 1). These mentored will allow me to gain deep understanding of metabolic regulation and autonomic physiology, as well as to acquire additional surgical skills to manipulate the GI tract. The target of vagal neurons is the nucleus tractus solitarius (NTS), a key metabolic center that integrates neural and circulating humoral signals and generates complex physiological and behavioral commands to maintain energy balance. In the independent phase of this grant, I will build upon my graduate and postdoctoral training to further investigate how the downstream neuronal circuits in the NTS integrate and process interoceptive signals, and how they generate output commands to drive metabolic responses (Aim 2 and Aim 3). This grant will allow me to expand my current experimental and intellectual skills and develop additional expertise in metabolic regulation as well as electrophysiology guided circuit mapping, under the guidance of my advisory committee who are expertise in those fields. This will further link my sensory neurobiology skillset to the study of physiologic function and advance my career goal of being an independent researcher. Wale Bamidele, Ph.D.

MAYO CLINIC ROCHESTER, ROCHESTER, MN

Gastrointestinal Cues Influence Metabolic Control of Regulatory T cell Function

PROJECT ABSTRACT

Human inflammatory bowel disease (IBD) is characterized by inconsistent response to therapies and persistent activation of pathogenic effector CD4+ T cells implying regulatory T cell (Treg) dysfunction; however, the underlying mechanisms are poorly understood. Therefore, the OVERALL OBJECTIVE of this proposal is to elucidate the mechanisms by which gastrointestinal cues impact Treg metabolism and function with the therapeutic goal of defining pharmacological and adoptive Treg therapies to treat IBD. For the first time using complementary approaches, we have observed a defined mitochondrial ultrastructure (shape, cristae structure, and physical interaction with the endoplasmic reticulum [ER]) which correlated with Treg metabolic state. We are now poised to exploit how: i) mitochondrial ultrastructure and its associated metabolic state in Tregs suppress gut inflammation in various mouse models of experimental colitis; and ii) a breakdown in the regulation and function of mitochondrial ultrastructure can drive human IBD pathogenesis by analyzing Tregs from peripheral blood and lamina propria of IBD patients compared to relevant healthy individuals. Our preliminary data suggest that anti- inflammatory transforming growth factor beta 1 (TGF-β1) cytokine is a critical driver of mitochondria-ER contact (MERC) in Tregs via its associated molecular architecture, thus implicating intact MERC and subsequent pyruvate oxidation in Treg-mediated suppression of IBD. In our preliminary experimentations mimicking the proinflammatory milieu of IBD gastrointestinal tract, treatment of Tregs with proinflammatory cytokines impaired MERC and perturbed glucose metabolism, leading to excessive fatty acid oxidation as a compensatory mechanism in contrast to vehicle-treated Tregs (“proinflammatory cytokine-induced metabolic reprogramming”). Furthermore, we discovered that proinflammatory cytokine-induced metabolic reprogramming of Tregs was reversed by inhibiting the activity of glycogen synthase 3 beta (GSK3β) using a class of inhibitors currently being explored in clinical trials for other indications. Based on these novel observations, we formulated the CENTRAL HYPOTHESIS that TGF-β1 mediates mitochondria-ER contact that is essential for cellular metabolic homeostasis, Treg function, and suppression of IBD pathogenesis. The following independent SPECIFIC AIMS are designed to test three integrated hypotheses. First, we will directly test the hypothesis that TGF-β1 mediates MERC and consequently Treg function. Second, we will test the hypothesis that TGF-β1 potentiates mitochondrial pyruvate oxidation and consequently Treg function. Third, we will test the hypothesis that proinflammatory cytokines perpetuate IBD pathogenesis in vivo via MERC inhibition. We propose to utilize sophisticated approaches relevant to health and IBD athophysiology to test this hypothesis. This proposal, which is technically and conceptually innovative, is also significant because it presents a novel concept in Treg biology and identifies new mechanisms for therapeutically optimizing Tregs, namely combining pharmacological and Treg-based therapies, to halt the refractory nature of IBD. Irina Bancos, M.D.

MAYO CLINIC ROCHESTER, ROCHESTER, MN

Skeletal Consequences of Mild Autonomous Cortisol Secretion

PROJECT SUMMARY/ABSTRACT

Adrenal adenomas affect 5%-10% of the population, discovered annually in about 4 million adults in United States. Patients with adrenal adenomas demonstrate an abnormal steroid secretion and metabolism, the most prevalent being called “subclinical” mild, autonomous cortisol secretion (MACS). Available limited evidence suggests that patients with MACS suffer from very high-rates of vertebral fractures, many times not correlated with deterioration of bone density. Understanding how MACS affects bone metabolism and leads to fractures is significantly limited by heterogeneous patient populations and the multifactorial etiology of bone disease. The overall objective of the proposed project is to understand the epidemiology of fractures in patients with adrenal tumors and MACS, and to identify the elements of abnormal steroid secretion and metabolism that predict bone disease, and ultimately fractures. This will be accomplished in three specific aims. In Aim 1, the Olmsted county population will be accessed through the Rochester Epidemiology Project (REP) to determine the prevalence and incidence of fragility fractures in patients with adrenal tumors overall, and the effect of MACS severity on fragility on fracture occurrence will be established. The experiments proposed in Aim 2 will determine if the abnormal steroid metabolome observed in patients with MACS is associated with impaired bone density, quality, structure and metabolism that, in turn, are associated with an increased risk of both vertebral and non-vertebral fractures. The experiments proposed in Aim 3 will determine if the abnormal circadian pattern of cortisol secretion in patients with MACS is associated with abnormal calcium metabolism and alterations in bone turnover markers. The proposed career development award and associated candidate training address an important health problem of skeletal health in patients with MACS and prepare the candidate for an independent career in research, meeting the goal of the NIH mentored Patient-Oriented Research Career Development Award (Parent K23 - Independent Not Allowed), PA-18-375. The exceptional resources and institutional support at Mayo Clinic, outstanding multi-disciplinary mentorship team, and proposed career development activities will allow the candidate to achieve her long-term goal of becoming an independent investigator and nationally recognized expert on steroid effects on bone health and design interventions that are effective in improving skeletal health of patients with adrenal tumors. Anne Bantle, M.D.

UNIVERSITY OF MINNESOTA, MINNEAPOLIS, MN

A Weight Neutral, High Protein, Moderate Carbohydrate Diet for Treatment of Type 2 Diabetes Mellitus

PROJECT SUMMARY

Type 2 diabetes mellitus (DM2) causes significant morbidity and mortality. Diet can be a powerful tool for treatment of DM2, but the potential impact of diet has not been fully realized, which may be because usual recommendations include the challenging goals of caloric restriction and weight loss. An isocaloric, moderate carbohydrate, high protein diet (the Low Biologically Available Glucose, or LoBAG, diet) has been designed specifically for people with DM2, and is unique because it does not require caloric restriction or weight loss to produce beneficial effects. Data from preliminary efficacy studies are exciting, but are limited in generalizability. The purpose of the proposed project is to expand generalizability by testing the effectiveness of the LoBAG diet in people with DM2 who are free-living (given diet instruction and asked to prepare their own food). This project is proposed as a key component of a five-year career development plan for Dr. Anne Bantle, an endocrinologist and early career investigator at the University of Minnesota. Dr. Bantle’s long-term goal is to become an independent clinician investigator with expertise in treatment of DM2, with particular focus on diet and the gut microbiome. Her career development plan will allow her to 1) develop expertise in clinical research as a principal investigator, 2) develop expertise in DM2, diet, and gut microbiome, 3) develop skills necessary to lead an interdisciplinary research team, and 4) generate preliminary data to support future funding applications. Specific career development activities will include completion of a Master’s Degree in Clinical Research, hands- on training through implementation of the proposed research study and as a co-investigator in multicenter clinical trials, instruction in gut microbiome analysis, attendance and participation in research meetings, and ultimately, successful application for independent funding. The proposed research project will test the hypothesis that the LoBAG diet is effective in free- living participants with DM2, and specifically that consumption of the LoBAG diet will result in greater reduction of hemoglobin A1c than consumption of a control diet, without requiring caloric restriction or weight loss. Thirty-eight participants will be enrolled in a randomized controlled clinical trial of the LoBAG diet versus a control diet over a 12-week intervention period. Specific aims are to 1) determine the effect of the LoBAG diet compared to control on glycemic control, 2) determine the effect of the LoBAG diet compared to control on compliance and quality of life, and 3) determine if features of the gut microbiome are predictive of glycemic response to diet. It is anticipated that this project will generate preliminary data for a future larger, definitive effectiveness trial. The skills and expertise gained from the proposed career development plan and research project will allow Dr. Bantle to successfully emerge as an independent investigator, and will lay the groundwork for her to become a leading scientist in the field of DM2, diet, and gut microbiome. Kedryn Baskin, Ph.D.

OHIO STATE UNIVERSITY, COLUMBUS, OH

Cardiomyokines regulate inter-organ communication and metabolism

Project Summary

The heart has the remarkable capacity to adapt to metabolic, hormonal, and stress signals, in part by secreting factors that act in an autocrine/paracrine manner to optimize cardiac function. However, the endocrine function of the heart is a relatively new aspect of cardiac biology that is not fully understood. Dr. Kedryn Baskin is spear- heading investigations into the role of the heart in regulating systemic metabolism. We recently determined that “lean factors” secreted by the heart regulate metabolism of extra-cardiac tissue. These findings suggest that identification and exploitation of cardiac-secreted factor(s) has therapeutic potential. The PI’s long-term goals are to identify secreted and metabolites from the heart that have the potential to regulate systemic metabolism. Her short-term goals are to determine the metabolic functions of GBAS and KLK11. To accomplish these goals, the PI has performed proteomic analysis of secreted factors and screened them for their metabolic capacity. She has identified several “lean factors” secreted from the heart (called cardiomyokines) that enhance metabolic rates. The PI will investigate GBAS and KLK11, two novel cardiomyokines that regulate metabolism. Aim 1 will determine how cardiac-secreted GBAS and KLK11 regulate systemic metabolism. Aim 2 will identify the roles of these proteins during exercise and will determine the protective effect of GBAS and KLK11 in the setting of obesity, and Aim 3 will determine the mechanisms by which this occurs. This application will provide the PI with the scientific training, mentoring, and career development necessary as she transitions to inde- pendence. The scientifically stimulating, and highly collaborative nature of UT Southwestern provides an envi- ronment that supports the development of young investigators. The PI will be mentored by Dr. Eric Olson and the members of the PI’s advisory committee, Drs. Philipp Scherer, Rhonda Bassel-Duby, Jeffrey McDonald, and Hamid Mirzae. The proposed research and career development plan will enable the PI to investigate the metabolic functions of GBAS and KLK11. She will continue to elucidate the functions of GBAS and KLK11, delv- ing into the mechanistic details of how GBAS and KLK11 regulate metabolism. Aims 1-3 will provide the scien- tific tools and training needed to successfully advance the burgeoning field of metabolically active secreted factors. Identification of cardiomyokines with metabolic signaling properties provides a foundation for the dis- covery of novel therapeutic targets for metabolic diseases. Lisa Beutler, Ph.D., M.D.

NORTHWESTERN UNIVERSITY AT CHICAGO, CHICAGO, IL

Dissecting the Nutritional Regulation of Feeding Circuits

PROJECT SUMMARY/ABSTRACT

RESEARCH STRATEGY: Communication between the gut and the brain is essential for energy homeostasis, but how this communication is represented in the dynamics of hypothalamic feeding circuitry is unknown. Early studies of the gut-brain axis relied upon indirect measurements of the effects of nutritionally regulated peripheral signals on feeding circuitry. These studies led to a model in which the activity of key hypothalamic hunger neurons – AgRP neurons – fluctuates gradually as the animal's nutritional state changes. With the development of techniques to record the activity of genetically- defined neuronal populations in awake animals, the dynamics of AgRP neurons were recently observed in vivo for the first time. These studies revealed, contrary to the prevailing model, that AgRP neurons are inhibited rapidly when an animal sees or smells food, before it takes a single bite; however, food ingestion is required for maintenance of this inhibition. We have developed a tool combining in vivo monitoring of AgRP neuron dynamics with intragastric nutrient infusion to show for the first time that nutrient delivery to the gut, in the absence of the sensory stimuli normally associated with eating, is sufficient to inhibit AgRP neurons over a time-scale of minutes. This inhibition is independent of the macronutrient composition of the food but depends upon the number of calories ingested. The goal of this proposal is to determine the molecular and circuit-based mechanisms by which each macronutrient inhibits AgRP neurons. This will be accomplished across three aims: to identify the hormonal mediators responsible, to identify the nutrient sensors involved, and to dissect the pathway by which these signals reach AgRP neurons. CANDIDATE/ENVIRONMENT: Dr. Lisa Beutler is a senior fellow in the Division of Endocrinology at UCSF. She recently completed internal medicine residency at UCSF and an MD/PhD at the University of Washington, where she earned her PhD in Dr. Richard Palmiter's laboratory. She is finishing her fellowship research, which is the subject of a first-author publication in the journal Neuron, in Dr. Zachary Knight's laboratory at UCSF. Having gained expertise in in vivo neural recording and advanced rodent surgery, she now seeks to expand her expertise in the lab to include optical circuit dissection, single-cell resolution calcium imaging, and data analysis and programming skills prior to obtaining an independent position as an academic physician-scientist. CAREER DEVELOPMENT: This award will ensure that Dr. Beutler is able to launch her career as an independent investigator armed with a combination of experimental tools that both position her at the cutting edge of her field (calcium- based imaging techniques, optogenetics) and set her apart from others in the field (advanced rodent surgical techniques). Combined with her clinical training in endocrinology, this will make her uniquely poised to address questions that require detailed knowledge of both neural circuitry and peripheral metabolism. This award will also facilitate Dr. Beutler's acquisition of other professional skills required for independence including formal training in scientific writing, leadership, and management. Pavan Bhatraju, M.S., M.D.

UNIVERSITY OF WASHINGTON, SEATTLE, WA

The Assessment of Acute Kidney Injury Sub-phenotypes in the Intensive Care Unit

Project Summary/Abstract

Pavan Bhatraju, MD, MSc plans for a career as a molecular epidemiologist physician scientist in the field of acute kidney injury (AKI) in the intensive care unit (ICU). The objective of the proposed career development award is to provide necessary training in research methods to facilitate Dr. Bhatraju’s transition to independent research. The mentorship available at the University of Washington (UW) in the Division of Pulmonary and Critical Care Medicine (PCCM) and the Kidney Research Institute are outstanding. He will continue his work under the mentorship of Drs. Mark Wurfel (PCCM) and Jonathan Himmelfarb (Nephrology) at the UW. Collaborators on his multi-disciplinary research team span the disciplines of genome sciences (Dr. Gail Jarvik), epidemiology and biostatistics (Dr. Ronit Katz) and the application of translational research to the study of AKI (Dr. Chirag Parik). To complement the support from this research team, Dr. Bhatraju will augment his Masters in Science in Epidemiology with advanced coursework at the nationally-renowned UW School of Public Health. AKI is common in the ICU and is associated with morbidity and mortality. Currently we have no effective pharmacological therapy for AKI. Limitations in drug development may be partly explained by the biological and clinical heterogeneity in patients included in the AKI phenotype. To address these limitations, our lab has identified two distinct AKI sub-phenotypes (also known as endotypes) using an innovative modeling approach, latent class analysis. We found that the two AKI sub-phenotypes had different clinical characteristics, biomarker patterns and risk of poor short-term outcomes. We also developed and validated a 3-variable prediction model to prospectively identify the AKI sub-phenotypes. Overall, these findings provide the foundation for Dr. Bhatraju’s K23 research proposal. The objectives of the proposed research include: 1) evaluate the association of AKI sub-phenotypes with major adverse kidney events by 1 year; 2) determine the genetic risk factors associated with the development of AKI sub-phenotypes, and; 3) identify specific pathophysiologic differences between the AKI sub-phenotypes through a targeted urinary protein analysis. This award will provide essential research training in genome sciences, biostatistics, advanced epidemiology and will broaden his bench lab skillset. This proposal leverages Dr. Bhatraju’s unique access to multiple critically ill cohorts, access to a research infrastructure at the UW to enroll a prospective ICU cohort collecting urine, plasma and DNA samples and access to world-class didactic training program at the UW School of Public Health. His overall research goal is to use techniques of molecular epidemiology to develop a pathophysiologic based classification of patients with AKI to improve risk prognostication, discover novel genetic risk factors and inform precision based strategies to prevent and treat the development of AKI in the ICU. With the support of this K23, the research and education agenda Dr. Bhatraju has devised will assist him in achieving his long-term goal of becoming an independent translational investigator conducting AKI research. Jason Bini, Ph.D.

YALE UNIVERSITY, NEW HAVEN, CT

Direct measurement of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) enzyme levels in obesity using a novel positron emissiontomography radioligand

PROJECT SUMMARY

The prevalence of obesity in the United States population is over 30%, predisposing a large portion of the population to metabolic diseases. Cortisol, a steroid hormone, is of critical importance in obesity, as it is responsible for stimulating gluconeogenesis in the liver and promoting adipocyte differentiation and maturation. Cortisol is activated from cortisone by the intracellular enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). The overall aim of this proposal is to examine whole-body 11β-HSD1 enzyme distribution levels during normal physiology and in response to weight gain, insulin resistance and obesity using the novel 18F-FMOZAT PET radioligand in rodent and human populations. Whole-body PET imaging can provide a direct measure of the distribution of 11β-HSD1 allowing comparison to conventional methods using urinary or plasma cortisol metabolites in both lean and non-diabetic obese human individuals. We propose an obese Zucker fatty (ZF) rat model that will allow direct in vivo PET imaging of 11β-HSD1 levels in various tissues during progression to obesity (Aim 1). We also propose to examine 11β-HSD1 levels in lean and non-diabetic obese individuals with 18F-FMOZAT PET to assess tissue-specific variability in whole-body 11β-HSD1 enzyme levels, and compare these novel direct measures with current methods using urinary metabolites of cortisol (Aim 2). The program of research and training described in this K01 Mentored Career Development Award application will provide the candidate with the requisite skills and experience to become an independent investigator in the field of endocrinology (obesity). Under the mentorship of field-leading experts in endocrinology and obesity, and PET imaging, the proposed aims will afford training in the design, conduct and analysis of novel translational PET imaging techniques to be uniquely applied to metabolic syndromes in endocrinology, such as obesity. In pursuit of this goal, the candidate proposes to undertake further training in three primary areas 1) build expertise in clinical endocrinology (obesity) research 2) receive education in the pathophysiology of obesity and 3) continue to enhance my knowledge of cutting-edge whole-body PET imaging. The opportunities afforded by this award would enable the candidate to embark on a comprehensive, structured 5-year program of training and research designed to develop an expertise in innovative endocrinology research methods toward a career as an independent investigator. Therese Bittermann, M.S., M.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

Impact of immunosupprion variability on outcomes after liver transplantation

PROJECT SUMMARY

Over 95% of liver transplant (LT) recipients have only one opportunity to receive a new liver during their lifetimes and long-term patient survival depends upon prolonged graft functioning. As a result of advances in immunosuppression (IS), over 70% of recipients survive more than 5 years after LT, yet wide variability exists in clinical outcomes at the center-level. Most late post-LT deaths are not directly due to liver failure, but reflect the adverse consequences of prolonged IS therapy. Due to the lack of national recommendations to guide IS after LT, its management differs across centers. Moreover, comparative efficacy and safety data for IS regimens are only available from small trials and meta-analyses. Their interpretation and generalizability are limited due to the following factors: 1) care is often provided in settings that do not reflect “real world” clinical practice; 2) few IS regimens are compared, often at a single time point; 3) late or rare outcomes are missed, and 4) publication bias is evident. Population- level research is therefore needed to evaluate comprehensively the comparative effectiveness and safety of IS regimens for LT. Understanding center differences in IS management will also identify suboptimal practices, and further encourage the development of standardized guidelines. Since fewer than 10% of the observed variability in post-transplant outcomes can be explained by pre-LT factors, we hypothesize that targeting differences in post-LT management, such as IS regimen selection, has the potential to change practice and improve outcomes on a wider scale. Using a novel linkage of transplant data from the United Network for Organ Sharing and Medicare claims, the primary aims of the proposed research are the following: Aim 1 – to describe center variability in IS management in the US; Aim 2 – to evaluate the association between IS regimen and the risks of graft failure and mortality; and Aim 3 – to determine the association between IS regimen and the risks of acute rejection, severe infection and de novo cancer as potential mechanisms for the relationships identified in Aim 2. This proposal will also foster Dr. Bittermann's career development into a fully independent NIH-funded clinical investigator with a scientific focus in LT pharmacoepidemiology and practice variability, facilitated through a comprehensive mentorship plan. This will be accomplished by: 1) additional coursework on advanced methodologies through the Center for Clinical Epidemiology and Biostatistics (CCEB) at the University of Pennsylvania, 2) direct implementation of these acquired techniques under the close supervision of a carefully selected team of faculty with extensive expertise in pharmacoepidemiology, transplant hepatology, and outcomes research, as well as longstanding experience with successfully mentoring prior grant recipients, 3) involvement in the Center for Pharmacoepidemiology Research and Training in the CCEB, and 4) development and submission of future grants during the latter part of the award period to further IS and related post-transplant management issues in LT recipients. BRIGID BOLAND, M.D.

UNIVERSITY OF CALIFORNIA, SAN DIEGO, LA JOLLA, CA

USING SINGLE-CELL RNA AND PROTEIN APPROACHES TO ELUCIDATE MOLECULAR DETERMINANTS AND PREDICTORS OF INFLAMMATORY BOWEL DISEASES

Abstract

Inflammatory bowel diseases (IBD) represent a spectrum of complicated intestinal pathology characterized by dysregulation of the adaptive and innate immune responses in genetically susceptible hosts. The precise molecular mechanisms underlying the pathophysiology and immune dysregulation have yet to be fully elucidated. A significant barrier to better understanding of the disease pathogenesis is the heterogeneity of intestinal tissue. Furthermore, substantial heterogeneity may exist even within the same phenotypic cell subset, limiting the ability to detect differences using bulk RNA sequencing or flow cytometry. Single cell RNA sequencing and single cell protein identification in combination, however, represent a significant technologic advance with the capability to identify novel cell subsets and states that could not be previously detected. We propose exploiting this technology to generate a single cell atlas of the intestinal and peripheral blood immune response in two subsets of IBD: ulcerative colitis (UC) and ileal Crohn’s disease. The overall objective of this proposal is to define molecular determinants of IBD in tissue and blood, then to harness these differences in gene expression to develop novel diagnostic testing for IBD using peripheral blood. Specific Aim 1 proposes to identify abnormal states of differentiation and pathogenic cell subtypes that are enriched in the intestinal tissue and peripheral blood from UC and Crohn’s ileitis as compared to healthy controls. The differences in gene expression between the intestinal cells from IBD patients and healthy controls will provide insight into molecular determinants of disease that are specific to UC and Crohn’s ileitis. Specific Aim 2 proposes to utilize the differences in single cell gene expression from the peripheral blood in a larger cohort of patients to develop diagnostic testing for UC and Crohn’s ileitis. Non-invasive testing with single cell gene expression has the potential to predict not only disease status but also severity of disease activity with potential therapeutic targets. In sum, this proposal aims to use single cell gene and protein expression to understand disease pathophysiology of IBD and develop novel diagnostic biomarkers. CARY RAGAN BOYD-SHIWARSKI, Ph.D., M.D.

UNIVERSITY OF PITTSBURGH AT PITTSBURGH, PITTSBURGH, PA

The function of kidney specific (KS)-WNK1 condensates during potassium stress

PROJECT SUMMARY/ABSTRACT

Potassium [K+]•deficient diets have contributed to the global epidemic of hypertension and chronic kidney disease (CKD). Given the low cost and ease of increasing dietary [K+], more research is needed to understand how [K+] imbalance leads to these diseases. The kidneys handle 90% of [K+], and the distal convoluted tubule (DCT) acts as a [K+] sensor via the WNK-SPAK (With-No-Lysine/Ste20/SPS-1-related proline-alanine-rich protein kinase) pathway. Gain-of-function mutations to this pathway lead to severe hypertension and hyperkalemia by activation of the thiazide-sensitive sodium/chloride co-transporter (NCC). Curiously, dietary [K+] depletion or loading causes the WNK-SPAK kinases to assemble into large DCT-specific cytoplasmic puncta, that are not seen in mice on normokalemic diets. For years, the structure and function of these condensates, which we call “WNK bodies”, remained a mystery. Dr. Boyd-Shiwarski’s initial work has identified that these DCT-specific punta are (i) dependent upon the expression of kidney specific (KS)-WNK1 (ii) potassium-sensitive; (iii) membrane-less; (iv) not associated with conventional organelles; and (v) associated with WNK-SPAK proteins. Based on these findings, we hypothesize that WNK bodies are membrane-less microdomains that sequester the WNK-SPAK pathway to modulate WNK signaling during potassium imbalance. This hypothesis will be tested in two aims that evaluate the physiological significance and biological basis of WNK body formation. This proposal’s physiology- •based aim will provide Dr. Boyd•Shiwarski with the opportunity to work with animal models and (i) implement microscopy techniques, (ii) quantify changes in urine, serum, and blood pressure, and (iii) develop transgenic mouse models. Whereas, the biology-based portion of this proposal will include implementation of (i) molecular , (ii) protein biochemistry, and (iii) mass spectrometry and RNA Seq. These skills will be reinforced by a team of mentors, advisors, collaborators, and core resources available at the University of Pittsburgh. The primary mentor, Dr. Arohan Subramanya, is an established NIH R01•funded physician-scientist with 13 years of experience in WNK signaling biology and prior experience mentoring over 20 trainees. The co•mentor, Dr. Tom Kleyman, is an internationally recognized physician scientist who directs the Pittsburgh Center for Kidney Research, and has mentored nine career development awardees and five R01 recipients within the last 10 years. In addition, an advisory committee of accomplished investigators with expertise in hypertension, WNK-SPAK signaling, and renal tubular transport will monitor Dr. Boyd- Shiwarski’s progress through biannual meetings. Dr. Boyd•Shiwarski will use this proposal to accomplish her short•term goal of scientific independence and her long•term goal of becoming a tenure track physician- scientist with expertise in potassium homeostasis, hypertension, and CKD. The results from this proposal will form the basis for an R01 studying the translational role of KS-WNKY1-dependent WNK bodies in human models of nephropathy.

Graham Brady, Ph.D., M.D.

UNIVERSITY OF MICHIGAN AT ANN ARBOR, ANN ARBOR, MI

Nuclear and the hepatocyte nuclear envelope in nonalcoholic fatty liver disease

Project Summary/Abstract

This revised K08 proposal will complete Graham Brady, MD, PhD's training toward his goal of improving our understanding and treatment of nonalcoholic fatty liver disease (NAFLD). Dr. Brady is a physician and scientist in hepatology at the University of Michigan with clinical expertise in liver disease, graduate training in cell biology and biochemistry, and established success in the field of NAFLD. This proposal builds on Dr. Brady's prior experience, with new training in , bioinformatics, and a unique model (zebrafish). His existing expertise and newly-acquired skills will be integrated to advance our understanding of the scope and mechanisms of nuclear lamina-related NAFLD. The research will be conducted under the guidance of primary mentor Bishr Omary, MD, PhD, and co- mentors Liz Speliotes, MD, PhD, MPH, and Jordan Shavit, MD, PhD, with Jun Li, PhD, as a collaborator, who have expertise in NAFLD, genomics, zebrafish, and extensive mentoring success. The 5-year career development plan includes formal coursework, professional development, and mentored research, with defined milestones to ensure productivity and a successful transition to independence. The estimated global prevalence of NAFLD is now ~25%, but medical therapies are limited in number and efficacy, partly due to incomplete understanding of its pathogenesis. A unique group of patients with mutations in encoding nuclear lamina components develop early-onset NAFLD that often progresses to steatohepatitis (NASH) with cirrhosis. Mechanisms of NAFLD/NASH due to nuclear lamina mutations are obscure, but their study may provide insights into NAFLD pathogenesis and avenues for future therapies. This proposal will test the hypothesis that genetic alteration of the nuclear lamina predisposes to NAFLD/NASH by altering nuclear protein organization and the transcriptome. The mentored research has two Specific Aims: AIM 1: Identify and validate variants in nuclear lamina- related genes in NAFLD/NASH cohorts. AIM 2: Establish new experimental models to study how LMNA variants alter the nuclear lamina, heterochromatin, and transcriptome to promote NAFLD/NASH. In completing these aims, Dr. Brady will analyze genomic data linked to electronic medical record data in unique NAFLD cohorts and develop new disease models that will provide mechanistic insights into lamina-related NAFLD and in vivo systems for drug screening. This will build to two R01 proposals: (1) to examine the mechanistic basis of NAFLD-associated variants identified in AIM 1 and (2) to examine the mechanisms and therapeutic potential of screened compounds in the models generated in AIM 2. In addition to building a foundation for a programmatic line of research to understand the role of the nuclear envelope in liver disease, this K08 will provide Dr. Brady with research skills applicable to additional domains of hepatology and genomics and equip him to establish himself as an independent investigator and leader in this field. Alana Brennan, Ph.D., M.P.H.

BOSTON UNIVERSITY MEDICAL CAMPUS, BOSTON, MA

Evidence of the burden of diabetes, gaps in the diabetes cascade-of-care and the impact of care and treatment on diabetes outcomes in HIV-positive and HIV-negative patients in South Africa

Project Summary

This proposed five-year development period will be used for research and training activities that will give Dr. Brennan the skills and experience necessary to become an independent investigator. Dr. Brennan’s long-term career goals are to use high volume, high variety and high velocity large-scale datasets to provide evidence for the development of effective policies, programs and interventions to improve management and clinical outcomes of patients with chronic communicable (i.e. HIV and ) and non-communicable diseases (i.e. diabetes, cardiovascular disease and chronic kidney disease) at a national level in South Africa. Although Dr. Brennan is a clinical epidemiologist and methodologist by training, she recognizes the need and has a strong desire to acquire applied data science and “Big Data” analytical skills to stay competitive in the field. Building on her already strong background in quantitative data analysis, Dr. Brennan has proposed a training plan to develop skills in novel applied data science methods to deal with large-scale data sets, primary data collection, data validation and epidemiological approaches to causal inference. In addition, she has proposed training in the clinical monitoring, diagnosis, treatment and management of metabolic and cardiovascular conditions. The training plan will involve formal coursework, directed readings on specific topics, and strong mentorship from experts in the field of metabolic disorders and HIV, domestically and internationally. The proposed research will combine analyses of existing data from the National Health Laboratory Services in South Africa, the sole provider for public sector labs, and patient-level data from a primary health care clinic in the government sector. Upon successful completion of Dr. Brennan’s K01 award she will have started to build an invaluable resource to assess the burden of non-communicable chronic diseases in South Africa. Dr. Brennan’s high impact research is not only generalizable to other low- and middle-income countries, but would also be relevant to high-income countries where similar populations exist, but not in the numbers necessary to assess meaningful associations and interactions between desired exposures and outcomes. Through the creation of this resource, in addition to further strengthening her quantitative skill set and clinical knowledge of diabetes, Dr. Brennan will have developed competency as an independent researcher capable of securing R- series NIH funding. Kyle Burghardt, Pharm.D.

WAYNE STATE UNIVERSITY, DETROIT, MI

Acutely Induced Insulin Resistance by Antipsychotic Medication in Healthy Volunteers: Impact of Skeletal Muscle Epigenomic and Proteomic Mechanisms

PROJECT SUMMARY/ABSTRACT

This K23 Patient-Oriented Mentored Research Award builds on the candidate’s expertise in pharmacology and epigenetics to provide the support necessary to complete training in responsible conduct of research and three other needs: 1) clinical research design and execution, 2) in vivo insulin sensitivity measurement, 3) design and execution proteomics experiments. This training will be accomplished through a combination of expert mentor- ship, didactic (short courses and certificate program) and hands-on experiences (human insulin sensitivity meas- urements and proteomics). The career development activities and research will be mentored by Dr. Zhengping Yi (Primary) and Dr. Renu Kowluru, and supplemented by a physician key collaborator (Dr. Berhane Seyoum), consultants and a yearly, external scientific advisory committee. The candidate’s transition to independence will be aided by an R01 grant writing program, presentation of findings at national conferences and publication of results in high-impact journals. The research plan will provide a platform for training and address a gap in the understanding of how atypical antipsychotics cause insulin resistance. The long-term goal of the proposed work is to establish and sustain an independent career focused on the impact of molecular factors and mechanisms in medication outcomes. The objective is to determine the causal relationship between the observed skeletal muscle epigenetic and protein changes in response to atypical antipsychotics, and the development of insulin resistance using a randomized, placebo-controlled, 7-day trial of olanzapine in healthy volunteers. The central hypothesis, based on preliminary data, is that obesity- and mental illness-independent atypical antipsychotic- induced insulin resistance is caused by DNA hypermethylation and altered protein abundance and regulation in the skeletal muscle. The rationale for this work is that it will establish the molecular mechanisms that underlie atypical antipsychotic-induced insulin resistance, while providing the training and expertise for a research pro- gram to develop precise, tractable targets for future interventions to alleviate epigenetic and/or protein-based dysregulations. The central hypothesis will be tested by obtaining baseline and endpoint basal and insulin-stim- ulated skeletal muscle samples from the 7-day trial in the following specific aims: 1) Identify the relevant genes affected by the hypermethylation seen with atypical antipsychotic- induced insulin resistance and 2) Determine the relevant protein changes underlying obesity- independent atypical antipsychotic-induced insulin resistance. The approach is innovative, in the candidate’s opinion, because it seeks to change the status quo of assessing molecular changes in psychiatric populations already using antipsychotics and because it seeks to assess these changes in a tissue-specific manner using powerful epigenomic and proteomic approaches. The proposed work is significant because it is expected to have a positive impact both on the field, and in positioning the candidate for independence. This work will provide the basis for targeting the molecular dysregulations caused by antipsy-chotics and preventing mortality from insulin resistance and diabetes. Lilia Cervantes, M.D.

DENVER HEALTH AND HOSPITAL AUTHORITY, DENVER, CO

Improving Outcomes for Latinos on Hemodialysis with Limited English Proficiency

PROJECT SUMMARY/ABSTRACT

Latinos with End-Stage Renal Disease (ESRD) represent 17% of the US adult ESRD community and compared to non-Latino whites, they are 1.6 times more likely to develop depressive affect. Depressive affect among ESRD patients is associated with a reduced Health-Related Quality of Life (HRQOL), reduced renal therapy adherence, greater morbidity, and increased mortality. In addition, depressive affect is adversely affected by Limited English Proficiency (LEP) and many other social factors. With support from the Amos award from the Robert Wood Johnson Foundation (RWJF), Dr. Lilia Cervantes assessed the feasibility of a 5- visit lay Peer Navigator intervention to support Latino ESRD patients with social challenges and to activate them to seek mental healthcare. Her peer navigator intervention is feasible; however, her research showed that two-thirds of Latino ESRD patients with limited English proficiency report depressive affect yet do not seek care due to cultural preferences and language barriers. The proposed study will build on her Amos RWJF work by expanding the peer navigator intervention to include a bilingual Licensed Clinical Social Worker (LCSW) that will provide individual dialysis Chairside Cognitive Behavioral Therapy (CCBT) sessions while the peer navigator provides support with social factors. This patient-centered approach is culturally tailored and will bridge the gap to mental health services. The overall aims of this proposal are to: 1) engage key operational and clinical stakeholders early-on to develop a Peer Navigator-Licensed Clinical Social Worker (PN-LCSW) intervention; 2) conduct a pilot RCT of the PN-LCSW intervention versus standard care to test feasibility and acceptability; and 3) assess the efficacy of the intervention on depressive affect (primary outcome) as well as health-related quality of life and hemodialysis adherence (secondary outcomes). The candidate for this K23, Dr. Lilia Cervantes, is a 1st generation bilingual Latina and her long-term objective is to be an independent physician investigator and leader developing culturally tailored models of care for Latino ESRD patients with limited English proficiency to improve patient-centered and clinical outcomes. This K23 will provide Dr. Cervantes with the training to develop expertise in: 1) key stakeholder engagement; 2) clinical trial design; and 3) analysis of patient-centered and clinical outcomes including longitudinal data and mediation analysis. Dr. Cervantes will accomplish her research and training aims in the rich and supportive environment at the University of Colorado Anschutz Medical Center and Denver Health with the support from her excellent primary mentor Dr. Michel Chonchol (nephrology). Dr. Cervantes will make a significant contribution toward improving patient-centered and clinical outcomes among vulnerable populations with ESRD and limited English proficiency while obtaining the experience and training necessary to begin her career as an independent physician investigator. Teresa Chen, M.H.S., M.D.

JOHNS HOPKINS UNIVERSITY, BALTIMORE, MD

Biomarkers of Immune Activation in African Americans with CKD

PROJECT SUMMARY/ABSTRACT

Teresa K. Chen, MD, MHS is an Assistant Professor in the Division of Nephrology at Johns Hopkins University School of Medicine. She is applying for a K08 Mentored Clinical Scientist Research Career Development Award in order to acquire the necessary skills and mentored research experience to become an independent investigator in the field of chronic kidney disease (CKD). The proposed 5-year plan includes advanced coursework in epidemiology; practical experience in the measurement of biomarkers; and mentorship by an extraordinarily experienced, committed and diverse mentorship team [co-primary mentors, Lawrence Appel, MD, MPH and Morgan Grams, MD, PhD; co-mentor, Michelle Estrella, MD, MHS]. With resources provided by this award, Dr. Chen will develop proficiencies in study design, genetic and molecular epidemiology, and longitudinal data analysis related to CKD. Her proposed research project focuses on the role of immune activation in CKD progression and the interactive effects of immune activation with APOL1 risk variants. African Americans are disproportionately affected by CKD. This is in part due to the higher prevalence of APOL1 risk variants, genetic risk factors for kidney disease among individuals of African ancestry. The role of immune activation in the progression of non-diabetic CKD, particularly in APOL1 high-risk individuals, is unclear. The objectives of the proposed research are to: 1) study the associations of biomarkers of immune activation with CKD progression among non-diabetic African Americans with CKD attributed to hypertension; 2) determine whether the APOL1-associated risk for CKD progression is augmented by immune activation; 3) assess whether blood pressure interventions and dietary factors are associated with longitudinal changes in biomarkers of immune activation; 4) identify metabolomic predictors of biomarkers of immune activation. With over 10 years of follow-up, rigorously collected data, and stored biospecimens, the African American Study of Kidney Disease and Hypertension (AASK) represents an ideal cohort in which to study these potential associations. APOL1 genotyping and metabolomics measurements have previously been completed in AASK through other NIH-funded mechanisms. We propose to use stored serum samples from the baseline and 12- month visits of the trial phase to measure the following biomarkers of immune activation: tumor necrosis factor alpha (TNF-α), soluble TNF receptors 1 and 2 (sTNFR1 and sTNFR2), and interferon gamma (IFN-γ). The results of the proposed study will clarify the role of immune activation in CKD progression and perhaps identify novel targets for intervention. This could have important clinical implications in the treatment of African Americans with non-diabetic CKD, particularly among those with the APOL1 risk variants. The proposed research will also support Dr. Chen's long term-goal of transitioning towards an independent research career that ultimately improves the management and outcomes of African Americans with CKD. The rich training environment of Johns Hopkins University will ensure that she achieves these goals. Erika Cheng, Ph.D., M.P.H.

INDIANA UNIV-PURDUE UNIV AT INDIANAPOLIS, INDIANAPOLIS, IN

Obesity Prevention in Early Life (OPEL): Risk Screening and Targeted Intervention

Given the difficulty of reversing obesity once present, there has been increasing focus on the primary prevention of obesity early in the lifecourse. Few attempts have been made to prevent obesity during the first years of life. This proposal summarizes a 5-year program of mentored professional development tied to a multi-method research project intended to improve the identification and potential treatment of infants and toddlers with high risk for obesity. My long-term goal is to prevent obesity by identifying infants at greatest risk and providing for them an effective, family-centered intervention that targets modifiable, life course factors. My central hypothesis, based on my prior research, is that identifying infants at risk for obesity prior to the onset of unhealthy weight gain will enable early intervention. My research plan aims to: (1) create risk prediction models for obesity at age 24 months by linking three existing data systems that combine birth certificate, contextual- level, and health outcome data; (2) test the feasibility of linking these data prospectively to validate the Aim 1 obesity risk prediction models over a 24-month period within a contemporary, clinical cohort; and (3) identify best approaches for family-focused risk communication regarding the prevention of excessive weight gain and obesity in infants and toddlers using a human-centered design approach. Through my career development plan and guidance from my mentors, I will expand upon a foundation in epidemiology and pediatric health services research to develop expertise in machine learning, health informatics, data integration, qualitative methods, human-centered design, and behavior change. Together, the research and educational aims of this proposal will provide me with the necessary groundwork to compete for additional funding as an independent investigator. Specifically, I will seek R03-level grant funding from the NIDDK in year 4 of my K01 award to test the communication strategy developed in Aim 3 and to partner with families to modify an existing behavioral change intervention for use in infancy. By the end of this award, I will be well positioned to apply for funding from the NIDDK to conduct a robust R01-level study that combines the validated prediction models, family- focused communication strategy, and modified intervention to determine whether we can effectively prevent obesity in those infants and toddlers identified as being at the highest risk. This line of research will help ensure that prevention efforts are deployed in an efficient, cost-effective manner and accepted by those who need them. I will accomplish this work under the mentorship of Dr. Aaron E. Carroll, a child health services researcher, and a multidisciplinary team of faculty with expertise machine learning, health informatics, data integration and surveillance, qualitative research, human-centered design approaches, behavior change, and childhood obesity. I am ideally suited to complete this research due to my past research productivity, current mentorship team, open access to health care data, and the established clinical decision support infrastructure at the Indiana University School of Medicine. Pui (Susan) Cheung, M.S., D.O.

MASSACHUSETTS GENERAL HOSPITAL, BOSTON, MA

Characterization of Novel Signaling Pathways Involved in Water Balance Disorders

PROJECT SUMMARY/ABSTRACT

Maintenance of water homeostasis is a vital function of the kidneys and is essential for adaptation to terrestrial life. To reabsorb water effectively, vasopressin (VP) is released to induce aquaporin-2 (AQP2) phosphorylation and cytoskeletal remodeling within kidney principal cells in the collecting ducts, which increases apical membrane expression of AQP2. Dysregulation of AQP2 trafficking results in disorders of water balance; decreased AQP2 membrane expression causes nephrogenic diabetes insipidus (NDI), whereas an increase in plasma membrane AQP2 is associated with fluid retention in the syndrome of inappropriate ADH secretion (SIADH), congestive heart failure and cirrhosis. While VP/cAMP/PKA is the major signaling pathway that facilitates AQP2 membrane trafficking and water reabsorption, the process is in fact far more complex, and can be induced or inhibited by other signaling pathways. One such “alternative” pathway involves the epidermal growth factor receptor (EGFR), whose inhibition induces AQP2 membrane accumulation and phosphorylation similar to VP, but bypasses V2R, cAMP and PKA, and my goal is to deepen our understanding of the crosstalk between VP and EGFR pathways that modulate AQP2, with a long term career goal to characterize the role of this novel signaling pathways in dysregulated water retention observed in patients with SIADH, congestive heart failure and liver cirrhosis, in order to eventually design therapies to alleviate disease symptoms encountered in the clinic. For the last year and a half, I have been making significant progress in dissecting the pertinent pathways between VP and EGFR that regulate water balance. However, as I am focusing on my research career development, life not only brings us surprises, but also concerns about interruption in research progress. Therefore, I am applying for this new NIH- NIDDK Administrative Supplement in order to hire a technician during my maternity leave, expected to span between early to mid-July to early October 2020, in order to preserve or even increase research productivity. The Brown Laboratory, including my mentor Dr. Dennis Brown, my close collaborator Dr. Richard Bouley, and I will maximize the intellectual and technical resources to ensure progress during my leave, and I will remain updated on the progress to resume full productivity upon my return. This research proposal is carved out of Aim 1 of my parent K08, and the proposed experiments are carefully designed and expected to be completed by the end of the supplemental period. Obtaining this supplement funding would provide highly valuable support at this critical time of my research career. Christina Ching, M.D.

RESEARCH INST NATIONWIDE CHILDREN'S HOSP, COLUMBUS, OH

Urothelial IL-6 Signaling in the Host Defense Against Urinary Tract Infections

PROJECT SUMMARY/ABSTRACT

UTIs are a common source of patient morbidity that can result in permanent renal damage. Historically, antibiotics have been the standard in UTI treatment and routinely used for prevention. However, the emergence of multidrug resistant uropathogens limits the long-term viability of antimicrobial strategies based solely on antibiotic therapy. As a result, alternative methods for UTI treatment and prevention are needed. The lining of the urinary tract, or urothelium, serves essential roles in the detection and elimination of invading bacteria, including production of cytokines and antimicrobial peptides (AMPs). These intrinsic qualities of innate defense within the urothelium might be harnessed therapeutically to prevent and treat UTI. A fundamental knowledge gap exists, however, in our understanding of how these different factors contribute to limiting UTI susceptibility. The objective of this research project is to define the mechanisms by which the bladder urothelium defends the urinary tract from invading uropathogenic bacteria. This proposal tests the hypothesis that the cytokine interleukin-6 (IL-6) promotes clearance of uropathogenic Escherichia coli through activation of the Signal transducer and activator of 3 (Stat3) transcription factor and AMP production. The anticipated outcome of this research is to establish the contributions of the IL-6/Stat3/AMP signal transduction cascade to the host defense properties of the urothelium. Aim 1 will investigate how IL-6 directly limits urothelial susceptibility to UTI. Aim 2 will establish the roles for urothelial Stat3 during UTI. Aim 3 will explore the impact of AMPs in preventing UTI. Together, these Aims look to implicate the IL-6/Stat3/AMP pathway as a key driver of urothelial susceptibility to UTI and thus as a potential target of UTI treatment. The completion of this award will combine structured career development activities and mentored research programs to enable the transition from a junior investigator to an independently funded surgeon-scientist. Timothy Chlon, Ph.D.

CINCINNATI CHILDRENS HOSP MED CTR, CINCINNATI, OH

The Role of DDX41 in Inherited Myelodysplastic Syndromes

Project Summary

Myelodysplastic syndromes (MDS) are genetic disorders caused by impaired hematopoietic stem/progenitor cells (HSPC), which can transform to acute myeloid leukemia (AML). MDS typically occurs in the ageing population, however MDS can also manifest as a result of inherited germline mutations in younger individuals. Mutations in the DEAD/H-box gene DDX41 are among the most common alterations associated with inherited MDS. Inherited DDX41 mutations are heterozygous and are typically frameshifts, suggesting that these mutations result in loss of DDX41 function. DDX41 mutations are also observed in de novo MDS and AML, and are typically missense mutations frequently resulting in the amino acid substitution R525H. DDX41 is an RNA helicase that hydrolyzes ATP, and can function as an innate immune sensor, RNA splicing factor, and regulator. The precise mechanism(s) by which DDX41 mutations alter HSPC function and contribute to MDS/AML remains unknown. As such, the proposed project will define the role of DDX41 mutations in the pathogenesis of MDS/AML. To mimic the frameshift mutations observed in human MDS and determine the role of DDX41 in normal hematopoiesis, we generated hematopoietic-specific and conditional Ddx41- deficient mice. Our preliminary data has revealed that DDX41-deficiency (complete knockout) is not compatible with HSPC function and hematopoiesis, whereas DDX41 heterozygosity increases BM HPSC. In addition, through integrative proteomic and biochemical approaches we identified a novel DDX41- interacting protein in leukemic cells, SAMHD1, a dNTPase and host defense factor, which controls cellular pools of dNTPs. Collectively, our preliminary data indicate that DDX41 has a critical role in hematopoiesis and HSPC function. We hypothesize that diminished DDX41 expression and/or function contributes to ineffective hematopoiesis and to the pathogenesis of MDS and AML, in part due to increased SAMHD1 activity and altered cellular dNTP pools. Therefore, the objectives of this proposal are to model somatic and germline DDX41 mutations in normal and malignant hematopoiesis, and to elucidate the molecular function of DDX41 required for HSPC function. Through extensive hematopoietic approaches, we will define the role of DDX41 deficiency and R525H expression in MDS incidence and progression to AML (Aim 1). Since DNMT3A and DDX41 mutations commonly co-occur in MDS patients that have progressed to AML, we will determine whether DDX41 heterozygosity or R525H expression combined with DNMT3A-deficient mice will result in high-risk MDS or overt AML. Reduced cellular dNTP pools impair cell cycle progression and result in genomic instability; therefore, we will determine the role of SAMHD1 activity in DDX41-deficient HSPC or mutant MDS, and whether diminished cellular dNTPs pools lead to genomic instability and development of AML (Aim 2). By elucidating the function of DDX41 malignant hematopoiesis, we predict to uncover novel mechanisms underlying inherited and de novo MDS. Melissa Crane, Ph.D.

RUSH UNIVERSITY MEDICAL CENTER, CHICAGO, IL

Engaging men from blue-collar industries in weight loss: Tailoring recruitment and treatment approaches

ABSTRACT

Men working in blue-collar industries (e.g., machine operation, transportation, construction, etc.) have a high prevalence of overweight and obesity and have high rates of comorbidities associated with obesity. Unfortunately, these men are unlikely to participate in evidenced-based weight loss interventions. To improve their participation in these programs, a multidisciplinary approach is needed. To address this need, I am applying for this Mentored Research Scientist Development Award to gain additional scientific and technical skills in 1) qualitative methods, 2) communication science, and 3) health economics and apply these skills to intervention development and evaluation. My mentoring team includes psychologists with expertise in behavioral intervention development (Drs. Appelhans and Powell), a men’s health communication and qualitative methodology expert (Dr. Baglia), and a health economist (Dr. Walton). Along with my mentoring team, I have developed a rigorous training plan that will allow me to simultaneously fill the gaps in my training while conducting impactful work focused on an understudied population. I will investigate how to develop recruitment messages that encourage participation in weight loss programs and adapt behavioral weight loss strategies for use among men working in blue-collar industries. Aim 1 of this project is to determine how to tailor recruitment messages and intervention components to maximize reach and engagement of men in weight loss programs. I will conduct semi-structured interviews and focus groups with men from the target population followed by an online closed-response survey. The survey will include a discrete choice which provides data to develop models of participants’ relative preferences for intervention and message components. The second aim is to test the feasibility of using refined messages and intervention components in a randomized trial. The results of this project are crucial for developing appropriate ways to reach this high burden and underserved population and will yield compelling preliminary data for my R01 application. Further, by completing this project I will have the training to launch my career as an independent investigator focused on developing behavioral interventions for underserved populations. Clair Crewe, Ph.D.

UT SOUTHWESTERN MEDICAL CENTER, DALLAS, TX

Extracellular Vesiclemediated Regulation of Metabolism

Project Summary/Abstract

Adipose tissue dysfunction is at the forefront of metabolic disturbances in obesity and type II diabetes, making it a promising target for pharmacological intervention. This active, energy-sensing, endocrine organ secretes number of factors that have profound effects on systemic metabolism, in addition to its role in sequestering potentially toxic lipids species. Proper function of adipose tissue is maintained by cross-talk between resident tissue cells including adipocytes, endothelial cells, immune cells and fibroblasts, a process that is disrupted in the obese condition. The efficiency at which the adipose tissue responds to nutrient stresses can mean the difference between sustained whole-body metabolic homeostasis or pathology. My recently published work describes the finding that cells in adipose tissue exchange extracellular vesicles (EV) that are rich in signaling proteins, lipids, and, potentially miRNAs. These transfer events are dominated by an endothelial-to- adipocyte axis; however, adipocytes also secrete EVs that are taken up but other cells in the tissue such as macrophages or mural cells. Furthermore, we found that under the energetic stress of fasting, endothelial cell EV secretion is enhanced and targeted to adipocytes. This work has opened up vast potential for the discovery of novel signaling pathways between these cells that may provide an understanding of what pathways support healthy vs maladaptive adipose tissue remodeling under the nutrient stress of obesity or fasting. Preliminary data suggests that endothelial cell EVs support adipocyte ATP production during mitochondrial energetic stress by increasing adipocyte glycolytic reserve. Furthermore, adipocyte EV production is enhanced in the context of mitochondrial dysfunction, which we predict will regulate systemic metabolism. Thus, I hypothesize that under energetic stress, adipose tissue endothelial cells and adipocytes work synergistically through EV production to modulate whole body metabolism. The first Aim will test the hypothesis that endothelial cell EVs reprogram adipocyte metabolism to promote efficient adaptation of the adipocyte to metabolic stress. Aim 2 will evaluate the concept that energetic stress stimulates adipocytes to secrete EVs that alter systemic metabolism. The general approach will take advantage of both in vitro techniques as well as recently generated mouse models of cell-specific mitochondrial dysfunction or cell-specific suppression of EV production. This proposal will give me the opportunity to be trained in metabolic tracing techniques, mouse physiology, and human tissue acquisition by highly skilled specialists in the Scherer lab and throughout UT Southwestern. Successful completion of these aims has the potential to open a new area of research to decipher EV- mediated signaling pathways in metabolic regulation. Oriana Damas, M.D.

UNIVERSITY OF MIAMI SCHOOL OF MEDICINE, CORAL GABLES, FL

The impact of diet patterns and PUFA-related polymorphisms on ulcerative colitis in Hispanics

Ulcerative colitis (UC) is a devastating immune- mediated disease that is on the rise globally. In our prior study, we found that Hispanics have less severe disease than non-Hispanics, but yet within one US-born generation develop a disease course that is similar. Differences in adoption of a Western diet between US- born and foreign-born Hispanics could explain differences in UC severity observed. A Western diet, high in n-6 to n-3 polyunsaturated fatty acids (PUFA), is up to 10-20 times the recommended intake and is considered pro-inflammatory. However, heterogeneity of results in dietary intervention studies suggests that underlying genetic variation in PUFA metabolism may modify diet effects. Our preliminary data indicates that Hispanics with UC have polymorphisms in PUFA not present in Hispanic controls. In UC, disease activity measured by rectal bleeding, number of bowel movements, and urgency correlates with endoscopic inflammation. I hypothesize that Hispanics with UC whose diet follows a higher Western dietary pattern, defined by a reproducible dietary pattern score, have more active disease, than those whose diet scores are lower. Further, I propose that carriage of PUFA variants augments the effect of a Western diet on relapse leading to a disproportionate pro- inflammatory effect of the diet. In this study, I examine the influence of a Western dietary pattern on remission and relapse of UC among Hispanics (Aim 1). We will test whether PUFA variants influence relapse and enhance the effect of a Western diet on UC relapse (Aim 2). A total of 320 self- identified, Hispanic participants with UC are followed at our GI clinics and we will include these for our diet portion in Aim 1. We will include a total of 691 Hispanics with UC and 900 Hispanic controls for our Aim 2 genetics portion. We have detailed demographic and clinical information on these patients, as well as whole genome sequencing of PUFA variants. Participants will be asked to complete a validated food-frequency dietary questionnaire, at baseline and at 1-year follow up. Disease activity using the validated simple clinical colitis activity index will be assessed at baseline and at 1 year follow up in combination with fecal calprotectin. We will calculate scores for a Western dietary pattern and determine the influence of diet on relapse using multivariable regression analysis. In Aim 2, we will examine the effect of PUFA variants on relapse and also determine gene-environment interactions influencing disease activity between PUFA variants and Western diet. Results gathered from this study will be a step towards personalizing IBD treatment by targeting specific dietary patterns in Hispanics with highest metabolic genetic risk. Dawn Eichen, Ph.D.

UNIVERSITY OF CALIFORNIA, SAN DIEGO, LA JOLLA, CA

Novel Executive Function Training for Obesity

PROJECT SUMMARY/ABSTRACT

Current behavioral obesity treatments (e.g., behavioral weight loss; BWL) are not effective for nearly 80% of people in the long term. Accordingly, there is a pressing need to develop novel treatments to target alternative mechanisms that might be interfering with the success of current treatments. Growing research has identified significant neurocognitive deficits, particularly in relation to executive function, among adults with overweight or obesity. Compensatory Cognitive Trainings have been used to improve executive function in various populations including schizophrenia and traumatic brain injury, as well as normal aging adults. In line with a recent NIH Working Group Report, that concluded there is a great need for a “deeper understanding of cognitive function” in regards to its impact on weight loss and maintenance, this application seeks to develop a Novel Executive Function Treatment (NEXT) for obesity. NEXT will be adapted from previous compensatory cognitive trainings and will be iteratively pilot tested with 20 adults to refine the treatment based on initial qualitative feedback. Then, a preliminary randomized control trial will compare NEXT prior to BWL (NEXT+BWL) to a nutrition education comparison group prior to BWL (CON+BWL) to evaluate initial feasibility and acceptability of NEXT and NEXT+BWL. Lastly, NEXT+BWL will be compared to CON+BWL on weight loss, attendance and executive function outcomes at post-treatment and at a 6-month follow-up. The proposed research aims to advance the field’s understanding of current obesity treatment failure. The findings will be used as a basis for the applicant’s future R01 proposal. The proposed study fits well with the applicant’s career development goals. The candidate has a strong background in obesity and eating disorder research with prior clinical training in neuropsychology. The overarching goal of this 5-year training program is to evolve the candidate into an independent clinical scientist. The specific training goals include: 1) training in treatment development and alternative study design to evaluate new treatments; 2) advanced training in neuropsychology related to obesity and neuropsychological assessment, and; 3) training in advanced statistics, with a strong focus on how to manage and analyze data for longitudinal treatment trials. The mentorship team consists of internationally-recognized experts in obesity, compensatory cognitive treatment, and statistics who will oversee the execution of the training plan and foster career development. Research and training will occur at the University of California San Diego, which is a ripe environment for fostering junior investigators through a transition to independence. This award will provide the required time, funding, and training needed to broaden the candidate’s expertise in the obesity field while simultaneously allowing her to become an independent investigator. As such, this proposal represents a critical first step in developing novel behavioral interventions for obesity with a focus on neurocognitive deficits. Laya Ekhlaspour, M.D.

STANFORD UNIVERSITY, STANFORD, CA

Modeling and modulating insulin delivery in automated insulin delivery systems to accommodate for meal compositions

PROJECT SUMMARY

The candidate, Laya Ekhlaspour, MD, is dedicated to advancing diabetes management by decreasing the burden of diabetes care. This proposal will provide a structured clinical research training experience with formal mentorship that will enable Dr. Ekhlaspour to become an independent clinical researcher with expertise in closed loop systems. Currently, carbohydrates are considered the primary and major determinant affecting postprandial glucose control and the insulin bolus is based solely on the carbohydrate content of the meal along with the blood glucose at the time of the bolus. Given the known benefits of normalizing postprandial glucose excursions, the goal of this research is to specifically address problems related to providing adequate insulin coverage for meals that have variable fat and protein content that might result in both early hypoglycemia and prolonged hyperglycemia, when automated insulin systems are used. This proposal will take advantage of existing data collected during an at the Barbara Davis Center, and the feasibility closed-loop trials at Stanford. In order to characterize the effect of macronutrient food content on postprandial glucose levels, a retrospective review of meals with known glucose, protein and fat content combined with CGM and insulin values in both open loop and closed loop situations will be conducted. This analysis will provide data to allow modeling for insulin requirements with meals of variable patterns of postprandial glycemic levels. It will also determine whether this modeling will allow for setting the percent of insulin required upfront and how long the insulin delivery needs to be extended in pumps which allow for extended meal boluses. The result of this analysis can be used in closed-loop control real-time modeling of meal boluses when there is a real-time adaptation to food absorption patterns, which will contribute to eventual fully closed-loop glucose control. The developed model will be based on assessing the hourly glucose and insulin requirements which could be integrated into prandial dosing algorithms in closed loop systems in order to optimize postprandial glycemic control. The long-term goal is to have a fully-closed loop algorithm, which will recognize the need for additional insulin with a high fat or protein meal based on the CGM postprandial tracing and insulin requirements during closed-loop control. This will reduce the burden of diabetes management significantly because the patient does not have to announce a meal. The proposed studies will provide the preliminary meal data for modeling of meal-responses that will account for both the carbohydrate, protein and fat content of the meal without user input. The following step will be to validate this model through experiments and then conduct a randomized, controlled trial of the implementation of a potential refined meal algorithm that could handle postprandial glucose levels without an extended bolus as part of a competitive R01 grant application. Amy Engevik, Ph.D.

VANDERBILT UNIVERSITY MEDICAL CENTER, NASHVILLE, TN

The role of Vb in hepatocyte protein trafficking

PROJECT SUMMARY

Background and aims: Inclusion Disease (MVID) is a form of congenital diarrhea caused by inactivating mutations in Myosin Vb (MYO5B). The available treatment options for individuals with MVID are either lifelong total parenteral nutrition or full intestinal transplantation. MVID patients frequently present with cholestasis. This cholestasis was previously thought to arise from prolonged administration of total parenteral nutrition. However, recent publications have described mutations in MYO5B that do not result in MVID, but instead patients have isolated cholestasis. Given the complex nature of MVID and the limited treatment options available, understanding the pathogenesis of cholestasis resulting from mutations in MYO5B represents an important scientific question which needs to be addressed. The central hypothesis of this research proposal is that mutations in MYO5B result in aberrant expression of apical/canalicular membrane transporters preventing the normal secretion of bile salts. We base our hypothesis on preliminary data generated from our mouse and pig models of MVID which demonstrate mislocalization of BSEP and other canalicular transporters. Our findings are mirrored by published reports from patients with mutations in MYO5B, demonstrating aberrant expression of the bile salt export protein (BSEP) in hepatocytes. To date, no experimental animal model has been used to define the mechanism of cholestasis in the setting of MVID. For this K01 Career Development Award, I propose the use of germline MYO5B KO mice, an MVID pig model and a novel mouse model of MYO5B point mutation to address deficits in apical transporters in hepatocytes that arise from mutations in MYO5B. Specific Aim 1 will define the function of MYO5B in maintaining hepatocyte polarity and homeostasis. Specific Aim 2 will determine the mechanism by which the C266R mutation in MYO5B contributes to cholestasis, but does not result in MVID. At the completion of these studies I expect to have elucidated the role of MYO5B in the regulation of protein trafficking in hepatocytes in vivo and in vitro. This proposal highlights the need for a better understanding of the function of MYO5B in hepatocytes with the ultimate goal of improving current therapeutic treatments for MVID. Long-term objective and aims: Being the recipient of a K01 Career Development Award would provide the mentorship, training and support necessary to achieve my goal of becoming an independent investigator. This research is well suited for the National Institute of Diabetes and Digestive and Kidney Diseases as it relates to digestive and liver disorders. Vanderbilt University Medical Center offers all of the scientific resources required to complete this proposal. I have assembled a group of renowned scientists to serve as my mentors and mentorship committee, and as collaborators. Additionally, I have developed a training plan to enhance my scientific repertoire, increase my publication record and secure independent funding. This will ensure success in securing an independent position to start a new laboratory by the completion of this award. Raquel Espin Palazon, Ph.D.

IOWA STATE UNIVERSITY, AMES, IA

The impact of inflammation on hematopoietic specification

Hematopoietic stem cells (HSCs) are rare cells within human bone marrow that are responsible both for the life-long replenishment of all blood cell lineages and for the curative effects of bone marrow transplantation. The creation of human induced pluripotent stem cells holds great promise for cellular regeneration therapies, but it is not currently possible to instruct these cells to generate HSCs in vitro. The goal of this application is to determine how pro-inflammatory signaling via NF-kB instructs HSC fate in the vertebrate embryo, and how this process is regulated by Progranulin a (Pgrna), with the ultimate goal of replicating HSC generation in vitro for clinical utility. My working hypothesis is that early pro- inflammatory inputs converge to activate NF-kB, which in turn activates key signaling events in the specification of HSC fate. These pro-inflammatory signals need to be downregulated soon after HSC specification; my preliminary results suggest that Pgrna functions in this manner. To test these hypotheses, this proposal consists of 2 aims: (1) Characterize the role of NF-kB in HSC specification; and (2) identify the role of Pgrna in HSC emergence. This study will be conducted in zebrafish, which are an ideal system for direct visualization of HSCs and have served as a to study human disease. To achieve this application’s goals, novel transgenic and mutant lines will be generated, and qPCR, FACS-sorting, RNA-seq and confocal microscopy techniques will be utilized. Dr. Espin Palazon is a postdoctoral fellow in David Traver’s laboratory at UCSD whose ultimate career goal is to lead an independent research group focused on stem cells at a major research institution. Her short- term goals are (1) to determine the spatiotemporal requirements of NF-kB within hemogenic endothelium, and the downstream genes regulated to specify HSCs; and (2) to elucidate how Pgrna governs HSC emergence. She was recruited to join Dr. Traver’s group because of her strong background in and the zebrafish animal model. The project outlined in this proposal will allow Dr. Espin Palazon to transition from a mentored scientific position to an independent research career, helping her gain expertise in FACS sorting, RNA-seq, genome editing techniques, and HSC biology, all of which are fundamental to establish her independent lab. Dr. Espin Palazon will meet and present her work to experts in development, immunology and hematology, in addition to presenting her data to a formal mentorship committee comprised of senior experts that will aid her transition to an independent researcher. UCSD offers numerous courses that Dr. Espin Palazon will attend, as well as seminars on career development and laboratory management. The vibrant, collaborative scientific atmosphere at UCSD is an ideal environment for Dr. Espin Palazon to develop during the mentored phase of her award, and will be instrumental in forming the foundation for the future success of these studies. She is well poised to execute the proposed work, achieve her career development and training goals and to contribute high impact research to the scientific community. Di Feng, Ph.D.

BETH ISRAEL DEACONESS MEDICAL CENTER, BOSTON, MA

The interaction between mechanical forces and cytoskeletal impairments in podocyte mediated kidney disease

Project Summary/Abstract

This K01 grant proposal describes a five-year mentored training program designed to transition Dr. Di Feng to become an independent academic investigator. Dr. Feng obtained her Ph.D. at the Medical College of Wisconsin under the mentorship of Dr. Allen Cowley. She is now completing her postdoctoral fellowship in the lab of Dr. Martin Pollak, an international leader in studying the genetics of glomerular kidney disease. Dr. Feng has focused her research on elucidating the mechanism by which mutations in ACTN4 – an important protein – lead to a form of glomerular kidney disease called focal segmental glomerulosclerosis (FSGS). The inability to better characterize the podocyte dysfunction that underlies FSGS has hindered the field in establishing more specific, personalized treatments beyond broad immunosuppression and anti- hypertensive therapy. Dr. Feng has focused her research on the mutant podocyte’s response to the mechanical stresses it experiences while filtering blood flow in the glomerulus. She has so far shown that the biophysical changes conferred by disease-causing mutant ACTN4 render the podocyte brittle, exhibiting failure of contractile forces and actin cytoskeleton disruption in response to periodic stretch. In the current proposal, Aim 1 seeks to further define the impaired response of human podocytes caused by mutant ACTN4, not only to stretch but also to shear stress. She will employ organ-on-a-chip methods to better simulate these stresses while quantifying the associated biomechanical and molecular responses of podocytes. Aim 2 will determine whether post- translational phosphorylation of ACTN4 also impairs the response of podocytes to mechanical stress, using mouse models and biomechanical studies of podocytes isolated from these mice. Aim 3 plans to use CRISPR/Cas technology to generate a mutant ACTN4 rat model and use intravital microscopy to measure the in vivo mechanical stresses within mutant and WT glomeruli. Through the proposed research, she will learn organ-on-a-chip methods, mass spectrometry, super-resolution imaging, and intravital microscopy. She has assembled a team of mentors and advisors under Dr. Pollak entailing leaders in these respective disciplines, including Dr. Donald Ingber, Dr. Bruce Molitoris, Dr. Hanno Steen, Dr. Douglas Richardson, as well as Dr. Roger Tung, who will provide advice related to the translational value of her work. Dr. Feng will spend 95% of her time under this award toward the proposed research, and her training plan includes didactic courses, seminars, and career development workshops at Harvard. The proposed project will make Dr. Feng competitive for independent research awards, for which she plans to apply her findings from ACTN4 and the above multidisciplinary methods to further study how defects in the actin-based cytoskeleton impair the podocyte’s response to the mechanical stresses experienced in vivo. The advancement of her goals will take place within Harvard’s vast resources and connections to thought-leaders, situated in the unique environment of Boston that integrates academics and industry. Aaron Fobian, Ph.D.

UNIVERSITY OF ALABAMA AT BIRMINGHAM, BIRMINGHAM, AL

A sleep and media intervention to improve adolescents weight and risk of type 2 diabetes

Project Summary

Dr. Davis-Fobian's long-term goal is to become an independent, patient-oriented investigator with a focus on implementing and evaluating interventions to address childhood and adolescent obesity and prevent type 2 diabetes. Protected time provided by successful completion of this K23 career development award will provide the foundational support necessary for achievement of this goal. The long-term objective of this research is to establish an effective intervention incorporating sleep to improve adolescent health and reduce obesity and risk for the development of type 2 diabetes. Dr. Davis-Fobian is an Assistant Professor in the Department of Psychiatry at the University of Alabama at Birmingham (UAB). Her position allows her to interact with distinguished researchers on a daily basis and provides her with the opportunity to utilize the most innovative technology. Dr. Davis-Fobian is an experienced clinical psychologist who has extensive experience in treating obese adolescents with or at risk for type 2 diabetes. It was through these clinical experiences that Dr. Davis-Fobian realized the need for new approaches to obesity treatments and developed her hypothesis that improving sleep may assist in adolescent weight loss and prevention of type 2 diabetes. The foundation of Dr. Davis-Fobian's successful career development is the distinguished mentoring team she has developed for her project, consisting of Drs. Barbara Gower, David Allison and Karen Gamble and her scientific advisory panel. Their expertise complements all areas in which Dr. Davis-Fobian would benefit from additional training. These mentors are all faculty at UAB and have a strong history of research collaborations. Career development planned for this award includes 1) training in insulin sensitivity, beta-cell function, chronobiology, biostatistical and clinical trials research and research ethics; 2) laboratory training in metabolism, body composition, and chronobiology; and 3) professional skills training. These skills will be fostered through coursework, short courses, didactic opportunities and hands-on experiences, allowing Dr. Davis-Fobian to ultimately become a nationally recognized obesity and type 2 diabetes prevention researcher. The purpose of the proposed study is to assess the effects of a sleep and media use intervention on body composition and risk for type 2 diabetes in obese adolescents aged 15-17. This will be accomplished through 2 specific aims: 1) Conduct a two-arm parallel group randomized controlled trial of the relative effects of an intervention for adolescent sleep duration and media use on sleep duration and quality, insulin sensitivity, appetite-regulating hormones, and body composition; and 2) Evaluate whether the sleep intervention affects adolescents' nighttime media use at both the end of the intervention and the 3-month follow-up. Craig Forester, Ph.D., M.D.

UNIVERSITY OF COLORADO DENVER, Aurora, CO

Profiling Translational Control of Gene Expression and Nascent Proteomic Responses in Erythropoietic Progression and Ribosomopathies

SCIENTIFIC ABSTRACT

Erythropoiesis is guided by a complex interplay of signaling cues triggering expression of gene networks necessary for production of mature red blood cells. Rapid response to these signals in erythroid precursors temporally orchestrate functions required for erythroid differentiation. Timely and appropriate selection of mRNAs for translation by the ribosome is essential for maintaining the balance between maintaining erythroid precursors differentiation. This concept, known as translational control governs efficiency of mRNA translation and thus plays a crucial role in regulating responses to extracellular cues such as anemia and hypoxia. Failure of the ribosomal machinery to regulate this process underlies the ribosomopathies, a set of human diseases resulting from ribosomal mutations leading to profound consequences for hematopoietic maturation. This proposal focuses on outstanding, fundamental questions in how translational control 1.) directs mRNA selection in normal erythropoiesis and 2.) is impaired in ribosomopathies in response to signaling cues. Aim 1 will explore how eIF4E, a key factor in translation initiation is repressed in a dynamic, cell-intrinsic manner to direct translation of a specific set of transcripts. Failure to repress eIF4E activity in maturing erythroblasts leads to inappropriate expression of genes necessary for maintenance of early erythroid precursors and impaired differentiation. This aim will mechanistically decipher motifs in target mRNA and decipher how eIF4E specifically recognizes these transcripts for ribosomal recruitment. Dependence of tight regulation of eIF4E on erythropoiesis will be tested in vivo employing a novel mouse model utilizing doxycycline- responsive expression of eIF4E localized to specific phases of hematopoietic maturation. A paradigm example of ribosomopathies, Diamond Blackfan Anemia (DBA) results in erythroid aplasia and failure to respond to erythropoietin (Epo), a key signal in differentiation of early erythroid precursors. However, why hematopoietic precursors in DBA fail to generate the appropriate nascent gene expression response to these cues is still poorly understood. Aim 2 will use employ a universally-applicable, mass spectrometry-based method (OPP-ID) pioneered by Dr. Forester to characterize the nascent proteomic landscape in healthy and DBA-derived iPSCs in response to erythropoietin. Identification of impaired proteomic responses in DBA iPSCs will give insight into how specific ribosomal mutations prevent translation of key genes crucial in early differentiation. This technology will be applied to understand how dexamethasone, a known mediator of hematopoietic rescue of unclear mechanism, remodels the erythroid proteome in response to Epo. Successful completion will result in both a unique contribution into the fields of translational control and erythropoiesis as well as a novel approach to understanding how the nascent translatome is impaired in ribosomopathies. Accomplishment of this proposal is crucial in Dr. Forester’s long-term goals of becoming an independently funded researcher and an expert in the field of proteomic networks governing early hematopoiesis. Krystle Frahm, Ph.D.

UNIVERSITY OF PITTSBURGH AT PITTSBURGH, PITTSBURGH, PA

Impact of CREBRF and its obesity-risk variant on hypothalamic glucocorticoid and neuroendocrine output using molecular, cellular, and physiological approaches.

PROJECT SUMMARY

Strong evidence links central hypothalamic neuroendocrine output to complex biological processes and behaviors that control energy and metabolic homeostasis. Dr. Kershaw’s collaborative research group recently identified a novel obesity-risk variant in a putative transcriptional regulator, CREBRF, that is highly expressed in the hypothalamus. Although virtually nothing in know about CREBRF, it has been linked to glucocorticoid signaling, raising that possibility that its energy and metabolic effects in humans is mediated, in part, by influencing hypothalamic glucocorticoid and/or neuroendocrine output. The overarching goal of this proposal is to determine how CREBRF and its obesity-risk variant contribute to the central regulation of energy homeostasis and the stress response. The overall objective, which is the next step in the pursuit of this goal, is to characterize the role of this gene/variant in central hypothalamic glucocorticoid and neuroendocrine output. The central hypothesis is that CREBRF is expressed and regulated in key hypothalamic neurons and that the loss of CREBRF or expression of its missense CrebrfR457Q will differentially impact the metabolic control transcriptome and the chromatin landscape within the hypothalamus, thereby influences behavioral and metabolic outcomes. This hypothesis is based on the following: 1) endogenous CREBRF is highly expressed in the central nervous systems including the hypothalamus where it co-localizes with the glucocorticoid receptor (GR); 2) ectopic expression of CREBRF in cells influences subnuclear GR targeting and stability; 3) global CrebrfKO mice have dysregulated glucocorticoid signaling and abnormal postpartum maternal behavior; and 4) global CrebrfKO mice have reduced body weight. The above hypothesis will be tested using molecular, cellular, and physiological approaches with the following specific aims: 1) To characterize the impact of CREBRF and its obesity risk variant (CREBRFR458Q) on hypothalamic output in vivo using murine models; and 2) to determine the function of CREBRF and its obesity risk variant (CREBRFR458Q) on chromatin accessibility and cell-specific transcriptome using murine models. This research is innovative because 1) it examines obesity in a murine model without the complications of diabetes, 2) examine the mechanisms for the phenotype observed in Samoans and 3) will lay the foundation for numerous avenues to pursue the potential therapeutic benefits of targeting central CREBRF. This contribution will be significant because it will identify CREBRF expression, regulation, and function in specific hypothalamic neuropeptide-containing neurons and HPA-axis output during defined stress conditions and determine the impact of CREBRF and its obesity-linked variant on the genome-wide chromatin landscape and single cell transcriptomes within the hypothalamus. Such findings are expected to have a broad translational impact by improving strategies for prevention and/or treatment of obesity and associated metabolic diseases. Finally, at the completion of this proposal, Dr. Frahm will be well poised to lead her own independent research program in the field of neuroendocrinology. Dana Fuhrman, M.S., D.O.

UNIVERSITY OF PITTSBURGH AT PITTSBURGH, PITTSBURGH, PA

Renal Fitness in Young Adults with Congenital Heart Disease

ABSTRACT

Acute kidney injury (AKI) and chronic kidney disease (CKD) are strongly associated with morbidity and mortality across multiple patient populations. Given that there is currently no specific therapy for AKI or CKD once either has occurred, having a means to predict a patient at risk for renal injury in order to prevent or attenuate damage is invaluable. The overarching goal of this proposal is to predict AKI and AKI to CKD transition in high risk patients. As an assistant professor at the University of Pittsburgh in both pediatric nephrology and intensive care medicine, Dr. Fuhrman has a firm commitment to the study of measures to predict and ultimately prevent the AKI to CKD transition. The main goal of this study is to establish an objective method to quantify renal fitness in young adult congenital heart disease (CHD) patients, a growing population that is at risk for numerous kidney insults across a lifetime. A lack of renal fitness, defined by reduced glomerular reserve and increased susceptibility to tubular injury, may indicate a poor ability of the kidney to respond to external stressors. There currently is no readily available means to quantify renal fitness clinically. Dr. Fuhrman plans to quantify glomerular reserve in CHD patients ages 18-28 by measuring estimated glomerular filtrate rate (eGFR) values using cystatin C before and after a protein load, an easily replicated method that she piloted as a T32 Training Grant recipient along with her mentor, Dr. George Schwartz, during her nephrology fellowship years. When these patients are subsequently admitted to the hospital for coronary bypass (CBP) exposure, she plans to determine the association of glomerular reserve and AKI risk. In addition, prior to a CPB exposure, Dr. Fuhrman will assess the expression of tubular biomarkers, which have been previously shown to be early markers of kidney damage. The association of these biomarkers with AKI similarly will be determined at the time of hospital admission for CBP exposure. Patients will be followed for 1 and 2 years in order to determine if preoperative glomerular reserve and tubular biomarkers estimate the risk of eGFR decline and/or the development of proteinuria. No prior studies have examined the long- term effects of glomerular reserve or preoperative tubular biomarker values in any patient group. Dr. Fuhrman has assembled a mentoring team of internationally recognized researchers led by Dr. John Kellum, her current mentor and an internationally renowned researcher who has done extensive work on predicting AKI and the use of biomarkers. The strong institutional support along with the guidance of a multidisciplinary mentorship team that Dr. Fuhrman will receive under this award will provide her with the necessary foundation to become an independent investigator with expertise in methods to predict and prevent adverse kidney outcomes in numerous patient groups. Meghana Gadgil, M.P.H., M.D.

UNIVERSITY OF CALIFORNIA, SAN FRANCISCO, SAN FRANCISCO, CA

Prevention of Type 2 Diabetes through Diet and Metabolomics

PROJECT SUMMARY

Type 2 diabetes affects 1 in every 11 adults in the United States, and is a leading cause of death. High quality dietary intake is independently and strongly associated with lower risk of type 2 diabetes. The metabolic processes connecting dietary intake and diabetes, however, remain unclear. Understanding these biological processes between dietary intake and diabetes can help to establish new and effective individualized targets for treatment and prevention. The expression of compounds related to metabolism and cellular processes is called metabolomics. The objective of this proposal is to determine the identity of metabolites associated with a low-diabetes-risk dietary pattern, to investigate the association of these metabolites with incident diabetes, and the causal pathways related to the action of these metabolites, and to examine changes in the levels of these metabolites in a pilot dietary intervention with a low-diabetes-risk food. Aim 1 will use data and biospecimens from the Multi-Ethnic Study of (MESA), the Mediators of Atherosclerosis in South Asians Living in America (MASALA) study and the Rotterdam Study (RS) to determine the association of a low-diabetes- risk dietary pattern with known and novel metabolites. We will validate newly discovered metabolites within the Airwave cohort. Aim 2 associates these newly identified metabolites with incident diabetes and examines the extent to which they mediate the relationship between diet and diabetes. It also places the metabolites within a biological context using network analysis techniques. Aim 3 uses data and biospecimens from an existing pilot trial of a short-term walnut supplement in individuals with prediabetes to evaluate changes in metabolites with the intervention. The candidate for this Mentored Patient-Oriented Research Career Development Award, Dr. Meghana Gadgil, is a General Internist and an Assistant Professor in the Division of General Internal Medicine at the University of California, San Francisco. Dr. Gadgil is a physician-investigator with a strong background in nutrition and diabetes. In this proposal, she will examine novel associations between dietary intake, metabolite expression and type 2 diabetes incidence. This work will serve as the foundation for her overarching career goal: to become an independent clinical investigator focused on prevention of diabetes in high-risk populations through the use of dietary interventions. Dr. Gadgil will commit to structured didactic training and mentored research experience in the areas of 1) techniques for metabolomic analysis and interpretation; 2) training in network and pathway analysis; 3) clinical trial design and management; and 4) career development skills. This study is relevant and timely, as the incidence of both diabetes and its risk factors continues to rise, and personalized dietary strategies for prevention are few. The proposed investigations will contribute understanding, new knowledge, and potential targets of dietary intervention for diabetes prevention. Nicole Gilbert, Ph.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

Role of Gardnerella vaginalis in the Urinary Tract

ABSTRACT

The overall goal of this proposal is to provide me with the skills and expertise necessary to launch my career in translational in the field of urologic disease, particularly bacterial infections of the urinary tract. To achieve this goal, I will use the K01 mechanism to meet the following training objectives: Objective 1: Generate innovative data for publication in highly regarded peer-reviewed journals. Recurrent urinary tract infections (UTI) disproportionately affect women and are common, costly and sometimes life threatening. Escherichia coli is the most frequent causative agent of UTI, and has been shown in mouse models to establish antibiotic-resistant tissue reservoirs that persist long after acute infection of the bladder lumen is resolved. Bacterial emergence from such reservoirs has long been regarded as one likely origin of recurrent infections in humans. We have shown in a novel mouse model that Gardnerella vaginalis, a common member of the urogenital microbiota, causes E. coli to emerge from bladder reservoirs and initiate a recurrent UTI episode. The experiments outlined in this research plan test the hypothesis that G. vaginalis induces UTI recurrence by damaging the bladder epithelium (Aim 1) and modulating the host inflammatory response (Aim 2). These studies will increase our understanding of the mechanisms driving UTI recurrence and have the potential to identify G. vaginalis as a biomarker of risk in susceptible women. Objective 2: Gain expertise in performing translational research. I will engage in training activities, including seminars and didactic coursework offered by the NIH Clinical Center and through the Clinical Research Training Center (CRTC) of the Institute for Clinical and Translational Sciences (ICTS) at Washington University School of Medicine. This instruction will teach me the language and procedures of and execution and is essential for me to meet my goal of leading a transdisciplinary team doing T1 clinical research. With guidance from my co-mentors and the Research Design and Biostatistics Group of the ICTS, I will hone my skills in this area by performing a nested case-control study that seeks to confirm the findings of our mouse model in human clinical specimens. Objective 3: Develop my professional abilities and grow my professional network. Washington University has a wealth of career development mechanisms offered through the Center for the Integration of Research, Teaching, and Learning, the Offices of Postgraduate and Faculty Affairs and the CRTC. I will attend seminars and courses on research ethics, scientific manuscript and grant writing, academic job interviewing, laboratory finances and management. Additionally, I will attend instructive seminars at national meetings on urology and infectious diseases, where I will present my research findings and network and build collaborative relationships with leading researchers in the field. I have assembled a mentoring committee that includes leaders in the field of UTI research, which will evaluate the completion of both my scientific and career development milestones and facilitate my transition to an independent investigator. Ayush Giri, Ph.D.

VANDERBILT UNIVERSITY MEDICAL CENTER, NASHVILLE, TN

Influence of genetic variation, genetic ancestry, and obesity on gestational diabetes mellitus risk

PROJECT SUMMARY/ABSTRACT

Gestational diabetes mellitus (GDM), is among the most common pregnancy complications in the US. Shared pathophysiology with type-2 diabetes (T2D), evidence of familial aggregation, and evidence of racial disparity all support a role for genetic predisposition. Asian American (AAM) and Hispanic American (HA) women have lower prevalence of obesity on average than African American (AA) women, yet have higher GDM prevalence: 10.2%, 6.8%, 4.5% and 4.4% in AAM, HA, European American (EA) and AA women, respectively. Current studies are limited to candidate gene investigations with most investigating five to ten known T2D loci and only one genome-wide association study (GWAS) (468 cases; 1242 controls), in a South-Korean population. AAMs and HAs are the fastest growing populations in the US. Despite evident racial disparity suggesting a genetic etiology, no study has evaluated whether this is in part is rooted in differences associated with genetic ancestry. The overall goals of this proposal are to expand comprehensive genetic investigations of GDM and related traits by leveraging electronic health records (EHR) and bio-repositories to better understand the etiology which may inform personalized strategies for screening and prevention. We aim to develop, refine and validate reproducible and portable bioinformatics-algorithms to identity GDM cases and controls using de- identified EHR data at Vanderbilt. We will evaluate whether reported race/ethnicity modifies the association between maternal BMI and GDM in the Vanderbilt EHR database, the synthetic derivative (SD) (>8000 cases; Aim 1.1). In approximately 2,200 cases and 4,400 controls with genetic data, we will perform a Mendelian randomization study to test whether genetic instruments of central obesity (waist to hip ratio) or overall obesity (BMI) are more strongly associated with GDM (Aim 1.2). We will perform the first two-stage trans-ethnic GWAS of GDM in the US in EA, AA, HA, and AAM women from the SD and replicate associated variants (P < 1x10-6) in over 1000 GDM cases and many controls from the UK Biobank and Mount Sinai BioME EHR-linked bio- repository (Aim 2.1). By integrating GWAS data and expression quantitative trait loci (eQTL) data from various tissues with methods such as S-PrediXcan, we will prioritize candidate causal genes for GDM (Aim 2.2). Finally, we will explore whether genetically inferred Asian/Native American ancestry proportion is associated with increased risk of GDM (Aim 3.1) and T2D (Aim 3.2) in HAs. The well-tailored mentored training program supports the stated research aims and provides the candidate with the protected time to gain appropriate training in areas in which he lacks fully independent expertise, including phenotyping in the EHR setting, biomedical informatics and knowledge of gestational diabetes and classification of pregnancy outcomes. Successful completion of this award will facilitate the candidate's development into an independent multi- disciplinary researcher ideally prepared to contribute significantly to the fields of gestational diabetes, diabetes and associated complications, genetic epidemiology, racial disparity and women's health research. Eman Gohar, Ph.D., M.S.

UNIVERSITY OF ALABAMA AT BIRMINGHAM, BIRMINGHAM, AL

Sex Differences in Renal Sodium Handling

SUMMARY

This K99/R00 application from Eman Y. Gohar, PhD, is designed to acquire the knowledge and training necessary to transition into an independently-funded investigator leading a research program focused on sex differences in cardiovascular and renal physiology. Hypertension and kidney-related diseases are more common in men and postmenopausal women compared to premenopausal women. Renal purinergic signaling has emerged as an important system in the control of blood pressure and Na+ homeostasis. We have a recent evidence that estradiol (E2) stimulates Na+ excretion and increases the expression of renal purinergic receptors (P2Y2&4-R). Importantly, sex differences in the purinergic system has been demonstrated in non-renal tissues, however sex-related differences in renal purinergic signaling are not clear. The novel receptor, G protein-coupled estrogen receptor (GPER), is expressed in the renal medulla. GPER activation elicits cardiovascular and nephroprotective effects against salt-induced complications. However, its role in Na+ handling is not yet defined. Our preliminary data indicate that activation of GPER in the renal medulla promotes Na+ excretion via an endothelin-1 (ET-1)-dependent mechanism in female rats. In the current proposal, the overall hypothesis is that the enhanced renal Na+ handling in females is due, at least in part, to ATP/P2Y2&4-R/ phospholipase C/ epithelial Na+ channels (ENaC) and E2/GPER/ET-1/ENaC signaling within the renal medulla. This hypothesis will be tested by two specific aims. One aim will test whether female rats have enhanced P2Y2&4-R signaling in the renal medulla compared to males in response to high salt intake. The second aim will determine whether locally synthetized E2 activates GPER in the renal medulla to promote ET-1-dependent natriuresis in female rats via inhibition of ENaC. Under the mentorship of Dr. David Pollock and co-mentor, Dr. Edward Inscho, Dr. Gohar extends her current research on purinergic signaling and rapid estrogen signaling and proposes additional training that involves three major goals: i) technical development; Dr. Gohar will develop knowledge and technical expertise in the fields of calcium imaging, RNA sequencing and bioinformatics, beside refining her surgical skills, ii) professional career development in lab management, mentoring and grant writing, iii) training in translational research. This 5-year plan will prepare Dr. Gohar for an R01 submission and set her on a path for a successful independent career. Eric Gonzalez, Ph.D.

DUKE UNIVERSITY, DURHAM, NC

Underactive Bladder: Mechanisms and Recovery of Sensation and Function

PROJECT SUMMARY

Candidate (Eric J. Gonzalez, PhD): My long-term career goal is to develop a peripheral nerve stimulation device to manage lower urinary tract symptoms and improve bladder emptying in underactive bladder. I am seeking a Mentored Research Scientist Development Award (K01) to expand my translational research program and develop leadership skills to independently manage a research team. The K01 award will provide me with mentored training in human subjects research, didactic clinical coursework, and protected time for professional development. Environment: My career development will be guided by a multidisciplinary team with proven expertise in neural engineering, clinical research, basic science, and biostatistics. My training will be implemented at Duke University, a world-class academic medical center with innovative scientific research laboratories and neighboring medical facilities that enable rapid translation of novel ideas. Research: Underactive bladder is an understudied health concern that occurs in diverse patient populations and exhibits an age-related increase in prevalence. The palliative management options for persons with underactive bladder are associated with poor quality of life and patients often fail to completely resolve lower urinary tract symptoms. In order to improve therapeutic outcomes, there is a need to clarify the pathological mechanisms underlying underactive bladder. The objective of this proposal is to determine the role of motor and sensory nerve drive in the development of underactivity in a novel animal model, as well as in women with underactive bladder. We will first utilize a novel rat model of underactivity to determine the mechanisms of reduced motor drive and efficacy of pharmacological treatments and electrical neuromodulation in improving bladder emptying (Aim 1). We will also quantify the contributions of reduced sensory drive to the pathophysiology of lower urinary tract symptoms in neurologically-intact adult women with underactive bladder and determine the efficacy of electrical neuromodulation in resolving symptoms (Aim 2). These studies will increase our understanding of the muscular and neural alterations in the lower urinary tract that may accompany underactive bladder and will provide a foundation for clinicians to develop innovative therapeutics to improve patient outcomes. Russell Goodman, M.D.

MASSACHUSETTS GENERAL HOSPITAL, BOSTON, MA

A METABOLIC BIOMARKER OF HEPATIC NADH/NAD+ RATIO

PROJECT ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world, and is an increasing cause of significant morbidity and mortality. Despite its growing impact on global health there are currently no FDA-approved therapies for treatment. While the pathophysiology of NAFLD is complex, increasing evidence supports a direct role of alterations in hepatic NADH/NAD+ levels. However, testing the role of NADH/NAD+ in hepatic physiology is challenging given the limitations of existing tools with which to manipulate it in a precise manner, and the lack of circulating biomarkers for NADH/NAD+ which, as unstable intracellular metabolites, would be impractical to measure in most clinical and human research settings. The Mootha lab has developed a genetically encoded metabolic tool, called LbNOX, to directly and precisely manipulate cellular NADH/NAD+ in different tissues and cellular compartments. In preliminary work, I have used this tool both in vitro and in vivo to identify that a circulating metabolite, α-hydroxybutyrate (αHB) is sensitive to alterations in hepatic NADH/NAD+. αHB, has previously been shown to be a biomarker of insulin resistance and sensitive to polymorphisms in the gene GCKR, which in turn has been associated with NAFLD. Our preliminary work also shows LbNOX improves hepatic insulin resistance in vivo, and that GCKR influences hepatocyte NADH/NAD+. These findings link GCKR, NAFLD, αHB, and hepatic NADH/NAD+. The central hypothesis of this proposal is that αHB is a circulating biomarker of hepatic NADH/NAD+ which is causally linked to hepatic insulin resistance and steatosis, and that genetic modulators of hepatic NADH/NAD+ influence hepatic steatosis and insulin resistance In this proposal, I will use a combination of in vitro and in vivo hepatic LbNOX expression to further define the causal connection between hepatic insulin resistance, hepatic steatosis, and hepatic NADH/NAD+. I will define the mechanism by which LbNOX improves hepatic insulin resistance, and the mechanism by which GCKR influences hepatic NADH/NAD+. I am a clinical and research hepatologist dedicated to a research career as a physician scientist specializing in metabolic aspects of liver disease. The proposed research plan will allow me to develop new knowledge and expertise in metabolism, metabolomics, and hepatic physiology, and provide experience with animal models of chronic liver disease. Throughout the proposed research I will be guided by a formal research advisory committee comprised of outstanding mentors and experts in metabolism, NAFLD, and hepatic physiology, all in the setting of a stellar research environment comprised of MGH, the Broad Institute, and affiliated institutions. The research proposed, along with the guidance of my mentors and collaborators in this research, will help ensure my successful transition to scientific independence. Anjali Gopalan, M.S., M.D.

KAISER FOUNDATION RESEARCH INSTITUTE, OAKLAND, CA

The initial care of younger adults with newly diagnosed type 2 diabetes

PROJECT SUMMARY

Dr. Anjali Gopalan, MD, MSHP is a board-certified Internist at the Kaiser Permanente Northern California (KPNC) Oakland Medical Center and a Research Scientist I (equivalent to Assistant Professor) at the KPNC Division of Research (DOR). With the proposed K23 mentored career development award, Dr. Gopalan will gain the advanced training and expertise necessary to become an independent investigator who can develop and implement tailored interventions for patients with early-onset type 2 diabetes (EOT2D). By focusing on adults with EOT2D, she seeks to address the comparatively poor micro- and macrovascular outcomes in this population that may result partly from inadequate management of hyperglycemia and other cardiovascular disease risk factors. Dr. Gopalan’s research premise posits that it is particularly critical for newly diagnosed patients with EOT2D to achieve early disease control, since tighter glycemic control during the year following diagnosis confers long-lasting micro- and macrovascular benefits. In support of the premise, the Research Aims of this K23 are to: 1) use electronic health record [EHR] data to identify EOT2D patient subgroups at highest risk of inadequate early disease control, 2) identify barriers to and facilitators of initial EOT2D management, and 3) examine prospective associations between patient-reported factors (e.g., disease- related distress, health literacy) and inadequate early disease control. The proposed research will be supported by experienced mentors and advisors, as well as advanced training in the following areas: 1) the use of advanced statistical modeling, 2) qualitative data analysis to inform intervention development, 3) the role of social determinants in health, and 4) implementation science. KPNC and DOR are ideal environments for the proposed work. KPNC is an integrated learning healthcare delivery system with a diverse membership of 4.2 million people (>350,000 with diabetes). DOR has a long track record of effectively training early stage investigators and has well-established internal research programs and collaborative relationships with nearby institutions that provide the infrastructure needed for career development. This environment, along with the proposed Research and Training Plans and an outstanding mentorship team, will enable Dr. Gopalan to continue to build the skills and generate the foundational data needed to submit an R01-level proposal during the final years of the proposed award period. In this planned R01, Dr. Gopalan will refine and implement an intervention (with a design based on the results of the three K23 Research Aims) to improve early care for newly diagnosed EOT2D patients and evaluate its impact on early outcomes in this growing, high-risk population. In summary, the proposed K23 award will support the career development of Dr. Gopalan and is vital to her acquiring the skill set and expertise required to become a successful, independent investigator. Andrea Graham, Ph.D.

NORTHWESTERN UNIVERSITY AT CHICAGO, CHICAGO, IL

Testing Intervention Strategies for Addressing Obesity and Binge Eating

PROJECT SUMMARY

Obesity is a significant public health problem, with medical and psychosocial morbidities, impaired quality of life, increased mortality, and substantial health care costs. Up to 30% of treatment-seeking individuals with obesity have comorbid binge eating, an eating disorder behavior characterized by eating a large amount of food and experiencing loss of control. First-line interventions are face-to-face treatments for obesity or binge eating, but current approaches fail to address both conditions and cannot reach all people in need. To tackle comorbid obesity and binge eating, an intervention needs to be designed that: 1) targets both weight and binge eating; 2) improves the efficiency of the intervention while preserving its potency; and 3) is scalable to large populations of users. Thus, I propose a research agenda to design a mobile intervention that targets obesity and binge eating, which aligns with the NIH’s call for research that enhances the precision of intervention delivery. I will conduct two studies with adults with obesity (BMI ≥30) and recurrent binge eating (≥12 binge episodes in the past 3 months). First, I will employ a user-centered design approach to design a 16- session guided self-help, mobile intervention that combines components of psychological and behavioral weight loss (BWL) interventions. Overvaluation of weight and shape, unhealthy weight control practices, and negative affect are key treatment targets in psychological interventions for binge eating, but are not components of BWL. Thus, I will design an intervention that integrates these three components as adjuncts to BWL. I will conduct usability testing to inform intervention refinements prior to a pilot trial (Aim 1). Second, I will conduct a randomized pilot trial to determine the feasibility of delivering the intervention and to generate preliminary data on the effect of the intervention on changes in weight and binge eating at 16 weeks (post-intervention) and 3 months post-intervention (follow-up) (Aims 2 & 3). I will use a 23 full factorial trial design to randomly assign participants to a combination of the 3 components (n=8 conditions) in order to isolate which of the 3 intervention components are active ingredients. I will apply the Multiphase Optimization Strategy framework to optimize the intervention. These aims support my training plan in which I will receive expert mentorship and training in user-centered design and usability testing (Training Goal 1); using novel frameworks and efficient trial designs (Training Goal 2); and longitudinal data analysis of clinical trials (Training Goal 3). Expertise of the mentorship team, along with the academic and training environment of Northwestern University, will ensure my success. Results of this study will be used to optimize the intervention, which I will then test in a subsequent R01 trial. This K01 Mentored Research Scientist Development Award will equip me with the skills and data to facilitate my career goal of becoming a leader in increasing access to high-quality health care for obesity and weight-related issues, such as binge eating, using scalable interventions. Jason Greenberg, M.D.

YALE UNIVERSITY, NEW HAVEN, CT

Novel Biomarkers of Chronic Kidney Disease in Children

Project Summary. Candidate.

I am a pediatric nephrologist at Yale University dedicated to improving outcomes for children with chronic kidney disease (CKD). The goal of my application is to obtain mentored research training to develop a biomarker-augmented risk prediction model of pediatric CKD progression for use in future clinical trials. This research will build upon my prior training, which focused on the risk of CKD after acute kidney injury and the use of biomarkers to predict renal outcomes in children. This proposal will provide me with hands-on learning and formal didactic coursework in advanced statistics, biomarker methodology, and pediatric CKD. I will also intensely focus on developing the professional skills necessary for establishing effective collaborations, scientific writing, and obtaining funding to support my research. To accomplish my stated plan, I have the support of my highly qualified primary co- mentors (Drs. Chirag Parikh and Susan Furth) and mentoring committee (Drs. Haiqun Lin, Eugene Shapiro, and Prasad Devarajan) with interdisciplinary expertise in the fields of kidney injury biomarkers, translational research, biostatistics, and pediatric CKD. This multidisciplinary mentorship along with the highly skilled training environment at Dr. Parikh's, Program of Applied Translational Research will allow me to conduct my proposed research and establish an independently funded research program. Project. Progression of CKD in children leads to end stage renal disease (ESRD), which is associated with mortality rates 30-150 times higher than the general pediatric population. The traditional biomarkers, serum creatinine and proteinuria, are used to predict progression of CKD in clinical trials even though both correlate poorly with the progression of CKD and the response to interventions. There are numerous candidate therapies for CKD, but with a continued reliance on serum creatinine and proteinuria, clinical trials will likely continue to fail. The field of CKD biomarkers in children is a very promising area of research with a small amount of resources invested to date. Predicting progression of CKD will allow clinicians to better time follow-up, referral for transplant, and provide better guidance to families. More importantly, an optimal panel of biomarkers and risk prediction model can replace proteinuria and serum creatinine in biomarker guided clinical trials. We plan to measure urine and serum biomarkers of kidney injury, inflammation, repair, and fibrosis from the baseline samples of the 869 children with CKD enrolled in the CKD in Children (CKiD) cohort and determine their relationship with longitudinal measured GFR decline and incident ESRD. The optimal combination of biomarkers plus clinical variables from 2/3rd's of the CKiD patients will yield a risk prediction model to predict CKD progression. Our risk prediction model will be validated for longitudinal GFR decline, internally in 1/3rd of the CKiD patients, and externally in the 124 children of the Assessment, Serial Evaluation, and Subsequent Sequelae in AKI cohort. Developing a risk prediction model of CKD progression can be paradigm changing, transforming clinical care for children with CKD. Pinar Gumus Balikcioglu, M.H.S., M.D.

DUKE UNIVERSITY, DURHAM, NC

Serotonin as a Novel Biomarker for Progression to Type 2 Diabetes in Obese Adolescents

PROJECT SUMMARY:

Type 2 diabetes (T2D) is a worldwide pandemic affecting nearly 300 million adults and a rapidly increasing number of children, with profound individual and community health consequences. Estimated diabetes costs in the United States now exceed $200 billion. It is the major cause of blindness, leg amputation, and end-stage kidney disease and a principal cause of myocardial infarction and stroke. The two major determinants of T2D are obesity and insulin resistance (IR), yet the pathogenesis of T2D remains poorly understood. Only one-half of obese children are IR, and a far lower percentage progress to T2D. Why some obese children avoid metabolic dysfunction while others develop T2D is a critical gap in in the understanding of T2D pathogenesis and precludes the design and implementation of programs for diabetes prevention and treatment. My long-term career goal is to establish an independent research program dedicated to identifying and treating obese children at highest risk for IR and T2D. Using state-of-the-art metabolomic profiling of plasma samples, we previously found that childhood IR is associated with increased levels of the branch-chain amino acids (BCAA). Recently we have employed metabolic profiling of 24-hour urine samples, which has the advantage of integrating differences in metabolic status over time non-invasively. We identified a decrease in urine 5-Hydroxyindoleacetic acid (5- HIAA), the major metabolite of serotonin, as a novel biomarker associated with T2D in obese youth. This finding is particularly interesting given that serotonin increases pancreatic β-cell replication, β-cell mass, and glucose- stimulated insulin secretion. The reduction in 5HIAA in T2D likely reflects a decrease in cellular serotonin availability, since: (a) elevated BCAA compete with tryptophan, the precursor of serotonin, for uptake into β-cells and other tissues; and (b) in obesity, tryptophan metabolism is dysregulated and shifted towards production of kynurenine rather than serotonin. These findings suggest that reductions in tryptophan availability and serotonin production contribute to diabetes pathogenesis through inhibition of insulin secretion, promoting progression from obesity to glucose intolerance and T2D. We hypothesize that serotonin and serotonin metabolites including 5-HIAA constitute useful urine biomarkers that predict development of T2D in obese youth. This hypothesis will be tested in three Specific Aims. Aim 1 will identify urinary metabolic signatures that differ among non- obese and obese children with and without T2D. Aim 2 will determine if these urinary metabolic signatures correlate with parameters of glucose tolerance and glycemic control. Aim 3 will determine if urinary 5-HIAA measured at baseline or during follow-up predicts changes in disposition index, a composite measure of β-cell function in a longitudinal study of lean and obese subjects without T2D. My mentoring team, which includes Drs. Christopher Newgard (scientific mentor) and Michael Freemark (academic mentor), will provide guidance and quantitative and methodological expertise in clinical research. With an outstanding Duke University research environment, this award will facilitate my transition to independence as an academic pediatric endocrinologist. Autumn Harris, D.V.M.

UNIVERSITY OF FLORIDA, GAINESVILLE, FL

Sex Differences in Renal Ammonia Metabolism

Project Summary

Males and females differ in their basic physiology and pathophysiology in essentially all physiological systems. These biological sex differences can lead to differences in disease prevalence, progression rates and treatment outcomes. Understanding these differences can serve to identify new disease mechanisms and/or new and improved therapeutic opportunities. Thus, there is an immense need for additional research into the area of sex differences in both animal models and people. Acid-base homeostasis is of critical importance for maintaining normal health and renal ammonia metabolism has a major role in the maintenance of acid-base homeostasis. I have shown there are significant sex differences in basal ammonia excretion that result from sex differences in the expression of multiple important proteins involved in ammonia metabolism and transport. Furthermore, I have shown that there are significant structural differences between the female and male kidney. Hence, the overall objective of this proposal is to determine the underlying mechanisms of these sex differences in renal ammonia metabolism and to determine the role of these sex differences in physiological relevant disease models. This objective will be evaluated via the following specific aims: 1) determine the role of sex steroid hormones in the regulation of renal ammonia metabolism using a gonadectomy and hormone replacement model; 2) determine the role of sex steroid hormone receptors in the development of sex-dependent differences in ammonia metabolism through the use of kidney-specific AR and ERα deletion; 3) determine the role of sex in the regulation of renal ammonia metabolism using the four core genotype model; and, 4) determine the effect of sex on the renal response to conditions of altered ammonia metabolism such as metabolic acidosis. Autumn Harris, DVM, DACVIM (SAIM), is an NIH T32-funded post-doctoral research fellow at the University of Florida College of Medicine. This career development award will provide Dr. Harris with the additional training needed to achieve her goal of becoming an independent investigator, which includes training in the areas of 1) study design and implementation, 2) performance, interpretation, and evaluation of various laboratory techniques and diagnostic tests, 3) statistical methods and interpretation, 4) collaboration with a multidisciplinary and multi-institutional team, 5) grant and manuscript writing. Dr. Harris has developed a comprehensive program including didactic training, practical and laboratory experience, and a strong mentoring and advisory team. The University of Florida College of Medicine will provide a collegial and supportive environment with the equipment, laboratory space, and resources necessary to complete the research and training. At the culmination of this proposal, Dr. Harris will be poised to achieve independence as a researcher with continuing investigation in the field of nephrology and sex differences.

Alexander Hawkins, M.P.H., M.D.

VANDERBILT UNIVERSITY MEDICAL CENTER, NASHVILLE, TN

Novel Assessment of the Role of Surgery in Recurrent Diverticulitis

PROJECT SUMMARY/ABSTRACT

Diverticulosis of the sigmoid colon is a significant burden to the US health system. Complications lead to substantial morbidity and mortality. In 2004, diverticular disease was responsible for 313,000 hospitalizations, 1.9 million ambulatory care visits, and 3365 deaths in the United States. Indications for surgical management of chronic or recurrent diverticulitis are poorly understood. A number of studies have revealed that there is minimal mortality benefit to elective surgery for diverticular disease. Because of this, traditional surgical outcomes, such as 30-day survival, wound morbidity and in hospital complications are poor metrics to analyze this problem. This proposal describes a career development plan for a colorectal surgeon that will enable me to use my 75% protected effort to develop expertise in qualitative analysis, patient reported outcomes, comparative effectiveness, and analysis of longitudinal data. The research specific aims are: 1) to investigate alignment between the goals and expectations of patients and surgeons; 2) analyze the effect of surgery on the function and quality of life of patients with recurrent diverticulitis; and 3) assess the effect of surgery on health care utilization, work absenteeism and financial toxicity in recurrent diverticulitis. The career development plan integrates: a) advanced coursework and individualized mentor-directed instruction in qualitative methodology, patient reported outcomes, and surgical comparative effectiveness techniques, b) participation in local/national seminars and conferences to advance expertise in qualitative and quantitative aspects in development of patient reported outcomes, c) a multidisciplinary mentored research experience, and d) a highly supportive research environment. This environment includes the Center for Health Services Research, the Center for Surgical Quality and Outcomes Research, an AHRQ-funded Evidence- based Practice Center, a top-ranked Peabody School of Education, a Masters in Public Health program, and the Vanderbilt University Department of Surgery. Overall, this career development award will advance my ability in qualitative methods, patient reported outcomes and comparative effectiveness research by providing both didactic and experiential education under the supervision of proven mentors. We will generate important data on functional and quality of life outcomes for patients with diverticular disease. Importantly, this award provides critical support to seamlessly transition toward future independently funded studies that use patient reported outcomes to conduct comparative effectiveness studies to better understand the role of surgery in chronic disease states, including both diverticular disease and other disorders. Tim Heden, Ph.D.

UNIVERSITY OF MINNESOTA, MINNEAPOLIS, MN

Glycophagy in liver and skeletal muscle insulin sensitivity and energy metabolism

Project Summary/Abstract

Liver and skeletal muscle insulin resistance represent two core defects in individuals with Type 2 Diabetes. Although dysregulated glycogen metabolism is linked to insulin sensitivity, it is not entirely understood how glycogen metabolism modifies insulin sensitivity. New preliminary data generated by the applicant suggests that the autophagic degradation of glycogen (i.e. glycophagy) by the lysosomal enzyme alpha acid glucosidase (GAA) plays a previously unrecognized role in modulating liver and skeletal insulin sensitivity and energy metabolism. In mice, high-fat diet feeding reduced insulin sensitivity, an effect associated with an increase in liver GAA levels, whereas exercise or muscle contraction increased insulin sensitivity, an effect associated with reduced muscle GAA levels. Follow-up mechanistic studies in liver or skeletal muscle cells showed that acute inhibition of glycophagy reduced glycolytic capacity while increasing insulin sensitivity, mitochondrial biogenesis, and fatty acid oxidation, effects that were associated with an induction of SIRT1 signaling and reduced oxidative stress. To better define the link between glycophagy, insulin sensitivity, and energy metabolism in mice, the applicant used CRISPR/Cas 9 technology to generate a novel mouse model with inducible liver or skeletal muscle specific knockout of GAA. The overarching objective of this K01 proposal is to define the role of glycophagy in liver and skeletal muscle insulin sensitivity and energy metabolism in mice. Additionally, the applicant has assembled an interdisciplinary grant advisory panel to provide mentorship and guidance for the proposed research. The training plan was designed to build upon the applicant’s prior expertise in metabolism by providing additional training in analytical techniques, research concepts, data analysis, grant and manuscript writing, didactic training in tracer methodology, establishing productive collaborations, and leadership skills. This proposal will help the applicant initiate his own investigative niche in the field of glycophagy and serve as a springboard to his independent research career. LaTonya Hickson, M.D.

MAYO CLINIC JACKSONVILLE, JACKSONVILLE, FL

Mesenchymal stem cell senescence in diabetic nephropathy

PROJECT SUMMARY/ABSTRACT

I am a practicing nephrologist at Mayo Clinic and I will use the acquired skills from my clinical and epidemiology background to bring the proposed basic research investigations into truly translational research for patients with diabetic nephropathy (DN). I have assembled a multidisciplinary team of mentors who will provide guidance, infrastructure, and tools needed for performance of the proposed studies. My immediate career goal is to gain laboratory research skills, clinical trial experience, regenerative medicine fund-of- knowledge, and regenerative medicine coursework. My long-term goal is to become a leader in Regenerative Nephrology and an independent, productive clinician-investigator. My career development plan will provide me with the building blocks needed to transition into independence. I have the strongest support from my division, department, and institution. In the proposed studies, I will explore the feasibility of a novel therapeutic platform that I believe may change the course of disease and improve the lives of patients with DN, a devastating disease with few therapeutic options. Mesenchymal stromal/stem cell (MSC) transplantation offers hope. MSCs are non- embryonic stem cells with anti-fibrotic, anti-inflammatory, and pro-angiogenic paracrine activity that improve regeneration in DN models. Senescence is an irreversible cell cycle arrest, which generates a pro-inflammatory secretory phenotype that impairs neighboring cell function. Hence, increased senescent cell burden in DN may substantially compromise MSC function and become a barrier to successful autologous MSC transplantation. Our overall goal is to characterize and optimize the functional properties of MSC in DN to allow these patients to benefit from future enrollment in clinical trials using stem cell transplantation. A central mechanism limiting MSC functional capacity, may be treatable through senolytic drugs that selectively eliminate senescent cells. We will examine senolytic therapy as a potential in vivo preconditioning method to improve stem cell function. Our central hypothesis is that adipose-derived MSC obtained from DN patients show increased senescence and decreased functionality, which can be ameliorated, both in vitro and in vivo, using drugs that clear senescent cells. First, we will compare cellular senescence and functionality in MSC from DN patients to MSC from age- and gender-matched controls. Second, we will incubate cells with senolytic agents in vitro and assess DN-MSC senescent cell clearance and function thereafter. Third, we will conduct a pilot study wherein DN patients will receive senolytic drugs, and MSC senescence and function will be measured at baseline and 14 days after treatment. Additional examinations will include blood and urine collection for kidney function and injury measurements. The proposed studies explore an innovative approach for preconditioning MSC and their deleterious microenvironment, and aid in developing a completely novel therapeutic strategy to delay DN progression. David Hill, Ph.D.

CHILDREN'S HOSP OF PHILADELPHIA, PHILADELPHIA, PA

The role of unique adipose tissue macrophage populations in obesity

PROJECT SUMMARY/ABSTRACT

This proposed five-year training program will develop my independent research career as an academic pediatric allergist/immunologist seeking to better understand how obesity and other metabolic derangements influence atopic disease states such as asthma and food allergy. I have completed an MD and PhD in innate immunology, pediatrics residency training, and am entering my final year of fellowship training in allergy and immunology at the Children’s Hospital of Philadelphia (CHOP). My immediate goal is to refine the essential skills necessary for a successful career as an independent investigator. Specifically, I want to be immersed in the field of metabolism, and gain new expertise with advanced and unbiased transcriptomic techniques to complement my prior training in experimental and clinical immunology. My mentor for this award is Dr. Mitchell Lazar, an eminent physician- scientist and expert in the fields of metabolism and epigenetics. To add depth and breadth to my scientific and career guidance, I am supported by an advisory committee composed of scientists and physician-scientists from relevant and complementary fields. I have secured the complete support of my institution, and will benefit greatly from the unparalleled resources and mentorship available at both CHOP and the University of Pennsylvania over the course of this award. My proposal focuses on how adipose tissue macrophages (ATMs) influence adiposity and obesity-related outcomes. ATMs can protect against or promote obesity depending on context, discordant observations that raise fundamental questions as to the number and functions of distinct ATM populations. Drawing on the expertise of the Lazar lab, I utilized single-cell mRNA sequencing and flow cytometry to identify two, transcriptionally-distinct ATM populations in obese mice and humans. analysis indicated that these ATM subsets are equipped to have distinct functions: one that is proinflammatory and one that is tissue- regulatory. These ATM subsets are differentially regulated in an inbred mouse strain that is genetically-resistant to obesity and an established model of genetic variation in humans, implicating ATM subsets in mediating obesity susceptibility. Together, these findings have led me to develop the hypothesis that ATMs exist in more than one functional state, and that differences in the relative proportion of functionally distinct ATMs influence the adipose tissue environment and susceptibility to obesity. This proposal outlines a series of studies that have the potential to fundamentally reshape our view of ATM biology, and have implications for the development of personalized medical approaches for obesity. Importantly, the skills that I gain through the proposed training program will facilitate my long-term goal of using basic and translational research approaches to understand how obesity and other metabolic derangements influence atopic disease states, such as asthma and food allergy. John Hollier, M.S., M.D.

BAYLOR COLLEGE OF MEDICINE, HOUSTON, TX

A New Paradigm for Treating Functional Abdominal Pain Disorders in Children

Project Abstract

John M. Hollier, MD, a board-certified pediatric gastroenterologist, is an Assistant Professor at Baylor College of Medicine who seeks to obtain essential skills and mentored research experience for an independent career as a pediatric clinical researcher utilizing health technologies. Many aspects of medicine are affected by poor medication adherence, ill-advised health behaviors, and poor access to medical care. The candidate’s career goal is to conquer these barriers by harnessing the potential of current (e.g., mobile) and future technologies as tools to fulfill these endeavors. Functional abdominal pain disorders (FAPDs) affect up to 20% of school-age children (and adults) worldwide and are associated with significant morbidity and health care costs. Up to 60% of children with FAPDs go on to have similar conditions as adults. Many affected children suffer from psychosocial distress like anxiety and depression. The most effective treatment for these disorders (psychological interventions) remains out of reach for most children with FAPDs. The applicant proposes to refine his innovative mobile application (App) prototype designed to remotely deliver audio- recorded guided imagery therapy (ARGIT App) for pediatric patients with FAPDs using accepted technology development techniques. Specific Aims: 1) Refine our ARGIT App prototype’s design, operation, and GIT session content via patient, caregiver, and expert stakeholders’ feedback by using qualitative interviews 2) Conduct a double-blind, randomized controlled trial comparing GIT versus deep breathing exercises (control) utilizing the refined ARGIT App over 8 weeks in children with FAPDs to assess the efficacy of guided imagery therapy and evaluate whether psychosocial distress is altered as a result of this intervention over time. 3) Assess acceptability of the refined ARGIT App among affected children and their caregivers through qualitative interviews. Dr. Hollier has assembled a multidisciplinary team of esteemed NIH-funded mentors and collaborators with a diverse range of expertise in mobile health App development, clinical trial methodology, qualitative research methods, and health behavior change specialists. In addition, the candidate will acquire additional skills and training through didactic coursework, workshops, directed readings, and conferences. The scientific training obtained through this grant will lead to publications, data, and experience that will enable the candidate to secure NIH funding and establish himself as an independent physician scientist. Katharina Hopp, Ph.D.

UNIVERSITY OF COLORADO DENVER, Aurora, CO

Defining the functional role of T-cells in Autosomal Dominant Polycystic Kidney Disease pathology

PROJECT SUMMARY/ABSTRACT

This NIDDK K01 application is designed to provide Dr. Katharina Hopp with the scientific, technical, and career training to enable her transition into an independent investigator studying mechanisms controlling Autosomal Dominant Polycystic Kidney Disease (ADPKD) cystogenesis. The project builds on Dr. Hopp's expertise in the genetics and cell biology of ADPKD, and extends her training in the immunological and microenvironmental aspects of this disease. The proposal encompasses a five-year training plan under the primary mentorship of Dr. Raphael Nemenoff, an expert in the cancer microenvironment, and an Advisory Committee comprised of accomplished immunologists and PKD physician/scientists. The project will investigate the functional role of T- cells in cyst initiation and progression, underlying mechanisms, and novel therapeutic approaches. Features of ADPKD parallel those of cancer, including induction of proliferation, genomic instability, and increased inflammation. In cancer, targeting T-cells in the tumor microenvironment has shown clinical success; however, the functional role of T-cells in PKD is poorly understood. Preliminary data generated by Dr. Hopp showed that, in a well-established murine ADPKD mouse model developed by Dr. Hopp, distinct T-cell subpopulations increased correlative to disease severity and localized specifically to cystic lesions. Importantly, depletion of CD8+ T-cell, which are generally anti-tumorgenic, increased disease progression, highlighting the functional importance of these cells in halting cyst progression. However, regulatory T-cells, which are generally pro- tumorgenic, rose early in disease, suggesting that distinct T- cell subpopulations may have opposing effects on cystogenesis. In addition, both PD-L1 and PD-1, components of an immune checkpoint pathway, were significantly increased in the mouse model and PKD patient kidney sections. Targeting this pathway has been therapeutically effective in numerous cancers. Thus the central hypothesis of this project is that interactions of distinct T-cell populations with the cystic microenvironment/epithelium result in both anti- and pro- cystogenic effects, and targeting T- cells represents a novel therapeutic strategy for APDKD. The specific aims of this project are (1) Define the functional role of T-cell subpopulations in cyst initiation and progression; (2) Elucidate the mechanisms how T-cells alter cellular pathways in the cystic epithelium; and (3) Evaluate the efficacy of checkpoint inhibitors in ADPKD. The project will be supported by the University of Colorado, Denver's outstanding PKD Program, the Renal Division, and the Immunology Department. The PKD Program has been successful in translating preclinical data into clinical trials and is motivated to incorporate results of this project into future clinical trial designs. In addition to the above aims, Dr. Hopp will (1) develop a strong knowledge of immunology and related novel technique/model systems; (2) expand her professional proficiencies in manuscript/grant writing, mentoring, and reviewing duties; and (3) submit a competitive R01 application towards the end of this K01 expanding upon findings from this application. Joanne Hsieh, Ph.D.

COLUMBIA UNIVERSITY HEALTH SCIENCES, NEW YORK, NY

Hepatoprotective Mechanisms of TTC39B Deficiency

Hepatoprotective Mechanisms of TTC39B Deficiency PI: Hsieh, Joanne

Project Summary/Abstract

This K01 application is designed to provide the necessary resources and training to transition Dr. Joanne Hsieh to full independence in the field of lipid metabolism in liver disease. The proposed work and training will be performed at Columbia University Medical Center (CUMC) in the Department of Medicine, which has demonstrated a commitment to junior faculty mentorship. She will be mentored by Dr. Alan Tall, who is a pre-eminent investigator in lipoprotein metabolism and atherosclerosis. With Dr. Joel Lavine in the Department of Pediatrics as her co-mentor, Dr. Hsieh will transition her focus towards non-alcoholic fatty liver disease (NAFLD). Dr. Lavine has an extensive record of clinical research in adult and pediatric NAFLD, and will be integral in Dr. Hsieh's training in building translational aims into her biomechanistically-focused research. Dr. Hsieh will continue her research on TTC39B, a novel gene significantly associated with HDL- in human GWAS. Dr. Hsieh recently showed that deficiency in TTC39B conferred a dramatic protection from steatohepatitis in mice. This hepatoprotection was associated with inhibited sterol-regulatory element binding protein-1 (SREBP-1) activation and decreased lipogenic gene expression. The overall goal of the proposal to determine how decreased expression of specific lipogenic genes contributes the hepatoprotective effects of TTC39B deficiency. Dr. Hsieh will conduct the investigations in a diet-induced mouse model of steatohepatitis that exhibits many of the histological features observed in human NASH, including inflammatory cell infiltration and fibrosis. In the first aim, TTC39B's effect on SREBP-1 activation will be further explored mechanistically to understand the early pathogenesis of NAFLD. The first subaim will determine whether the SREBP- 1 inactivation in TTC39B deficiency is driving the protection from steatohepatitis, while the second will investigate the role of phospholipid metabolism in this inactivation. The second aim will explore whether the suppression of phosphatidic acid signalling in TTC39B deficiency prevents the progression of NAFLD to the more severe non-alcoholic steatohepatitis (NASH). Dr. Hsieh's scientific research and career development will be further supported by members of her advisory board, including Dr. Muredach Reilly, Dr. Robert Schwabe, and Dr. Richard Deckelbaum. The mentoring and advisory team will help broaden Dr. Hsieh's approach to scientific hypothesis-testing and ensure her success as an independent investigator. Jing Hughes, Ph.D., M.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

GLUCAGON REGULATION BY A NOVEL BROWN ADIPOSE FACTOR

Project Summary/Abstract

This proposal aims to develop the applicant into an independent physician-scientist in the field of diabetes and metabolism. The principal investigator Dr. Jing Hughes has undergone PhD training in the basic biological sciences and completed clinical training in internal medicine and the endocrinology subspecialty. The proposed 5-year career development plan will build on Dr. Hughes's experiences in endocrinology and provide her with further expertise in the design and conduct of basic diabetes research. Dr. Hughes's long-term goal is to establish an independent laboratory dedicated to studying islet biology, with a goal to discover new endocrine and paracrine pathways modulating islet hormone secretion. The central guidance and training environment for this project will be provided by the mentor, Dr. David W. Piston, who is the Chair of Cell Biology at Washington University, also a recognized leader in islet physiology and imaging expert. Dr. Piston has an excellent track record for training physician-scientists, especially at the incipient stages of their careers. In addition to Dr. Piston's mentoring, Dr. Hughes will be able to take advantage of the rich resources available at Washington University, including scientific as well as career guidance from her faculty advisory committee and collaborators. The research set forth herein seeks to address how brown fat restores glucose balance by modulating glucagon secretion in diabetic animals. In the past few years, the Piston laboratory has revolutionized thinking about islet physiology by demonstrating new and unexpected roles in diabetes modulation by alpha cell hormones. We now examine the mechanism by which factors secreted by brown adipose tissue act on alpha cells to regulate glucagon production, a phenomenon that has never before been investigated. Dr. Hughes will use a proteomics strategy to identify the brown adipose factor and test the hypothesis that its effect on glucagon suppression is mediated through GPCRs on islet cells. The proposed studies have the potential to provide proof-of-principle for novel translational treatments and reduce suffering from diabetes. This K08 mentored research award will provide the necessary resources for Dr. Hughes to complete these studies and to develop the necessary intellectual foundation and skill set as an independent diabetes investigator. Anne Huml, M.D.

CASE WESTERN RESERVE UNIVERSITY, CLEVELAND, OH

Barriers to Care for Patients Transitioning from Transplant to Dialysis

DESCRIPTION (provided by applicant):

End Stage Renal Disease following failure of a previously transplanted kidney (Tx-ESRD) is the fourth leading cause of ESRD. However, its contribution to the burden of ESRD has been underappreciated because national registries do not report incident and prevalent cases of Tx-ESRD as a separate category. Several unique features of Tx-ESRD may adversely affect quality of care. Pre-dialysis planning such as dialysis modality education or vascular access placement may be delayed by efforts to prolong transplant function and uncertainty about risk for transplant failure. In addition, Tx-ESRD patients are exposed to immunosuppressive medications, are subject to psychosocial stress from the loss of a transplant, and are required to transition care from transplant to dialysis providers. However, the specific barriers that Tx-ESRD patients face are poorly understood and have not been studied in a systematic way. Moreover, little is known about feasible methods to improve Tx-ESRD quality of care. We propose a mixed methods research design to address three specific aims: A) to determine the quality of care that patients with Tx-ESRD receive during the first year of dialysis; B) To determine barriers to quality of care among patients who are transitioning from transplantation to dialysis; and C) to develop potential interventions to improve the quality of care of patients transitioning from transplantation to dialysis. Aim A will involve a retrospective of 3000 patients from Ohio, Kentucky, Indiana, and Illinois. Aim B will involve a of 70 greater Cleveland patients with failing kidney transplants and their providers. Aim C will present Aim A and B findings and a on transitions of care to a national panel of 95 transplant nephrologists who will use a Delphi process to develop and rank potential interventions. This innovative approach will allow us to incorporate patient, provider, and expert perspectives. As part of the proposed study, the principal investigator will also receive training in advanced statistical techniques, qualitative research methods, and intervention trials in order to become an established clinical researcher in transitions of care along the continuum of kidney disease. The results of these analyses will be an empirically informed model of the barriers to quality of care for patients with Tx-ESRD who are transitioning to dialysis as well as feasible potential interventions to improve quality of care. The proposed study will set the stage for future intervention trials designed to optimize care and outcomes for this patient population. Kristin Javaras, Ph.D.

MCLEAN HOSPITAL, BELMONT, MA

Understanding the Effects of Acute Stressors and Negative Emotion on Eating Behavior in Binge-Eating Disorder: The Role of Stressor-Induced Changes in Reward and Cognitive Control

PROJECT SUMMARY/ABSTRACT

Binge-eating disorder (BED) is characterized by recurrent episodes of binge eating (uncontrolled consumption of large amounts of food in a short time period). BED is the most prevalent eating disorder in the U.S. and globally, and it is associated with significant psychosocial impairment, and psychiatric and physical morbidity. However, even with the most efficacious treatments, a substantial proportion of those with BED do not experience good outcomes. Elucidating the biobehavioral mechanisms of binge eating is critical to gaining better understanding of treatments’ effects and why they vary. In this vein, converging evidence suggests that negative affect plays a significant role in binge eating in BED, but the link between negative affect and binge eating behavior, including its neurobiological mechanisms, is not well understood. In contrast, a growing body of literature suggests that stressors tend to increase preference for highly palatable food in the general population, and that this phenomenon may involve mechanisms of increased food reward and decreased cognitive control. Thus, the goal of the K23 research project is to examine whether stressors have greater effects on (palatable) food choice and related blood oxygenation level dependent activity in young women with BED, and whether these stressor-related effects relate to real-world binge eating behavior. Dovetailing with the research aims, the K23 will provide training and mentored research experience in the following areas critical to achieving the candidate’s long-term career goal of becoming an independent clinical investigator focused on understanding the mechanisms underlying binge eating, with the ultimate goal of developing more tailored and efficacious interventions: (1) neuroscience and neuroimaging; (2) the study of stressors and their effects; (3) assessment of real-world stressors, affect, and eating; (4) responsible conduct of research; and (5) general career development. To achieve these training goals, the candidate will attend relevant classes, seminars, and scientific conferences and receive mentoring and training from an expert team including Dr. Diego Pizzagalli (Primary Mentor), Drs. James Hudson and Shelly Greenfield (Co-Mentors), and Drs. Lisa Nickerson, Joanna Steinglass, Marian Tanofsky-Kraff, & Stephen Wonderlich (Advisors). The research and training will occur at McLean Hospital, a psychiatric teaching hospital with a state-of-the-art imaging center and eating disorder research program. McLean, an affiliate of Harvard Medical School, provides an ideal environment due to its clear institutional commitment to research and academic/professional training and its unique opportunities for diverse clinical and basic researchers to collaborate to better understand the brain in illness and health. In addition to providing the candidate essential training in designing and implementing patient-oriented research with sophisticated behavioral and neurobiological measures, the K23 award will provide novel data on the role of stressors and negative affect in BED. This line of research can yield critical information relevant to developing more efficacious interventions for BED, a compelling target for improving public health. Angie Jelin, M.D.

JOHNS HOPKINS UNIVERSITY, BALTIMORE, MD

Single gene pathogenic variants associated with BEEC (Bladder Exstrophy, Epispadias, Complex)

PROJECT SUMMARY

This K23 application is submitted by Angie C. Jelin, MD, Assistant Professor of Gynecology and Obstetrics/Genetics at the Johns Hopkins School of Medicine. Dr. Jelin’s long term goal is to become an independent investigator in fetal urinary tract anomalies. Towards this goal, she proposes a mentored career development plan that provides training in whole genome sequencing (WGS), genomics, CRISPR/Cas9, and mouse phenotyping. Urogenital anomalies account for 20-30% of prenatally detected structural defects. Bladder Exstrophy Epispadias Complex (BEEC) describes a subset of anomalies with a spectrum of developmental defects ranging from a mild form of epispadias, to classic bladder exstrophy, to omphalocele, exstrophy, imperforate anus, spinal anomalies (OEIS) complex. Patients with BEEC suffer substantial morbidity and mortality due to impaired genito-urinary dysfunction. The etiology of BEEC is largely unknown. Elucidating the underlying genetic component is critical to gaining a better understanding of the developmental signaling pathways and is likely the first step to developing targeted therapy. Variants in genes identified in other urogenital anomalies appear to be responsible for some cases of BEEC including IS, WNT3, WNT9b, PLAG1 and p63. We propose to take advantage of our extensive analytical experience in the Baylor Hopkins Center for Mendelian Genomics and perform WGS on parent-proband trios for whom the proband has BEEC. One study utilizing whole exome sequencing (WES), identified candidate genes (SLC20A1 and CELSR3) in 2 out of 8 affected patients, providing reassurance that our proposed strategy will be successful. Following WGS, we will explore the pathogenicity of genetic variants by employing a knockout mouse model using CRISPR/Cas9 technology via collaboration with the Jackson Laboratory. Final validation will include mouse phenotyping by dynamic contrast-enhanced MRI under the expertise of, Cory Brayton, mouse pathologist. Aim 1. To identify the genetic basis of BEEC through Whole Genome Sequencing (WGS) Aim 2a. To create the founder (F0) homozygous knockout mouse using CRISPR/Cas9. Aim 2b. To define the murine model phenotype using dynamic contrast enhanced MRI. Yuwei Jiang, Ph.D.

UNIVERSITY OF ILLINOIS AT CHICAGO, Chicago, IL

Molecular regulation of adipose tissue development, homeostasis and expansion

Project Summary

Childhood and adult obesity and their associated metabolic complications is a public health problem. In the United States alone, more than 67% of adults and 33% of children are currently overweight or obese, which can cause both immediate and long-term health problems. If this epidemic is uncorrected it will lead to a range of ensuing metabolic disorders such as heart disease, diabetes, atherosclerosis, and cancer. Adipose or fat tissues appear to be at the nexus of the obesity crisis. However, very little is known about the cells and molecular mechanisms that oversee the formation and the expansion of adipocytes and adipose tissues. Our recent findings using genetic mouse models demonstrate that there are two progenitor populations that give rise to adipocytes: developmental progenitors, for adipose development, and adult progenitors, for adipose homeostasis. In established adult adipose depots, not developmental adipose depots, adipocytes fate-map from a perivascular residing progenitor cell pool. Contrary to adult adipose tissue, even less is known about the cellular origin of developmental progenitor cells and the signals that control their cellular dynamics including pattern, proliferation, migration and formation. The central focus of this proposal aims to use our genetic tools to identify novel regulatory mechanisms important for adipose tissue development compared to adult adipose tissue homeostasis and expansion. Previous studies have suggested that platelet derived growth factor receptor α (PDGFRα) is an important potential source of developmental adipose progenitor cells. Furthermore, PDGFRα+ cells contribute to adipose tissue expansion in respond to high fat diet feeding. However, few studies have examined if PDGFRα, gene function, is critical for adipose tissue development or adult regulation. Based on our preliminary data, I hypothesize that PDGFRα signaling is a central regulator of adipose tissue development by establishing the adipose lineage. Thus the specific aims of this study are focused on: Aim 1: Determine if PDGFRα is necessary for adipose tissue development (organogenesis). Aim 2: Determine the mechanisms by which PDGFRα controls adipose tissue development. Aim 3: Determine if PDGFRα is necessary for adult adipose tissue obesogenic expansion. Based on preliminary data, PDGFRα is critical for the development of adipose tissue but not for maintaining it. It appears that PDGFRα regulates adipose progenitor cell fate promoting adipose tissue organogenesis. These findings will be of great importance for three reasons: 1) how embryonic cells adopt an adipose tissue cell fate, 2) the molecular underpinning that govern this developmental cell fate, and 3) how adipose tissue expands in response to caloric excess compared to adult adipose tissue homeostasis. These important findings may lead to novel discrete therapeutic targets for childhood and adult obesity. Joshua Joseph, M.D.

OHIO STATE UNIVERSITY, COLUMBUS, OH

The Role of The Renin-Angiotensin-Aldosterone System, ARMC5, and Neprilysin in Glucose Metabolism among African Americans

PROJECT SUMMARY/ABSTRACT

Joshua J. Joseph, MD, is an Assistant Professor in the Division of Endocrinology, Diabetes and Metabolism at The Ohio State University. Dr. Joseph seeks a Mentored Patient- Oriented Research Career Development Award in order to obtain the skills, knowledge, and mentored research experience that are essential for a career as a clinician scientist in the field of type 2 diabetes mellitus (T2DM) prevention. This proposal is aimed at determining the role of the renin-angiotensin-aldosterone system (RAAS) in glucose metabolism and the development of T2DM among African Americans (AAs). AAs are 1.7 times as likely to develop T2DM in the US and are twice as likely to die from T2DM compared to non-Hispanic whites. Thus, this represents an area of critical need. The objectives of this proposal are to determine the role of the RAAS, endothelin-1, ARMC5 (armadillo repeat containing 5) and RAAS antagonism in glucose metabolism and the development of diabetes. The specific aims of this research proposal are: (1) to determine the associations of aldosterone and endothelin-1, individually and combined, with HOMA-insulin resistance, HOMA-β cell function, fasting plasma glucose and incident T2DM among AAs without T2DM at baseline in the Jackson Heart Study (JHS); (2) to determine a) the cross-sectional associations of predicted damaging ARMC5 mutations with plasma aldosterone, plasma renin activity, fasting glucose, and prevalent T2DM and b) the longitudinal association with incident T2DM among AAs in the JHS; (3) to determine the impact of RAAS antagonism or RAAS and neprilysin antagonism vs. placebo with changes in glucose metabolism over 6 months assessed via glucose clamp studies among AAs. For Aim 1, we propose predictive epidemiological analyses in the JHS, an observational investigation of cardiovascular disease among AAs, to determine the association of a combination of aldosterone and endothelin-1 with glucose metabolism, prevalent and incident T2DM. For Aim 2, we propose genetic analyses in the JHS, to determine the association of ARMC5 genetic variants with components of glucose metabolism, prevalent and incident T2DM. For Aim 3, we propose a 26- week clinical trial to test the effect of RAAS antagonism on β-cell function and insulin resistance in AAs with impaired glucose tolerance. The goals during the award period include developing expertise in the design, performance, analysis and presentation of clinical research through mentored research, didactic coursework, and formal training in clinical investigation of glucose metabolism, clinical trial methodology, genetic, genomic and other –omic analytic techniques and predictive/causal modeling. Long-term career goals include developing a career as an independent investigator focused on finding new approaches for preventing and treating T2DM, particularly among historically understudied populations in biomedical research. The proposed research aims to provide new insights into the contribution of the RAAS to changes in glucose metabolism in the development of T2DM. This work will lay the foundation to develop novel therapeutic targets for T2DM.

Evangelia Kalaitzoglou, M.D.

UNIVERSITY OF KENTUCKY, LEXINGTON, KY

Muscle –bone interaction and its role in diabetic bone disease of Type I diabetes

Summary

Bone fracture incidence observed in individuals with Type 1 diabetes (T1D) is much higher compared to the general population. The burden of diabetic bone disease is partially due to lack of evidence to support targeted prevention and interventions to reduce fractures in this population. Furthermore, those with T1D exhibit skeletal muscle dysfunction associated with decreased muscle strength and muscle mass. Skeletal muscle and bone communication is a potential modifiable factor that may contribute to development of diabetic musculoskeletal disease. More specifically, the candidate proposes that a myokine called myostatin, is directly involved in development of diabetic bone disease and may associate with skeletal muscle dysfunction. The role of myostatin in T1D and how it affects the musculoskeletal system in this disease are gaps in knowledge that will be addressed with this proposal. Specifically, the three aims of this proposal are: 1. Quantify the relationship between myostatin levels in serum and skeletal muscle, and bone parameters of humans with and without T1D; 2. Quantify the relationship between myostatin levels in serum and muscle and the bone phenotype of mice with insulin-deficient diabetes; 3. Evaluate whether inhibition of myostatin is beneficial for prevention of DBD in insulin-deficient diabetes; and lastly 4: Determine the mechanism of action of myostatin on osteoblastic bone cells under normoglycemic and hyperglycemic conditions. Myostatin, which is thought to negatively affect both bone and skeletal muscle, may serve both as a marker of musculoskeletal function and surrogate for risk for fracture in those with T1D. Additionally, it may offer an opportunity for targeted intervention to prevent or improve musculoskeletal dysfunction associated with T1D. The knowledge gained from these studies will set the ground for future studies in musculoskeletal health in diabetes and will offer the candidate an opportunity to transition towards an independent career in the abovementioned field. This proposal presents a five-year research career development program focused on the study of muscle and bone interactions in Type 1 diabetes; and specifically, how muscle derived molecules, called myokines contribute to diabetic bone disease, a serious and emerging complication of Type 1 diabetes. The candidate currently holds a position as an Assistant Professor of Pediatrics in the Division of Pediatric Endocrinology at the University of Kentucky. She has 75% protected time for research, independent office space, laboratory space and access to all equipment and resources offered by the Barnstable Brown Diabetes Center. The candidate is strongly committed to an academic career in the field of musculoskeletal research in diabetes and is supported by her mentors and her department. The proposed study and the complementary didactic work will provide the candidate with research skills in basic, translational and clinical research thereby enabling to transition to an independent clinician scientist. Kathryn Kanzler, Psy.D.

UNIVERSITY OF TEXAS HLTH SCIENCE CENTER, SAN ANTONIO, TX

Acceptance Based Coping (ABC) Skills Delivered by Promotores for Hispanic/Latino Patients with T2DM

Project Summary/Abstract

Dr. Kathryn Kanzler is applying for a Mentored Patient-Oriented Research Career Development Award (K23). This grant would provide intensive training and time to gain necessary skills and knowledge toward achieving her long-term goal of being an independent clinical investigator of scalable behavioral treatments to reduce health disparities and improve outcomes for patients with diabetes. Dr. Kanzler is a clinical health psychologist with experience as a clinician-educator in the US Air Force. Building on a strong foundation, additional training is sought in health disparities, clinical research and design, type 2 diabetes (T2DM), and dissemination and implementation science. Proposed training will be accomplished with guidance of multidisciplinary mentors and training advisors largely based at the University of Texas Health Science Center San Antonio. Activities include coursework, directed readings, seminars, workshops, and mentor meetings. The proposed study helps address an urgent need for more interventions for the Hispanic/Latino diabetes population. Despite continued biomedical advances, diabetes and related complications continue to take a higher toll in the Hispanic/Latino community, where there is 40% greater likelihood of death compared to non-Hispanic whites. Avoidance coping has emerged as a key factor in preventing optimal glycemic control, but interventions that address avoidance coping, such as Acceptance and Commitment Therapy (ACT), are limited by language and cultural barriers to care, as well as standard dissemination practices (e.g., ACT is usually provided by licensed providers in specialty settings). Community health workers (“Promotores”) serving on primary care teams could deliver key elements of ACT for diabetes, in addition to mitigating other barriers to self-management, such as limited resources, language differences, and low health literacy. This task- shifting paradigm allows for culturally-tailored and accessible care. This task-shifting paradigm allows for culturally-tailored and accessible care. Task-shifting core skills of ACT–Acceptance Based Coping (ABC) skills—to supervised Promotores could provide accessible and culturally appropriate treatment in the preferred language of patients. This study aims to develop an acceptable and feasible Promotores- delivered ABC intervention program for Hispanics/Latinos using REP pre-conditions and pre- implementation steps, in (a) qualitative and quantitative data from interviews and focus groups (Phase 1); and (b) repeated small-sample beta-tests (Phase 2). The second aim, in preparation for a larger pragmatic trial, is to demonstrate feasibility in a pilot randomized trial through: evaluating recruitment and retention; establishing methods of assessing intervention fidelity and integrity; engaging multi-level stakeholders; and estimating the magnitude of potential impact on selected mechanisms and outcomes. This line of research has potential to improve glycemic control and quality of life for Hispanic/Latino patients with T2DM. This award will lay the foundation for a research program of scalable interventions that reduce health disparities through improved diabetes outcomes.

Shakun Karki, Ph.D.

BOSTON UNIVERSITY MEDICAL CAMPUS, BOSTON, MA

FSP27 regulation of vascular function in human obesity

Project Summary/Abstract

This proposal describes a 5-year training program that will expand the applicant's scientific knowledge, advance her expertise in patient-oriented translational research, and establish independence from her primary mentor. A 4-member Mentoring Team and 3-member Advisory Committee will oversee her training and career development. The current application represents a patient-oriented clinical proposal that examines mechanisms of obesity-associated vascular disease in human subjects. Adipose tissue dysfunction, lipotoxicity, and insulin resistance are essential abnormalities linking obesity to the pathogenesis of cardiovascular disease. This proposal will employ a number of complementary approaches harnessing physiological studies of vascular endothelial vasodilator function and angiogenesis, pharmacological and biological methods to probe dysfunctional signaling pathways, and proteomics-based approaches to gain novel insight into the role of a newly identified protein FSP27 in the pathogenesis of vascular disease in obesity. In aim1, we will investigate the role of FSP27 in depot- specific mechanisms of vascular dysfunction in the human adipose tissue microenvironment, using videomicroscopy and angiogenic assays to examine microvascular responses in both subcutaneous and visceral adipose compartments biopsied during elective surgical procedures, including bariatric surgery, in 150 obese and 50 lean subjects. We will characterize vascular phenotypes in relation to FSP27 signaling and test the hypothesis that down-regulation of FSP27 is linked to insulin resistance and vascular dysfunction. In aim 2, we will seek to identify molecular mechanisms that contribute to FSP27 regulation of vascular function by employing proteomics-based approaches using mass spectrometry to identify proteins and subsequent pathways differentially modulated by FSP27 in relevance to lipotoxicity, insulin resistance, and vascular biology. In aim 3, vascular studies, metabolic phenotyping, and proteomics will be repeated at 6-months following bariatric surgical intervention in the same 150 obese subjects from aims 1 and 2. We will test whether relevant FSP27- related molecular pathways identified in aim 2 are influenced by marked weight loss and/or metabolic changes and hypothesize that weight loss will improve vascular phenotype and linked to FSP27 signaling. The overall project will combine molecular biology with human physiology in severely obese individuals where clinically very little vascular data currently exist. The long-term goal of the applicant is to develop an academic career in the field of obesity and cardiovascular disease, and this proposal will allow the applicant to expand her translational expertise in an area that is relatively unexplored and medically important. Obesity will remain one of the most important health care challenges worldwide, and improving our understanding of mechanisms of obesity-related cardiovascular disease is critical. Lisa Karstens, Ph.D.

OREGON HEALTH & SCIENCE UNIVERSITY, PORTLAND, OR

Functional Considerations of the Urinary Microbiome in Overactive Bladder

Project Summary

Overactive bladder (OAB) is a symptom complex that is highly prevalent and negatively impacts quality of life. Multiple mechanisms likely contribute to OAB symptoms, though current therapies only target a few potential mechanisms. Recent evidence suggests that the urinary tract, which traditionally has been thought to be sterile in the absence of a clinically identifiable infection, actually harbors a variety of microbes, known collectively as the urinary microbiota, even when clinical cultures are negative. These microbial communities appear to be altered women with urge urinary incontinence (wet OAB), however it is unknown if the urinary microbiota are also altered in the broader patient population with OAB without incontinence (dry OAB) or how the urinary microbiota contribute to OAB symptoms. This proposal aims to determine if the urinary microbiota are also altered in women with dry OAB (Aim 1). Additionally, the proposed study also aims to lay the ground work for understanding the functional contribution of the urinary microbiota to OAB. In other body sites, commensal microbes contribute to host health and disease through interactions with the immune system and their metabolic activity. The proposed studies will identify interactions with the host immune system by identifying immunoglobulin- A associated microbes (Aim 2) and identify metabolic products of the urinary microbiota through metabolomics studies (Aim 3). Together, information gained through completion of these aims will lead to key insights that will guide biomarker and mechanistic studies aimed at improving the clinical management of patients with OAB. Candidate: Dr. Karstens has a unique background including training in bioinformatics and analytical chemistry. Her long-term goals are to apply this knowledge to understand the mechanisms underlying complex urinary tract disorders to streamline therapeutic options for women suffering from these conditions. Career development plan: The career development plan includes hands-on and didactic training to gain expertise in women’s health and clinical research, as well as expand her knowledge in the molecular techniques required to understand the function of the urinary microbiota. Environment: Her interdisciplinary mentorship team brings together expertise in bioinformatics (Dr. McWeeney), urogynecology (Dr. Gregory), microbiology (Dr. Wolfe), immunology (Dr. Rosenbaum), and host-microbe interactions (Dr.Braun). Training will be completed at Oregon Health & Science University, a leading biomedical research and training institution in the state of Oregon. OHSU has an excellent translational research and training infrastructure to facilitate the successful transition to an independent career in translational bioinformatics research focused on understanding bladder disorders. Michael Kattah, Ph.D., M.D.

UNIVERSITY OF CALIFORNIA, SAN FRANCISCO, SAN FRANCISCO, CA

Mechanisms of intestinal epithelial injury in intestinal inflammation

PROJECT SUMMARY AND ABSTRACT

There is a great need for new treatments for Inflammatory Bowel Disease (IBD). IBD affects over 1% of US adults, leading to disability, hospitalization, and surgery. IBD is characterized by damage to the “lining” of the intestine, also known as the intestinal epithelium. Intestinal epithelial damage leads to inflammation, bleeding, and an increased risk of colon cancer. Since a damaged intestinal epithelium is the defining feature of IBD, we want to understand what genes preserve intestinal epithelial integrity in order to better treat IBD. Genome-wide association studies (GWAS) have linked hundreds of genes with small individual effects to IBD, but it is unclear how these genes cooperate to increase susceptibility to intestinal injury and inflammation. We have a mouse model to study the role of two IBD-associated genes, A20 and ABIN-1, in intestinal epithelial cells (IECs). We were the first to show that A20 and ABIN- 1 work together to maintain a healthy intestinal epithelium by preventing IEC injury. We grew intestinal organoids from the intestines of these mice. Intestinal organoids are miniature replicas of the intestinal epithelium grown in a dish in the lab. Using this system, we discovered that IECs lacking A20 and ABIN-1 are exquisitely sensitive to cell death from tumor necrosis factor (TNF). TNF is one of the primary death signals in the intestine, and antibodies against TNF are commonly used to treat IBD. We have dramatic evidence that A20 and ABIN-1 cooperate to preserve intestinal well-being, but we still need to understand the specific ways in which they preserve IEC health. We will use the mouse model and our organoids to determine which inflammatory cells injure IECs in the absence of TNF. We have evidence that inflammatory cells secrete a factor or factors other than TNF that kill A20 & ABIN-1 deficient IECs. This could be a very important clue as to why some patients with IBD do not respond to anti- TNF neutralizing antibodies. Additionally, A20 and ABIN-1 might cause epithelial injury by altering how intestinal stem cells “differentiate” or mature into other IEC subsets. Two subsets of particular interest are paneth cells and goblet cells, but there are many other subsets that we will study using our unique system. Finally, and perhaps most importantly, we will also grow intestinal organoids from IBD and non-IBD patients and test susceptibility to cell death. Using the organoids, we can attempt to rescue cell death with small molecules and drugs that are currently in clinical trials. We will analyze IEC subsets in the organoids, and see how TNF and other stimuli perturb IEC subsets. Lastly, we will work on “editing” human organoids with a technique called CRISPR/Cas9, to improve our ability to study other genes in IBD. The long-term goal of these studies is to identify approaches of treating IBD that protect epithelial cells from injury, rather than simply suppressing the immune system. Chia-Chi Key, Ph.D.

WAKE FOREST UNIVERSITY HEALTH SCIENCES, WINSTON-SALEM, NC

Role of Hepatic GDPD3 in Mechanisms of Lipid Metabolism

PROJECT SUMMARY/ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), affecting ~30% of the U.S. population, is projected to replace hepatitis C as the leading cause of liver transplantation by 2020. Developing effective NAFLD treatments is hampered by a poor understanding of its underlying mechanisms, including complex interactions between genetic and environmental factors. Glycerophosphodiester phosphodiesterase domain- containing protein 3 (GDPD3) is a newly discovered enzyme containing lysophospholipase D activity that converts lysophospholipid to lysophosphatidic acid (lysoPA) in non-hepatic cells. Mammalian GDPD3 has not previously been implicated in hepatic lipid metabolism. Our preliminary data indicates a positive correlation between human GDPD3 expression and triglyceride (TG) accumulation in hepatocytes and mouse livers, suggesting a novel gene in the regulation of hepatic TG homeostasis. Nonetheless, the intracellular locations, substrate specificity, physiological function, and molecular mechanisms of human GDPD3 in hepatocytes/livers are unknown. Therefore, in this study, with the guidance of a highly experienced multi-disciplinary mentoring group, we propose to investigate enzymatic properties of human GDPD3 and explore whether human GDPD3 is a causal gene for hepatic steatosis. More specifically, we are asking three questions: 1) Is human GDPD3 an endoplasmic reticulum membrane- associated enzyme containing lysophospholipase D activity? 2) Does human GDPD3 increase lysoPA production resulting in increased hepatic TG synthesis and accumulation via the glycerol phosphate pathway? 3) Does human GDPD3-produced lysoPA activate peroxisome proliferator-activated receptor gamma (PPARγ) which enhances hepatic steatosis via increased fatty acid (FA) uptake and TG synthesis? To answer these questions, we will overexpress human GDPD3 in hepatoma cell lines and in mouse liver to determine: a) the effect of human GDPD3 overexpression on oleic acid-induced TG accumulation in hepatoma cells and diet-induced hepatic steatosis in mice; b) the subcellular localization of human GDPD3 in hepatoma cells and mouse primary hepatocytes; and c) the amount and molecular species of GDPD3 lipid substrates and products in mouse livers. Liver-specific human GDPD3 overexpressing mice with loss-of-function or gain-of-function in PPARγ will be fed chow or a Western-type diet to induce hepatic steatosis. We will perform comprehensive hepatic and systemic metabolic phenotyping on these mice. Primary hepatocytes will be used to investigate de novo lipogenesis, FA uptake and incorporation into TG, FA oxidation, and very low density lipoprotein-TG secretion using radioactive isotopes. When the proposed aims are achieved, we will have a better mechanistic understanding of the relationship between human GDPD3 and hepatic steatosis, to address the gap in knowledge regarding NAFLD pathogenesis and inform strategies for its treatment. Finally, this proposal provides the necessary training and mentored guidance for the applicant to transition to a successful career as an independent investigator in lipid and glucose metabolism.

Timothy Kline, Ph.D.

MAYO CLINIC ROCHESTER, ROCHESTER, MN

Advanced MR Imaging and Image Analytics as a Precision Medicine Tool to Manage ADPKD

ABSTRACT

The goal of this NIDDK Mentored Research Scientist Development Award is to provide an organized scientific and educational environment for Dr. Timothy Kline to begin his transition into an independent research career focused on developing novel imaging technologies and image analysis techniques for abdominal organ pathologies. This proposal outlines a five-year training plan at Mayo Clinic under the primary mentorship of Dr. Bradley Erickson and a Mentoring Team comprised of accomplished researchers in the fields of: biology, nephrology, genetics, radiology, informatics; medical physics, biostatistics, image processing, and physiology. The focus of this proposal is to improve both research studies and disease prognosis for autosomal dominant polycystic kidney disease (ADPKD) patients through biomedical imaging techniques. It is well understood that imaging is essential for ADPKD diagnosis, monitoring, and outcome prediction. Clinical studies utilize total kidney volume (TKV) (as measured by MRI as an image-based biomarker) to follow the progression of ADPKD, as larger TKVs have been shown to correlate with worse prognosis in both human and animal-model studies. However, there are challenges with using TKV as a marker of disease progression. For one, it is a simplification of the disease state and does not inform on microscopic disease processes that are involved with piecemeal destruction of healthy renal tissue. In addition, measurements of TKVs are time consuming, costly, and poorly standardized. The introduction of automated approaches for measuring TKV will: greatly improve measurement throughput, significantly reduce costs associated with performing research studies, allow accurate and reproducible measurements to be obtained both within and across institutions; facilitate the search for new imaging biomarkers. The specific aims of this project are to: (i) develop and validate automated tools to characterize renal structure, such as TKV and cystic burden; (ii) explore new imaging biomarkers by image texture feature analysis and pattern recognition techniques; and (iii) develop a new technique to measure renal blood flow. This research will be facilitated by Mayo Clinic's outstanding clinical and research environment dedicated to improving patient care, as well as the Mayo Clinic Translational PKD Center, which focuses on translating basic science research into improvements in the management and treatment of ADPKD patients. Dr. Kline's background in imaging technologies and image processing makes him particularly suited to perform this research. In addition to the above aims, Dr. Kline will: 1) develop a strong knowledge base in both nephrology and radiology by attending relevant rounds, seminars, and national conferences; 2) enhance his knowledge of medical imaging, biology, physiology, genetics, and programming through coursework and mentoring; 3) attend workshops focused on grant and publication writing; and 4) submit a highly competitive R01 application expanding upon the findings from this research proposal. This proposal will lead to vast improvements to current analysis workflows, as well as an improved understanding of the prognostic power of new imaging biomarkers of ADPKD. Obtaining this K Award will greatly facilitate Dr. Kline's transition into a prosperous independent research career. Hilaire Lam, Ph.D.

BRIGHAM AND WOMEN'S HOSPITAL, BOSTON, MA

Impact of mitochondrial dynamics on the development of renal cysts in Tuberous Sclerosis Complex (TSC)

ABSTRACT

This K01 proposal describes a five-year research and training plan that will facilitate the transition of Dr. Hilaire Lam to an independent academic investigator in the field of mitochondrial dynamics in kidney disease. Dr. Lam has a strong background in molecular biology and pathological processes. As a postdoctoral fellow she investigated the molecular mechanisms and aberrant metabolism supporting kidney cystogenesis in tuberous sclerosis complex (TSC). While mTORC1 inhibitors slow TSC disease progression, disease activity resumes soon after treatment cessation, necessitating life long treatment. Therefore, Dr. Lam seeks to identify more durable therapeutic responses through a better understanding of mitochondrial dynamics in TSC. The candidate has confirmed that TSC-deficient cells are characterized by increased mitochondrial content, and discovered that Tsc2-deficient cells depend on the upregulation of certain adaptive molecules (p62/SQSTM1 and miR-21) to maintain mitochondrial integrity. The role of mitochondrial dynamics, particularly fission, a process directly regulated by mTORC1 in the development and progression of mTORC1-driven kidney cystogenesis represents a key knowledge gap. Dr. Lam will address the central hypothesis that mTORC1-hyperactivated cells depend on STAT3 to regulate mitochondrial function and fission to support metabolic reprogramming and cyst formation. This hypothesis will be addressed in the following Specific Aims: 1. To determine the effects of Stat3 signaling on mitochondrial dynamics and metabolic reprogramming in Tsc2-deficient cells. Aim 2: To determine the dependence of cyst formation and progression on mitochondrial fission. The significance and innovation of this project is to identify mitochondrial targets to increase the durability of therapy of the renal manifestations of TSC. The contribution of mitochondrial fission to cellular metabolism may have translational relevance to other cystic renal diseases. Dr. Lam will receive mentorship from her outstanding scientific advisory committee composed of distinguished scientists with expertise related to key areas of this proposal including mitochondria (Finkel), metabolism (Asara), signaling (Manning) and kidney redox homeostasis (Pal) and polycystic kidney disease (Zhou). The Department of Medicine is committed to Dr. Lam's success and will continue to provide resources for her as a member of the faculty. The candidate's mentor, Dr. Elizabeth Henske, is a NIH funded researcher in the field of TSC/LAM and metabolism with experience training and transitioning mentees to independence. A detailed career development and training plan is presented that includes research, coursework, readings, seminars and presentations at scientific meetings. The candidate details a timeline for completion of research aims, preparation of manuscripts and R01 application. The expertise and skills gained from this K01 will prepare Dr. Lam to obtain the R01 funding necessary to launch her career focused on mitochondrial dynamics in disease. Craig Lammert, M.D.

INDIANA UNIV-PURDUE UNIV AT INDIANAPOLIS, INDIANAPOLIS, IN

Autoimmune Hepatitis: Genetic and Environmental Risks

PROJECT SUMMARY

The principle genetic and environmental risk factors underlying the pathogenesis of autoimmune hepatitis (AIH) are not well investigated. Better understanding the explainable risks in AIH is essential to elucidate disease heterogeneity, predict outcomes, and transition to a model of individualized clinical care. In this career development application, the candidate is proposing a hypothesis-driven research strategy to address important knowledge gaps in the field of genetic and environmental risks in AIH. This builds on a recently completed prospective study of patients with AIH and matched controls funded by an institutional KL2 award. The specific aims of the research strategy are to (1) develop a large cohort of patients with AIH and matched controls with detailed assessment of environmental risk factors and linked biologic samples utilizing traditional and novel recruitment strategies; (2) interrogate selected environmental triggers associated with new onset AIH; and (3) identify novel disease specific HLA associations and assess for genetic and environmental interactions. To achieve these aims, the candidate will (1) develop a large cohort of patients with well phenotyped AIH and matched controls who undergo carefully assessment of environmental risk factors and provide their genomic DNA; (2) systematically interrogate viral and medication-related environmental triggers of newly diagnosed AIH patients and two control groups; (3) conduct HLA deep sequencing utilizing the Illumina MiSeq platform to identify putative HLA-specific associations and perform gene-environment interaction studies. The proposed career development plan integrates in-depth mentoring from a multidisciplinary team of senior scientists, advanced coursework in genomics, genetic and molecular epidemiology, and immunology; experiential learning through the conduct of the proposed research plan, and a highly supportive research environment. The mentorship team, which includes independent investigators with expertise in clinical and translational research in hepatology (Chalasani), immunologic-mediated drug- hypersensitivity and HLA genetics (Phillips), clinical virology and mentoring (Gupta); epidemiologic data analysis and biostatistics (Gao), will guide the candidate's research and career development. The superb institutional infrastructure for facilitating junior investigators and substantial institutional commitment to the candidate greatly strengthen this application. At the conclusion of this program, the candidate will be well positioned to become an independent physician-investigator in autoimmune liver diseases and hepatogenomics. Dominick Lemas, Ph.D.

UNIVERSITY OF FLORIDA, GAINESVILLE, FL

Human milk metabolomics and microbe-host interactions associated with pediatric obesity

PROJECT SUMMARY/ABSTRACT

Pediatric obesity affects 1 in 3 children in the US and represents a public health crisis [1]. In utero exposure to maternal obesity and caesarean section (C-section) are independently and jointly associated with offspring obesity [2, 3]. Accumulating data suggests maternal obesity and C-section may seed an “obesogenic” microbiome responsible for transmitting obesity risk from mother to child [4, 5]. Exclusive breastfeeding is associated with protection against pediatric obesity [6]; however, the association between human milk components and the microbiome that account for these observations are poorly characterized. Recent advances in untargeted metabolomics provides a novel opportunity to comprehensively investigate how the human milk metabolome in combination with microbe-host interactions are associated with infant adiposity during the first year of life. The Principal Investigator, Dr. Dominick J. Lemas, Ph.D. will use high-resolution mass spectrometry as a tool to interrogate the human milk metabolome and infant microbiome in a longitudinal maternal-infant cohort of obese and normal weight mothers that includes vaginally and cesarean section deliveries. The over-arching hypothesis of this proposal is that the human milk metabolome will be associated with novel changes in the early infant microbiome that alter risk for obesity and weight gain in the first year of life. In collaboration with a multidisciplinary team of expert mentors (including primary mentor Dr. Christian Jobin and co-mentors Drs. Josef Neu, Timothy Garrett, William Hogan, Janice Krieger), the primary goal of this K01 career development proposal is for Dr. Lemas to develop expertise in high-resolution metabolomics, biomedical informatics and longitudinal clinical microbiome studies. The University of Florida (UF) boasts an intellectually rich research environment that will facilitate these goals. More specifically, longitudinal data collection will be completed at the NIH-funded UF Clinical and Translational Science Institute and human milk and stool metabolomics analysis will be completed at the NIH-funded UF Southeast Center for Integrated Metabolomics (SECIM). Finally, microbial sequencing will be completed at the UF NextGeneration DNA Sequencing Core and high-throughput data analysis will be completed at the UF Research Computing Center. Specific Aim 1 will generate a data-driven recruitment plan using informant interviews and qualitative methods to identify barriers and facilitators to successful participant recruitment and strategies. Specific Aim 2 will characterize the role of the human milk metabolome on the development of the infant microbiome. Specific Aim 3 will identify how the human milk metabolome is associated with infant adiposity. The proposed research is focused on the pathophysiology of infant growth and adiposity during early life and will inform subsequent studies that seek to reduce the risk of pediatric obesity. The structured mentoring and training plan is designed to facilitate Dr. Lemas's long-term goal of developing an independently-funded clinical and translational research program focused characterizing the microbe-host interactions that mediate the protective role of breastfeeding on pediatric obesity, consistent with the mission of the NIDDK. Belinda Lennerz, Ph.D., M.D.

BOSTON CHILDREN'S HOSPITAL, BOSTON, MA

Effects of dietary carbohydrate on postprandial metabolism, brain function and type 1 diabetes control

PROJECT SUMMARY

The scientific goal of this application is to explore the mechanisms linking diet to metabolism and body weight control, with relevance to type one diabetes mellitus (T1D). The career development goal is to advance my training in neuroimaging, nutrition, and clinical trial conduct - in preparation for my transition to independence. Despite major technological advances, management of T1D remains suboptimal, putting millions of people at risk for immediate and long-term complications. Moreover, individuals with T1D have elevated rates of dysregulated eating, overweight, and metabolic syndrome, further contributing to chronic disease risk. In this proposal, I consider control of the postprandial state as mechanistically related to each of these pathophysiological challenges. After meals, a mismatch between carbohydrate absorption rate and insulin action typically leads to alternating periods of hyper- and hypoglycemia, compounded by potential adverse effects of higher systemic insulin exposure (e.g. anabolic effects, insulin resistance). A conceptually promising approach to control both problems is dietary carbohydrate modification to reduce postprandial glycemia and insulin needs. In prior work, I demonstrated that high- vs low- glycemic index (GI) carbohydrate increased hunger and activation of brain areas involved in addiction and craving in individuals without diabetes. A survey study documented exceptional glycemic control (HbA1c 5.7% and low acute complication rate) among 316 children and adults with T1D consuming a very-low-carbohydrate diet (VLCD). Here I propose two complementary studies: (1) Leveraging neuroimaging and metabolic data obtained from a controlled, cross-over feeding protocol involving intravenous insulin administration, I will explore whether the effects of high GI carbohydrates on hunger, metabolic outcomes, and brain function are mediated by mechanisms related to blood glucose, serum insulin or both. (2) In a 12-week randomized trial, I will assess the safety and preliminary efficacy of a VLCD in the management of adolescents and young adults with T1D. As a result of these studies, I hope to help elucidate mechanisms relating type and amount of dietary carbohydrate to glycemia, brain control of body weight and cardio metabolic risk in T1D, and provide data of direct relevance to the clinical management. Ken Lim, M.D.

INDIANA UNIV-PURDUE UNIV AT INDIANAPOLIS, INDIANAPOLIS, IN

Dissecting the role of soluble a-Klotho in cardiovascular aging

PROJECT SUMMARY/ABSTRACT

The proposed career development award will foster and promote the candidate's training and evolution toward independent investigator. Candidate: Dr. Kenneth Lim is a clinical-translational investigator at the Division of Nephrology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS). The proposed study integrates patient-oriented research, cardiopulmonary exercise testing (CPET) technology, proteomics and computational biology embedded in a rigorous training plan. Mentorship: Dr. Ravi Thadhani is the Chief of Nephrology at the MGH, Professor of Medicine at HMS and Executive Director of Clinical Trials at Partners Healthcare. He will serve as the primary mentor in conjunction with a multidisciplinary team of expert collaborators. Research: Age-associated changes of the cardiovascular system and its complications are the leading cause of death in patients with chronic kidney disease (CKD). Despite this, there are currently no direct therapies available to treat this condition today. Klotho is a protein present in circulation that exerts highly pleiotropic aging suppressive effects. Animal studies have demonstrated promising therapeutic properties of Klotho that could be used for the treatment of cardiovascular disease in CKD. However, several fundamental problems must first be overcome before future human interventional studies can proceed: Firstly, published studies examining circulating Klotho with cardiovascular outcomes to-date have focused mainly on morphological alterations, while clear evidence has shown that aging is tightly associated with reduced cardiovascular functional reserve. Secondly, the precise levels of the various circulating isoforms of Klotho and the nature of their specific roles in cardiovascular health are still undefined. Additionally, clinical studies to-date assessing circulating Klotho are limited by the lack of a reliable Klotho assay. The overall aim of the proposed study is therefore to bridge a critical gap in our understanding of the role of circulating Klotho in the regulation of the cardiovascular aging response in CKD. We hypothesize that circulating Klotho deficiency is a major determinant of premature cardiovascular aging in CKD. In specific aim 1, we will characterize the relationship of the various Klotho isoforms with cardiovascular structure and functional reserve using state-of-the-art CPET technology. We will define levels of circulating Klotho isoforms in health and advanced CKD (cross-sectional), and after kidney transplantation (prospectively). Circulating Klotho isoform levels will be assessed using an established immunoprecipitation and western blotting method. Additionally, the proposed study will provide a robust platform to validate and confirm a new targeted proteomics assay that we have developed for the precision assessment of circulating Klotho isoforms. In specific aim 2, we will conduct a cross-section study to characterize the relationship of circulating Klotho with premature vascular changes using human arteries from healthy and CKD patients. To further determine therapeutic properties of Klotho, we will conduct an interventional study using arterial explant organ cultures. Eugene Lin, M.S., M.D.

UNIVERSITY OF SOUTHERN CALIFORNIA, Los Angeles, CA

Investigating the Economic and Social Determinants of Home Dialysis Drop-Out

PROJECT SUMMARY/ABSTRACT

This K08 proposal aims to provide Dr. Eugene Lin, MD, MS, with the protected time, training, mentorship, and research experience to allow him to become an independent health services researcher in kidney diseases. As an academic nephrologist, Dr. Lin focuses his research on health policy issues and efficient care delivery pertaining to kidney diseases and, in particular, dialysis. Home dialysis offers patients independence and improved quality of life, and it is less costly to society. However, many patients choose to switch from home dialysis to in-center dialysis, especially when they do not receive adequate support or when they are not sufficiently prepared. This dropout from home dialysis is costly and a potential loss in patient and societal well- being. Recent dialysis payment reform through the end- stage renal disease (ESRD) Prospective Payment System (PPS) catalyzed an increase in home dialysis use, by making it more profitable relative to in-center dialysis. However, it is unclear if payment reform simultaneously incentivized the recruitment of patients who are more likely to drop out or if it led to the emergence of facilities less equipped to support patients on home dialysis. Additionally, little is known about potential solutions to mitigate home dialysis dropout. To address this knowledge gap, this study aims to: (1) quantify changes in home dialysis dropout due to the ESRD PPS, (2) identify changes in the types of facilities offering home dialysis and changes in the composition of patients receiving home dialysis, and (3) evaluate potential strategies that might decrease home dialysis dropout, including retraining and periods of respite. Dr. Lin will accomplish these research goals by applying econometric techniques to a large claims database and by developing a dynamic simulation model of patients undergoing home dialysis. This proposal is highly likely to make a substantial impact on patient care by potentially changing how facilities manage patients on home dialysis. Studying the impact of payment reform will also help policy makers better understand its effects and will help inform the design and refinement of future policies aimed at improving the provision of home dialysis. To accomplish these aims, Dr. Lin will receive additional training in econometrics and design science, under the guidance of a multi-disciplinary mentorship team. The proposed work and career development plan are realistic and feasible within the award period and will ensure that Dr. Lin develops the skills to compete for R01 funding. In short, this K08 award will provide the training, research experience, and mentorship needed to help Dr. Lin successfully transition into an independent health services researcher. Elizabeth Lorenz, M.D.

MAYO CLINIC ROCHESTER, ROCHESTER, MN

Improving frailty and long-term outcomes after kidney transplantation

PROJECT SUMMARY/ABSTRACT

Improving the long-term survival of kidney transplants (KT) is a national priority in the US. The prevalence of end-stage renal disease is increasing, and the organ shortage is growing. Unfortunately, long-term graft and patient survival has not improved since the 1990s. The half-life of a deceased donor kidney is currently only fifteen years. The most common cause of kidney transplant failure is patient death. In order to address this health crisis, the transplant community needs to focus on modifiable risk factors for patient death and other adverse long-term outcomes after kidney transplantation, such as frailty. Frailty is a clinical syndrome characterized by decreased physiologic reserve and is common in patients with chronic kidney disease (CKD). Frailty prior to KT has been associated with increased post- transplant mortality and has been shown to be modifiable in non-transplant patients. However, there is a significant knowledge gap regarding frailty after KT, including risk factors for its development, biomarkers with which to identify it, and interventions with which to improve it. Cellular senescence is an exciting new area of frailty research that directly applies to these deficits. During the process of senescence, metabolic stressors cause cells to enter a state of permanent growth arrest. Senescent cells accumulate throughout the body and secrete factors collectively called the senescence- associated secretory phenotype (SASP) which induce formation of other senescent cells and cause surrounding tissue damage. Cellular senescence is a mechanism of aging and age-related diseases such as frailty. Components of the SASP serve as biomarkers of frailty in non-transplant populations and may help us identify KT recipients at high risk of functional decline and premature death. In addition, frailty biomarkers could ultimately serve as surrogate endpoints in clinical trials designed to improve frailty. The overall objective of this application is to 1) identify frailty trajectories after kidney transplantation, 2) identify biomarkers of frailty after kidney transplantation, and 3) conduct a phase II clinical trial examining the preliminary efficacy, feasibility and acceptability of an exercise intervention on post- transplant frailty. The proposed K23 application involves the use of innovative biomarkers and behavioral interventions to improve frailty and long-term outcomes after KT, a priority for the NIDDK which focuses on bridging translational research gaps to improve the health and quality of life of patients with CKD. The candidate has exceptional resources available to her: a multidisciplinary team of expert mentors; access to a large volume of transplant patients; and excellent career development activities, i.e., formal courses and workshops in statistical methods, biomarker development, and behavioral clinical trials. Together, these resources will allow the candidate to achieve her long-term goal of becoming an independent investigator and nationally recognized expert on the use of biomarker technology and behavioral interventions to improve frailty and long-term outcomes after KT. Keri Lunsford, Ph.D., M.D.

RBHS-NEW JERSEY MEDICAL SCHOOL, NEWARK, NJ

Defining Immunology Frailty as a Predictor of Human Liver Allograft Recipient Futility

Project Summary/Abstract

DESCRIPTION: This application for a Mentored Clinician Scientist Development Award (K08) is designed to evaluate the effect of cirrhosis-induced immune dysfunction on liver transplant outcomes. The candidate is a transplant surgeon and immunologist whose long-term goal is to develop treatments to prevent immune frailty and improve liver transplant recipient survival. In order to fulfill the educational objectives of this award, this proposal will expand the applicant’s knowledge base into novel lines of translational research inquiry and delve into new areas of investigation requiring focused mentorship. The mentors assisting in the applicant’s development will be crucial for her success, for the performance of the proposed studies, and for the educational mission of the award. Xian Li, PhD, a leader in basic science immunology, and Mark Ghobrial, MD, PhD, a leader in clinical and translational research, will serve as co-mentors. They will be assisted by Drs. Dale Hamilton and Anisha Gupte, who will provide mentorship in bioenergetics; Dr. Ed Graviss, who will provide expertise in biostatistics; Dr. Wenhao Chen, who will provide expertise in T cell exhaustion, and Dr. Todd Eagar, who will provide expertise in inflammation. A rigorous career development plan will be implemented, including structured workshops, biostatistics course work, scientific seminars, and lab meetings. This combination will be instrumental in ensuring the candidate’s successful transition to a career as an independent investigator. PROJECT SUMMARY: Due to organ shortage, livers are transplanted in order of recipient medical urgency; however, ethical principles dictate avoidance of futile transplantation. Due to imbalance in supply and demand for organs, illness severity (categorized by MELD score) has drastically increased. MELD does not correlate well with risk of death post-transplant; thus, better metrics to evaluate risk of mortality are necessary. The most common cause of death early after liver transplant relates to consequences of an immune system which is frail or dysfunctional prior to transplant. In pre- transplant cirrhotics, immune frailty may relate to metabolic deficiencies or T cell exhaustion and may affect liver transplant outcomes. Based on this, it is hypothesized that the pre-transplant state of immunologic frailty results from cirrhosis-related alterations in recipient metabolism, resulting in global T cell metabolic dysfunction, increased T cell exhaustion, and limited adaptive immune proliferation and function. Persistence of immunologic frailty following liver transplant results in increased recipient mortality. This will be addressed by three Aims, which will (1) evaluate metabolic alterations in liver transplant recipients and determine the bioenergetics pathways involved in immune frailty, (2) examine the role of T cell exhaustion in immune frailty, and (3) determine the longitudinal effect of frailty on the recipient post-transplant immune response. IMPACT: These studies are critically important to identify biological markers of immunologic frailty. Defining pre- transplant frailty will improve patient selection for liver transplant, prevent wasting of livers in patients at high risk of futility, and identify therapeutic targets to reverse frailty and improve patient survival. Shan Luo, Ph.D.

UNIVERSITY OF SOUTHERN CALIFORNIA, Los Angeles, CA

Neural Mechanisms of Overeating Among Children Exposed to Gestational Diabetes Mellitus In Utero

PROJECT SUMMARY

Childhood obesity rate has been increasing dramatically, at great cost for health care and wellbeing. It is highly likely that obesity will persist into adulthood; thus, identifying risk factors contributing to childhood obesity is extremely critical so that prevention strategies can be taken early to reduce the obesity rate. Research suggests that children who were exposed to gestational diabetes mellitus (GDM) in utero have increased propensity of developing obesity. However, there are few human studies aimed to understand the mechanisms explaining this relationship. The goal of this study is to test the hypothesis that in utero exposure to GDM leads to abnormal development of brain pathways that regulate eating behavior, which in turn increases food intake and risk for obesity in children. To accomplish this goal, the applicant and her team plan to 1) examine effects of GDM on food intake; 2) associations of GDM exposure with structural and functional connections between brain appetite and reward pathways; 3) explore whether changes in brain circuitry involved in feeding behavior would mediate the link between GDM exposure and increased food intake thus increasing risk for obesity. This will be the first study in humans to use multi-modal imaging techniques to examine neural mechanisms of the link between GDM exposure and obesity risk. Through extensive mentor-directed training, course work and workshops, manuscript and grant writings, scientific presentations at national conferences, the candidate will achieve three training goals 1) strengthen knowledge about GDM physiology, pediatric obesity and nutrition in children (co-mentor Dr. Kathleen Page); 2) build skills in multi- modal imaging, and learn computational methods (e.g., psychological-physiological-interaction, independent component analysis, DTI fiber tracking, graph theory) to analyze functional and structural connectivity data under supervision of Drs. Paul Thompson (primary mentor) and Neda Jahanshad (co- mentor); 3) strengthen skills in advanced statistical modeling (e.g., mediation analysis) (co-mentor Dr. Anny Xiang). By the end of the proposed training period, the candidate will be positioned uniquely as a leader in investigating neural mechanisms for obesity risk in children exposed to maternal diabetes in utero with skills from various disciplines, and compete for an independent R01 application. Furthermore, this proposed project will help the candidate progress towards achieving her long-term career goal, which is to become an independent and innovative research scientist dedicated to investigating underlying mechanisms of obesity from a joint perspective of neuroscience, psychology and endocrinology. Janet Lydecker, Ph.D.

YALE UNIVERSITY, NEW HAVEN, CT

Development and Initial Efficacy Testing of a Cognitive-Behavioral Intervention to Treat Adolescent Binge Eating

PROJECT SUMMARY/ ABSTRACT

Binge-eating episodes involve eating an unusually large amount of food and feeling unable to control the type or amount of food consumed while eating. Among adolescents, loss-of-control (LOC) eating is a precursor to excess weight gain and binge-eating disorder. Binge/LOC eating is also associated with physical and mental health impairments, and psychosocial distress. Although excess weight and binge/LOC eating have serious immediate and long-term consequences, assessment and treatment have received minimal attention for adolescent patients. Among adults, cognitive-behavioral therapy (CBT) has a strong evidence base for binge-eating disorder. Among adolescents, there are no well-established treatments for binge/LOC eating. Adolescents have unique social, cognitive and emotional needs because of their developmental stage; unique treatment approaches are essential to meet these unique needs. This Mentored Patient-Oriented Research Career Development Award (K23 with Clinical Trial) builds upon the candidate’s programmatic clinical-research interest in the treatment of weight and eating disorders. Training objectives and research activities for this K23 are to develop new skills and expertise in (a) treatment development and initial efficacy testing as described by the Stage Model of Behavioral Therapies Research, and (b) advanced statistical skills for repeated and dyadic data. Development of CBT for adolescent binge/LOC eating will make a significant contribution to our understanding of treatment in an important step towards reducing health impairments and psychosocial distress associated with adolescent binge/LOC eating and excess weight. Research activities are tightly integrated with training activities and mentorship from an advisory team of internationally-recognized scholars to facilitate the candidate’s development of necessary skills to become an independent and impactful investigator in the fields of childhood obesity and eating disorders. The training plan enlists the mentorship of Carlos Grilo, PhD, Professor of Psychiatry and Psychology and Director of the Yale Program for Obesity, Weight, and Eating Research, and Wendy Silverman, PhD, Alfred A. Messer Professor in the Yale Child Study Center and Professor of Psychology and Director of the Yale Child Study Center Program for Anxiety Disorders. Dr. Grilo’s expertise in the treatment of adult binge-eating disorder and obesity is complemented by Dr. Silverman’s expertise in youth treatments. Together, their mentorship will facilitate successful execution of the proposed research aims and career objectives. Research and training activities will also make use of other clinical-research educational programs at Yale (e.g., CTSA, Psychotherapy Development Center) that offer many formal training activities, opportunities, and resources. Skills (treatment development and advanced statistical analysis) will yield a new treatment and initial efficacy data for future tests of efficacy and effectiveness throughout the candidate’s career.

Matthew Lynes, Ph.D.

JOSLIN DIABETES CENTER, BOSTON, MA

The Lipidomics of Adipose Tissue Thermogenesis

Project Summary/Abstract

Obesity is a global pandemic with enormous financial burden and health cost due both to the increasingly large proportion of the population who are overweight or obese and the broad range of associated sequelae such as cardiovascular disease, type 2 diabetes, and some cancers. The development of strategies to prevent or treat human obesity is therefore extremely important. Recently, a great deal of interest has been centered on the metabolic capacity of brown fat in humans and the discovery and standardization of treatment regiments that activate brown fat thermogenesis and expend energy. In order to activate brown adipose tissue, several research strategies have been pursued, including both pharmacological interventions and physiologic cold exposure. Unfortunately, the thermogenic effect of many compounds that occurs in mice is not observed in humans, although recently B-3 adrenergic agonists that activate human BAT have been recently been reported. To this end, an innovative approach is proposed wherein I plan to investigate bioactive lipids as a novel class of circulating factors with potential pro-thermogenic effects in brown adipose tissue. Recently, studies have demonstrated that lipid molecules can act as hormones secreted to act as signaling molecules in distal organs. These lipids promote insulin sensitivity and glucose tolerance through their interaction with proteins located on the cell membrane. Conventionally, lipids have not been considered as potential endocrine factors. With this in mind, I have investigated the potential of lipids to act as secreted molecules that mediate, at least in part, the physiologic response to cold challenge. Preliminary studies have made the novel discovery that the lipid one specific target lipid species increases in circulation of mice and humans that are exposed to cold and further, systemic injection of this lipid can increase thermogenesis. This approach is innovative because lipid molecules have not been previously reported as secreted mediators of thermogenesis. In this proposal, I will systematically determine the effect of this lipid on whole body energy metabolism and determine the underlying molecular mechanisms that mediate enhanced thermogenesis after treatment with our target lipid. These results could a have significant impact in the development of treatments for obesity and cardiovascular disease. Faisal Malik, M.S., M.D.

SEATTLE CHILDREN'S HOSPITAL, SEATTLE, WA

Leveraging Social Media to Improve Health Outcomes in Adolescents with Diabetes

PROJECT SUMMARY/ABSTRACT

This proposal describes a five-year research training program that will allow me to achieve my long-term goal of becoming an independent faculty-scientist dedicated to improving healthcare quality and patient health for youth with diabetes. The majority of adolescents with type 1 diabetes (T1D) in the United States are not meeting the goals of therapy. I propose to develop tools to provide innovative diabetes management support to adolescents outside of the clinic setting through the use of social media (SM) and evaluate its feasibility and acceptability to adolescents with T1D. SM has become an integral part of adolescents’ lives and at this time, there is an absence of high quality empirical studies investigating the use of SM in the healthcare setting. Given the need for impactful interventions to support adolescent T1D management, this work has the potential to result in a major advance for disease management in this vulnerable population. The goal of this proposal is to develop a patient-centered SM intervention that will enhance patient self-efficacy and improve health outcomes for adolescents with T1D. I will achieve this goal by first identifying several different SM intervention strategies for distinct segments of adolescents with diabetes. In Aim 1, I plan to conduct a survey of a large sample of adolescents with T1D to examine perceptions on how SM can be utilized in diabetes management and then using latent class analysis techniques, I will segment adolescents based on patient characteristics and preferences around the use of SM. In Aim 2, I will incorporate additional stakeholder input, including caregivers and healthcare team members, through the use of user-centered design workshops to develop a pilot SM intervention based on previous formative work. By engaging adolescents, caregivers, and healthcare team members in an iterative process of user-centered design workshops I hope to develop a SM intervention that is consistent with key stakeholder values, preferences and goals. In Aim 3, I will pilot the SM intervention in a randomized controlled trial and examine its feasibility and acceptability. In addition, I plan to explore the impact on patient-reported outcomes (e.g. self-efficacy) and glycemic control. My research will be supported by an outstanding group of mentors with expertise in social media research, pediatric diabetes, survey research, and the design of clinical interventions. By accomplishing the aims in this proposal, I will address current gaps in our understanding of how to leverage SM to support adolescents with chronic disease. In addition, this proposal will allow me to take the next critical steps on my research training trajectory by providing me the opportunity to acquire additional research skills required to design, evaluate and implement behavioral health interventions using social media. Ultimately, this proposal will allow me to build a research program to investigate system-oriented, patient-centered solutions to improve the delivery and quality of medical care to youth with diabetes. Andrew Malone, M.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

Single Cell Analysis of Kidney Transplant Antibody Mediated Rejection

PROJECT SUMMARY/ABSTRACT

Kidney transplantation offers the greatest survival advantage to patients with end stage kidney disease and is vastly more cost effective than dialysis. Long-term survival of kidney transplants has not improved in recent decades. Antibody mediated rejection (AMR) has been identified as a major cause of transplant failure. Currently, management of patients with AMR is inconsistent among centers and frequently fails. We hypothesize that the cell types and cell states unique to AMR can be resolved by single cell RNA sequencing (scRNA-seq) of biopsy samples taken from patients at the time of diagnosis. This approach uses an unsupervised framework for dissecting transcriptional heterogeneity within complex tissues such as the kidney. This allows for the interrogation of cell states and subpopulations using an unbiased clustering approach that is independent of previous knowledge and can provide unprecedented resolution. To test our hypothesis we propose the following aims: In Aim 1, we will perform scRNA-seq of 40 research biopsy cores (20 AMR and 20 non-AMR). From this data we will identify genes expressed in endothelial cells and antibody secreting cells that define AMR specific phenotypes. We have demonstrated feasibility of this approach by generating single cell data from human biopsy samples as outlined in the research plan. For Aim 2, we will use immunohistochemistry to validate these AMR specific markers on a set of independent tissue samples. For Aim 3, we will use publicly available Affymetrix microarray datasets from kidney transplant biopsies with associated outcomes data to determine which cell types are associated with allograft outcome. This proposal logically builds on the principal investigator’s previous research experience and clinical training. To date he has been working full time in clinical transplantation and continuing his research endeavors on an ‘out of hours’ basis. Despite this his research output and experience continue to grow and he has recently published a report on the first successful application of this technology to human kidney biopsy tissue (co-first author). This proposal now focuses on expanding his scientific skills by attaining additional knowledge and practical research experience in single cell methods, bioinformatics and immunology. The career development goals will be achieved through a multi-faceted approach involving mentoring by Dr. Benjamin Humphreys and an advisory committee consisting of Drs. Barbara Murphy (transplant genomics), Phil Payne (biomedical informatics and translational science), Rob Mitra (single cell applications) and Paul Allen (translational immunology), didactic coursework, scientific investigation, and training in scientific communication and research ethics. This work will take place in Washington University which has a rich history of mentoring successful physician-scientists. Successful completion of this career development award will result in a better understanding of AMR, result in the principal investigator’s transition to an independent physician-scientist, and provide a solid foundation from which he will apply for RO1-level funding. Laura Mariani, M.D.

UNIVERSITY OF MICHIGAN AT ANN ARBOR, ANN ARBOR, MI

Integrative Molecular Epidemiology Approach to Identify Nephrotic Syndrome Subgroups

ABSTRACT

Although patients with Nephrotic Syndrome (NS) present with shared clinical signs and symptoms (proteinuria, hypoalbuminemia, hyperlipidemia and edema), there is dramatic variability in prognosis and response to therapy, frustrating patients, families and their clinicians. Even within the histopathologic categories used in the current diagnostic approach (e.g. minimal change disease, focal segmental glomerulosclerosis), there is dramatic variability in disease progression and response to therapy, highlighting the underlying biological heterogeneity within the groups. Small studies with broad, clinical patient inclusion criteria have demonstrated that a subset of patients respond well to anti-TNF therapy, but accurate pre-treatment response of those individuals is not possible based on routine clinical parameters. This project will leverage the Nephrotic Syndrome Study Network (NEPTUNE) cohort study, a multi-center prospective study of 600 patients with FSGS, MCD and MN with rich clinical data, kidney biopsy tissue and gene expression profiles. This study will leverage the kidney tissue gene expression data to identify a subgroup of patients with TNF-alpha pathway activation, assess associated clinical outcomes and identify non- molecular predictors of the subgroup. The aims are: Aim 1: To identify a subgroup of Nephrotic Syndrome patients with homogeneous activation of the TNF- alpha transcriptional pathway. Aim 2: To compare molecular subgroups with conventional clinical- pathologic classification in clinical outcome prediction. Aim 3: To identify non-invasive markers (e.g. demographics, blood and urine markers), standard pathology features and novel pathologic biopsy descriptors associated with the TNF-alpha subgroup. To accomplish this project, the applicant will pursue formal training in , genetic epidemiology and bioinformatics. She will be mentored by a multi-disciplinary team with expertise in systems biology, epidemiology and bioinformatics. The long term objective is to improve the clinical care of patients with Nephrotic Syndrome by improved understanding of the underlying biology, identifying novel biomarkers and potential therapeutic targets for future validation in animal models and mechanistic-based interventional clinical trials. Andrea McAlester, Ph.D.

BAYLOR COLLEGE OF MEDICINE, HOUSTON, TX

Lipid dysregulation of immune mediated intestinal epithelial healing

Lipid dysregulation of immune mediated intestinal epithelial healing A single layer of epithelial cells protects us from harmful interactions with the intestinal microbiota and harmful agents we inhale and ingest daily. Unresolved damage to this epithelial layer can lead to the development of chronic inflammatory diseases, including inflammatory bowel disease (IBD). High fat diets (HFD) correspond to increased incidence and severity of many chronic inflammatory diseases. Diets high in fat have been demonstrated to directly induce pro-inflammatory functions in macrophages, promoting chronic inflammation. In the intestine, HFD has been shown to decrease barrier function and promote chronic inflammation. However, it’s impact on intestinal immune function and tissue repair processes is less understood. Our immune system supports the proper functioning of tissue barriers. In response to tissue damage, tissue macrophages induce pro-inflammatory immune functions aiding in protection from pathogens. In the resolution phase of this response, these cells produce anti-inflammatory cytokines to dampen inflammation and promote tissue repair. Perturbations to any aspect of this response can lead to ineffective repair of tissue injury and development of inflammation. A key signal involved in this molecular switch of macrophages from pro- to anti-inflammatory responses is clearance of apoptotic cells, specifically apoptotic neutrophils. Defects in this response have been linked to many auto-immune and chronic inflammatory diseases including lupus, type 1 diabetes, atherosclerosis, COPD and cardiovascular disease. Tissue macrophages are important in intestinal homeostasis and their dysfunction is thought to drive disease pathogenesis in IBD. Further, defective barrier repair is seen in intestinal disease, but it is unclear whether defective apoptotic cell clearance by intestinal macrophages is involved and what mechanisms could modulate this process. To determine the impact of HFD on intestinal tissue repair responses, we utilized short-term HFD feeding in the context of mouse models of intestinal injury. Our data demonstrates that HFD increases susceptibility to colitis with increased weightloss, aberrant epithelial cell proliferation, loss of goblet cells and tight junction proteins necessary to prevent microbial penetration into the body. Further, we identified dysregulated immune responses resulting in an inability of intestinal immune cells to properly support barrier function and repair. We also find that HFD alters macrophage responses to and clearance of apoptotic neutrophils. Together, we find that HFD feeding directly promotes altered functions of intestinal immune cells leading to barrier repair defects. These findings led us to hypothesize that dietary lipids directly interfere with apoptotic cell recognition and uptake receptors and activation of downstream pro-repair signaling pathways resulting in dysregulated intestinal barrier repair. This is the basis of my K01 application. Wendy McKimpson, Ph.D.

COLUMBIA UNIVERSITY HEALTH SCIENCES, NEW YORK, NY

Gut cell plasticity during diabetes treatment

Project Summary

There is irreversible destruction of insulin-secreting pancreatic β-cells during type 1 diabetes. Consequently, individuals with this disease are dependent on continual insulin injections to manage their diabetes which is burdensome to patients. However, if one could trigger a patient's own cells to generate endogenous insulin it would essential cure these individuals of their diabetes. Elimination of the transcription factor Foxo1 in a population of intestinal endocrine progenitor (Neurogenin3+) cells prompts them to secrete functional insulin and reverse diabetes in mice. Interestingly, there are also more Ngn3+ cells in the intestines of these mice. Using a reporter mouse, the candidate has found that Foxo1 is expressed in Ngn3+ and some acid-secreting parietal cells in the stomach epithelium. While ablation of Foxo1 in both of these cell populations triggers the appearance of insulin immune-reactive cells, it also increases Ngn3 expression and parietal cell numbers. The candidate hypothesizes that elimination of Foxo1 in the stomach changes the cell plasticity of this organ. To further understand this process, using three specific aims, this research proposal will 1) study specific changes to gastric enteroendocrine and parietal cells when Foxo1 is deleted in mice and in human cells and 2) define molecular pathways important to insulin-cell conversion. To accomplish this, Foxo1 will be abolished, using cre recombinase technology, in primary stomach cultures (Aim1.1), in two separate populations of cells in mice (Aim1.2), and in human induced pluripotent cells (iPSC) (Aim2). Changes to cell type, distribution, proliferation, and death will be measured using immunohistochemistry and qPCR. Important to career development, new research skills (iPSC culture and genome modification by CRISPR) will be learned in Aim2 of this proposal. Using FACs to isolate cells, Aim3 will analyze Foxo1-deleted primary stomach cells and stomach insulin+ cells using RNA-seq. Unique multi-color fluorescent reporters will be used in both Aim2 and Aim3. Taken together, the proposed experiments will likely reveal new molecular targets/pathways important in the conversion of stomach cells to those that produce insulin, ultimately to discover a new avenue of treatment for type 1 diabetes. These experiments are complimented by a career development plan in which the candidate will learn new research techniques, broaden her scientific network through attending conferences, and develop her writing skills (through coursework, submitting manuscripts/R01 application) so that she is poised to become an independent investigator. Both the career development plan and research proposal will be completed at Columbia University, a prestigious institution. Joanna Melia, M.D.

JOHNS HOPKINS UNIVERSITY, BALTIMORE, MD

Genetic variation in the metal transporter ZIP8 and impact on the innate immune response in the gut

PROJECT ABSTRACT

Inflammatory bowel diseases (IBD), including Crohn’s disease (CD) and ulcerative colitis, affect 1 in 200 Americans. IBD arises through a complex interplay of genetic, immune, microbial, and environmental factors disrupting intestinal homeostasis. There is a strong call from patients to study the role of diet-derived micronutrients in intestinal homeostasis. Recent genetic studies have focused attention on ZIP8 and the role of metal transport in CD through the discovery of an association between CD and a nonsynonymous single polymorphism (SNP, rs13107325; Ala391Thr) in SLC39A8, the gene encoding ZIP8. This SNP has also been associated with six other diseases, including obesity and schizophrenia. Further, individuals with ZIP8 A391T shared a common fecal dysbiosis independent of disease diagnosis. The function of ZIP8 in the gut is not known, but in other cell types, ZIP8-mediated transport negatively regulates NF-κB signaling by complexing with IKKβ and ZIP8-mediated manganese transport balances arginine metabolism away from nitric oxide synthase to reduce oxidative stress. Our preliminary data demonstrate that ZIP8 is increased in the inflamed terminal ileum of patients with CD and ZIP8 A391T impairs negative regulation of NF-κB signaling with reduced zinc transport. We have established two model systems for the studies proposed in this application: (i) ZIP8- knockdown in human ileal enteroids and (ii) a novel knock-in mouse with ZIP8 A393T, the mouse equivalent of the human variant. We hypothesize ZIP8-mediated metal transport regulates the innate immune response in intestinal epithelial cells, and this function is changed by ZIP8 A391T to promote CD pathogenesis. The aims of this project are (1) To establish the role of ZIP8 in intestinal epithelial cells in the innate immune response, (2) To study the effect of the CD-associated genetic variation in ZIP8 (A391T) on ZIP8 function in intestinal epithelial cells, and (3) To study the effect of the CD-associated genetic variation in ZIP8 on colitis susceptibility in a novel knock-in mouse model (ZIP8 A393T). The candidate is an Assistant Professor of Medicine at the Johns Hopkins School of Medicine in the Division of Gastroenterology with research and clinical training dedicated to IBD. The goal for this applicant is to use this project to enhance her molecular biology and immunology expertise and position her to build an independent career as a physician- scientist dedicated to studying the role of micronutrients in the pathophysiology of IBD. In addition to hands-on training and didactic education, the Training Plan includes strong mentorship from a Scientific Advisory Committee with diverse expertise in epithelial biology, metal biology, NF-κB signaling, oxidative stress, host- microbiota interactions and IBD, complemented by the strong support of the Hopkins Conte Digestive Diseases Basic and Translational Research Core Center and the institution. Kristin Meliambro, M.D.

ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, NEW YORK, NY

The Role of KIBRA Signaling in Podocyte Injury

Project Summary:

Despite the acceptance of the glomerular podocyte as the target cell for injury in proteinuric kidney disease, cell-specific therapy remains absent in clinical nephrology. A critical barrier is the limited understanding of the mechanisms of podocyte injury during disease. Our preliminary data has identified KIBRA (KIdney BRAin protein) as a potential mediator of podocyte injury. KIBRA's expression is increased in human disease, and silencing of KIBRA in podocytes is protective in vitro and in vivo. We have demonstrated that KIBRA inhibits the signaling of the Hippo pathway effector Yes-associated protein (YAP) and disrupts normal actin cytoskeletal dynamics. KIBRA expression was also increased in Tgfbr1 transgenic mice and in CD2AP knockdown podocytes. Our central hypothesis is that TGF- β/Smad signaling leads to upregulation of KIBRA in podocytes, resulting in podocyte injury. Additionally, as a disease correlate, we hypothesize that KIBRA overexpression will be sufficient to induce and promote glomerular disease progression, while KIBRA deletion in models of chronic glomerular disease will be protective. The rationale for the proposed research is that defining the role of KIBRA in glomerular disease progression will increase understanding of the mechanisms of podocyte injury and advance the quest for targeted therapeutics. Our hypothesis will be tested by three Specific Aims: Aim 1 will define the upstream regulation of increased KIBRA expression in podocytes. Aim 2 will determine whether KIBRA overexpression in podocytes promotes glomerular disease progression in vivo. Aim 3 will determine if KIBRA deletion reduces podocyte injury in chronic glomerular disease. Candidate and Training: The primary objective of this application is to support Dr. Kristin Meliambro's career development into an independent basic scientist in the fields of podocyte cell biology and glomerular diseases. Dr. Meliambro's proposed training activities are in four areas: 1) animal models of glomerular diseases, acute kidney injury (AKI), and chronic kidney disease (CKD); 2) podocyte cell biology and cell signaling, with a focus on the Hippo signaling pathway; 3) advanced imaging techniques; 4) scientific writing and oratory skills. To achieve this, she has assembled a mentoring and advisory team led by Dr. John Cijiang He, Chief of the Division of Nephrology, and Dr. Kirk Campbell, Associate Professor and Director of the Nephrology Fellowship Program at the Icahn School of Medicine at Mount Sinai (ISMMS). Both Drs. He and Campbell are former K08 awardees with combined four R01 grants between them who have expertise in the field of podocyte cell biology. Environment: The ISMMS is an international leader in medical and scientific training, iomedical research, and patient care. Research is a top priority, as ISMMS is ranked 13th among U.S. medical schools for NIH funding by US News and World Report and 2nd in research dollars per principal investigator by the Association of American Medical Colleges (AAMC). The Division of Nephrology at ISMMS is an international leader in research, particularly in the area of podocyte cell biology. DAVID M. MERRICK, Ph.D., M.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

Identification of a mesenchymal progenitor cell hierarchy in adipose tissue

PROJECT TITLE

Characterization of a novel adipogenic mesenchymal progenitor population inhabiting a unique interstitial tissue niche.

ABSTRACT

Obesity and the associated metabolic syndrome represent a profound public health challenge for which there are few effective therapeutics. Fundamentally, obesity arises in the setting of nutrient excess, which stimulates adipose tissue expansion. The healthy growth of adipose tissue depends on the capacity of progenitor cells to undergo de novo adipogenesis. However, the cellular hierarchy and mechanisms governing adipocyte progenitor differentiation are poorly understood. Adipose mesenchymal progenitors represent a complex pool of highly diverse cell types, and previous attempts to characterize these populations using candidate gene approaches have been significantly limited by lack of specificity and loss of spatial information. Using unbiased single-cell RNA transcriptomics coupled with immunohistochemistry, we identified a novel progenitor population which we term Interstitial Progenitor Cells (IPCs). IPCs are multipotent progenitors, marked by expression of Dpp4 and Wnt2, which can give rise to committed preadipocytes and mature adipocytes. Importantly, we show that IPCs inhabit the Reticular Interstitium (RI), a fibrous tissue that envelops many organs including adipose depots and represents a previously unrecognized anatomical niche for multipotent mesenchymal progenitors. The scientific objectives of this proposal are to determine the in vivo contribution of IPCs to adipose tissue biology and define the niche elements that maintain IPC identity and direct lineage allocation. The Aims of the proposal are: 1) Trace the in vivo lineage allocation of IPCs during early adipose organogenesis and adult stimulated adipogenesis. 2) Investigate the role of TGFβ and Wnt signaling in IPC maintenance and differentiation. In addition to the scientific contributions of the proposed aims, this proposal outlines a structured, focused training plan that will equip me with the techniques and expertise in progenitor cell biology that will serve as the foundation of a successful transition to independence.

Evanna Mills, Ph.D.

DANA-FARBER CANCER INST, BOSTON, MA

Mechanistic characterization of the anti-inflammatory function of thermogenic fat for the treatment of metabolic disorders

PROJECT SUMMARY:

Obesity and associated “meta-inflammation” are major risk factors for type-2 diabetes, cardiovascular disease, non-alcoholic fatty liver disease (NAFLD) and cancers. Visceral “white” adiposity drives these deadly sequelae of obesity. In contrast, thermogenic “brown” and “beige” adipose can uniquely catabolize stored fat, and are potently anti-obesogenic. The anti-obesity activity of thermogenic adipocytes requires activation by peripheral signals, and the identification of these signals is key to leveraging the therapeutic potential of these cells. Such thermogenic adipose is also thought to be anti- inflammatory but the mechanistic basis of this is unclear. Understanding the mechanisms that promote this anti-inflammatory function of thermogenic fat would have major therapeutic potential. Thermogenic fat is rich in mitochondria which produce a vast array of metabolites, many of which possess signaling capacity. Interestingly the tricarboxylic acid intermediate succinate is an important signaling molecule that controls both thermogenic adipose function and inflammation. I identified that thermogenic fat has a unique capacity to sequester succinate from the extracellular milieu (through an undefined mechanism) to promote thermogenesis; while immune cells express a G-protein coupled receptor (termed succinate receptor 1), the ligation of which is potently pro-inflammatory. I hypothesize that activation of thermogenic adipose tissue will promote an anti-inflammatory phenotype by altering the levels of important signaling metabolites, such as succinate, that are known to regulate immune cell function. Moreover, I predict manipulation of these signals will aid in the treatment of metabolic disease. Objectives: 1) What is the succinate transport mechanism in thermogenic fat? 2) Can thermogenic fat limit circulating succinate and inflammation in obesity-driven pathologies? 3) Can we manipulate thermogenic fat to modify the pathology of NAFLD? This work will use both in vitro primary brown fat cell culture and in vivo models of visceral adipose expansion and thermogenesis. This will be coupled with liquid-chromatography/mass spectrometry (LCMS), genetic manipulation, and pharmacological methods to clarify the causality between thermogenic fat and inflammatory signaling. My research experience has afforded me with a skill set that is unique in the world, that will allow me to study the metabolic cross-talk between the adipose and immune systems. Working with Drs. Chouchani and Spiegelman, experts in the fields of thermogenesis and metabolism, I will become proficient in the study and manipulation of adipocyte biology and LCMS analysis and these skills will be coupled with my strong background in immunology. The diverse and rich scientific environment at DFCI adds fuel to my enthusiasm to establish myself as an independent investigator and drives my determination to ensure that I make a success of it. DENNIS G. MOLEDINA, Ph.D., M.D.

YALE UNIVERSITY, NEW HAVEN, CT

Identification of Non-Invasive Biomarkers and Indices for Diagnosis of Drug-Induced Acute Interstitial Nephritis

Candidate: The candidate, Dr. Dennis G. Moledina, is a board-certified nephrologist at the Yale School of Medicine. The proposed research builds on his past work on evaluating novel biomarkers to phenotype human acute kidney injury. After completing a clinical fellowship in nephrology at Yale, he received three additional years of training in methods of clinical translational research. He is a PhD candidate with Yale’s Investigative Medicine Program, which provides research training to aspiring physician-scientists. Through this program, he attended didactic coursework on clinical research methodology, biostatistics, and immunobiology. During this award, the candidate will develop additional skills that are required to achieve his long-term goal of becoming an academic translational physician-scientist studying immune-mediated kidney diseases. These skill areas will be developed through hands-on, mentored research training and advanced didactic coursework. The candidate has strong institutional commitment from Yale including assured transition to a faculty position. He will conduct the proposed research under the mentorship of Dr. Chirag R. Parikh, who is a world-renowned clinical investigator with expertise in biomarker research in acute kidney injury. Additional, he will receive guidance from a highly-qualified mentorship committee at Yale. Project: Drug-induced acute interstitial nephritis (AIN) results from immune-mediated kidney injury, which is triggered by commonly used drugs. Patients with AIN may escape clinical attention because they have a subtle clinical presentation with minimal symptoms and subacute loss of renal function. Moreover, since there is no noninvasive diagnostic test for this disease, its diagnosis requires a kidney biopsy, which carries risks. As a result, many cases of AIN remain undiagnosed, which leads to permanent kidney damage and chronic kidney disease. The overall goal of this proposal is to improve the ability to non-invasively diagnose AIN by identifying biomarkers and developing indices. The candidate hypothesizes that AIN is a delayed hypersensitivity reaction mediated by type 1 and 2 T- helper cells (Th1/Th2), and predicts that the characteristic inflammatory mediators produced by these cells, specifically interferon-γ, tumor necrosis factor-α, and interleukin(IL)-2 (Th1), and IL-4, IL-5, IL-9, and IL-13 (Th2), will be higher in the plasma and/or urine of AIN participants as compared with study participants without AIN. In aim 1, the candidate will identify biomarkers that distinguish AIN from other causes of acute loss of renal function. In aim 2, the candidate will develop two diagnostic indices for AIN; the first will use currently available clinical and laboratory variables and the second will combine currently available variables with novel biomarkers (from aim 1). These indices will provide probability of AIN diagnosis without requiring a kidney biopsy. In aim 3, the candidate will validate the biomarkers and indices from aims 1 and 2 in three, external, biopsy-based cohorts. These findings will improve patient outcomes through timely diagnosis and intervention, and guide biomarker-based enrollment in future clinical trials of intervention(s) for AIN.

Lindsey Muir, Ph.D.

UNIVERSITY OF MICHIGAN AT ANN ARBOR, ANN ARBOR, MI

Myeloid lineage activation and reprogramming in metabolic dysfunction

PROJECT SUMMARY

The high prevalence and health impact of obesity drives a critical need to understand the link between obesity and disease. Monocyte and macrophage activation that contributes to low grade inflammation is one such link. These myeloid cells contribute to inflammation by producing proinflammatory cytokines, activating other immune cells, and monocytes can also differentiate into proinflammatory macrophages, all of which are associated with insulin resistance. However, the mechanisms underlying myeloid cell proinflammatory activation in human obesity and diabetes are not resolved. My preliminary data identify lysosomal dysfunction in SRhi myeloid cells as high priority targets for investigation in diabetes. Furthermore, strategies to alleviate inflammation in metabolic disease have been ineffective, partly due to low cellular specificity. We identify polymer nanoparticles (NPs) as excellent candidates for a more specific approach. My preliminary data show increased NP-monocyte interactions in obese diabetic patients vs. non-diabetic, making NPs a promising approach for targeting myeloid cells in diabetes. My central hypothesis is that obesity disrupts SRhi myeloid cell lysosomal processing, increasing production of inflammatory mediators, and that NPs can modulate SRhi myeloid cell inflammatory activation. To examine this hypothesis, I will use monocytes and ATMs from a valuable obese bariatric surgery cohort, focusing on diabetic vs. non-diabetic comparisons in two research aims: (1) Identify the genes and pathways by which myeloid cells are dysregulated in human obesity. I will use a combination of powerful, unbiased high-throughput genomics platforms with novel bioinformatics tools to identify specific pathways and regulators in SRhi myeloid cells from diabetic patients. I will use assays of endocytosis and lysosomal function, proinflammatory cytokines, and flow cytometry to determine functional changes in SRhi myeloid cell subtypes in diabetes. (2) Determine the specificity and efficacy of NPs for myeloid cell modulation in human obesity. I will test internalization and impact of NPs on human SRhi myeloid cell subtypes, determining whether they can attenuate proinflammatory signatures through cytokine assays and RNA-seq. By completing these aims I will identify the molecular and cellular signatures mediating activation of SRhi myeloid cells and determine efficacy of novel NP-therapeutics to modulate SRhi myeloid cells in human metabolic disease. I will gain expertise in nanotechnology and high-throughput genomics platforms. The mentorship team will be led by co-primary mentors Dr. Robert O'Rourke and Dr. Lonnie Shea. Dr. O'Rourke is an expert in human obesity and clinical biosamples. Dr. Shea is an internationally recognized researcher at the interface of regenerative medicine, drug and gene delivery, and immune tolerance. Co-primary mentors and experts in immunometabolism, diabetes, obesity, and genomics and bioinformatics will guide me in completing the research and training proposed. Completing these goals will be a critical step toward independent research in translational immunology and immunometabolism. Naoka Murakami, Ph.D., M.D.

BRIGHAM AND WOMEN'S HOSPITAL, BOSTON, MA

Protection of kidney from autoimmunity by modulating co-stimlatory signaling

Project Summary

Kidney inflammation occurs in response to ischemic injury, infections, and activation of autoreactive or alloreactive lymphocytes and contributes to chronic kidney damage, fibrosis and end-stage kidney diseases. Adaptive immune responses are tightly controlled in the kidney to prevent excessive inflammation and to maintain tolerance against self-antigens. However, the fundamental understanding of the mechanisms supporting immune regulation in the kidney is incomplete. Clinical observations suggest that the co-stimulatory molecules such as PD-1 and CTLA4 play pivotal roles in regulating immune responses in the kidney, as observed in acute interstitial nephritis or kidney allograft rejection in patients treated with immune checkpoint inhibitors. Studies have elucidated the contribution of adaptive immunity in kidney inflammation. However, the mechanistic study on the roles of antigen- specific T cells in kidney inflammation is far from complete due to the lack of appropriate animal models to precisely track antigen specificity and this hinders development of specific targeted therapy in autoimmune kidney diseases. To close this knowledge gap, we developed two new transgenic mouse models in which the expression of self-antigens can be specifically induced in proximal tubules or podocytes, the main target anatomical segments in the autoimmune kidney diseases. By combining these animal models with a tetramer-based antigen-specific T cell tracking technique to detect endogenous T cells that recognize specific peptide-MHC complexes, we are able to analyze their phenotype and function in vivo. The overall goal of this project is to dissect the mechanisms of tolerance against kidney-restricted antigens. Our preliminary data indicated that kidney-specific expression of self- antigens induced tolerance against these antigens; however, administration of anti-PD-1 and anti-CTLA4 could overcome the tolerance, manifested as infiltration of antigen-specific T cells into kidney interstitium. We hypothesize that the tolerance in the kidney is regulated by the co-stimulatory molecules expressed in antigen-specific T cells and their ligands found in kidney parenchymal cells or antigen presenting cells; and that disruption of the tolerance would lead to maladaptive inflammation in the kidney. To test this hypothesis, we will investigate T cell tolerance mechanism at steady state (Aim 1), characterize kidney-infiltrating pathogenic T cells (Aim 2), and dissect the roles of antigen presenting cells in tolerance and autoimmunity (Aim 3). These models provide a novel and innovative approach to study antigen-specific adaptive immune response in autoimmune kidney disease and represent unique preclinical tools, which will lead to identification of novel therapeutic targets. In addition, after completing this K award, I will establish my own translational research project in the intersection of immunology and kidney biology. Developing this research project will be an exceptional opportunity to become proficient in specific basic research methodologies (e.g. transcriptomics) and in kidney biology research. Sreejith Nair, Ph.D.

UNIVERSITY OF CALIFORNIA, SAN DIEGO, LA JOLLA, CA

Enhancer Codes with Ligand Mediated Gene Regulation and Chromatin Architecture

PROJECT SUMMARY

The central objective of this research proposal is to gain novel insights into the cell biological principles underlying the regulation of gene expression in response to hormonal stimulation. My research focus is gene regulation mediated by transcriptional enhancers. Although recognized as the major organizer of gene regulation in metazoans, the precise mechanism with which enhancer complex assembled and regulate target genes in response to stimuli is still mostly not known. Preliminary data described in this proposal suggest that acutely assembled enhancers harbor ribonucleoprotein complex with a physicochemical property that is quite distinct from that of enhancers that are constitutively active. These enhancers get optimally activated once associated with inter-chromatin granules (ICGs). This proposal will link multiple experimental strategies from disciplines as diverse as molecular biology, biophysics, genomics, proteomics and microscopy to provide a comprehensive understanding of the underlying mechanism of inducible enhancer activation and chromosomal architecture. Together, this approach can significantly alter our current concepts about regulated gene transcriptional programs. Importantly, the finding and experimental skills acquired from this study will also allow the transition to an academic career as an independent investigator studying the biophysical and molecular mechanisms of ligand regulated transcriptional program using proteomic, bioinformatics and genomic approaches. Charles Neff, Ph.D.

UNIVERSITY OF COLORADO DENVER, Aurora, CO

Interactions of Intestinal Epithelial Cells, The Innate Immune System and Gut Microbiota During Dysbiosis

PROJECT SUMMARY/ABSTRACT

Candidate: The applicant is an instructor in the Allergy and Clinical Immunology Division at the University of Colorado Anschutz Medical Campus. The applicant’s thesis work under the mentorship of Dr. Ramesh Akkina at Colorado State University focused on translational research utilizing humanized mice to develop novel therapies against HIV infection. During his Post-doctoral training, Dr. Neff developed methods to characterize the im pact of the fecal m icrobiom e on adaptive im m une cells, found regulatory T cell inducing products in bacteria and characterized the impact of HIV infection on immune cells in the lung. Environment: At the University of Colorado, the applicant has a mentoring team with immunology, mucosal immunology, inflammation and microbiome expertise in primary mentor Dr. Brent Palmer, and co-mentors Drs. Catherine Lozupone, Sean Colgan, and Laurel Lenz. The applicant will continue close collaboration with Drs. Nichole Reisdorph and Timothy Bushnell who will serve as technical advisors for UPLC, mass spectrometry and CyTOF. The applicant’s training plan during the award includes bioinformatics, immunology, biostatistics, responsible conduct of research, and programs in grant writing and career development. Research: Chronic immune activation is a hallmark of HIV infection and recently, changes in the composition of the enteric microbiome have been implicated as a driving factor. However, the mechanisms of host:microbiome interactions are poorly understood. We showed that the fecal microbiome from those with HIV has inflammatory properties not seen in taxonomically similar healthy subjects. Using this HIV-associated microbiome as a model present a unique opportunity to identify causal bacteria and the mechanisms in which they lead to cell activation. The applicant’s long-term goal is to develop a deeper understanding of the bacterial products that drive chronic immune activation as a means to advance treatment strategies for inflammatory gastrointestinal diseases. This proposal will evaluate the mechanisms of microbiota:host interactions. Based on previous findings, which indicated peptidoglycan (PGN) and lipopollysaccharide (LPS) as drivers of infammatory responses, this proposal will also investigate if specific variants of PGN and LPS differentally induce aberrant activation. The central hypothesis is that specific PGN and LPS forms found with bacteria increased during HIV convey differential inflammatory properties. The first aim will characterize the interactions of epithelial cells, subsets of innate immune cells and the microbiome during HIV dysbiosis in order to determine rare bacteria that cause inflammation. The second aim will focus on purifying PGN and LPS to determine if strain specific forms of these bacterial products are responsible for inducing aberrant cell activation. The findings here could have global implications of the treatment of chronic immune activation seen in multiple diseases. Ravi Nistala, M.S., M.D.

UNIVERSITY OF MISSOURI-COLUMBIA, COLUMBIA, MO

Role of DPP4 in Kidney Inflammation and Injury

Project Summary/Abstract

The incidence of chronic kidney disease (CKD) is increasing due to the ever-expanding number of people who are either obese (33%) or diabetic (9%) or both. In a significant proportion of obese-diabetic subjects, CKD may progress to end stage renal disease (ESRD) and dialysis faster than non-obese diabetic subjects. Care of ESRD patients (1% of Medicare population) consumes Medicare dollars in a disproportional manner (7%). Hence, preventing progression of CKD to ESRD is of paramount importance. Currently, the standard of care is to use medications that block/suppress activation of the renin-angiotensin system. However, these medications are not enough to stop progression to ESRD in many subjects and novel therapeutic agents are needed. DPP4 inhibitors are conveniently placed at the intersection of diabetes and CKD due to recent data showing improvement in proteinuria in diabetic patients with CKD (SAVOR-TIMI trial and animal models). Dr. Nistala and others have shown that DPP4 inhibitors may have kidney specific effects independent of blood pressure or blood sugar improvement. However, the mechanisms of DPP4 inhibitor-mediated benefits in the kidney are relatively unknown. If DPP4 inhibitors are to be used for preventing progression of kidney disease, the biological function of DPP4 in the kidney needs better understanding. Data supports the notion that DPP4 may regulate sodium absorption and albumin/oligomeric peptide endocytosis in proximal tubules of the kidney. In addition, DPP4 is a co-stimulatory molecule for T-lymphocytes, activates mononuclear cells (including macrophages and antigen presenting cells [APCs]) and may mediate inflammation based on findings from fat depots in obese and plasma from diabetes patients. Tubulointerstitial inflammation and fibrosis plays an important role in the progression of CKD in the setting of obesity and diabetes and importantly, Dr. Nistala found that DPP4 activity is increased in the kidney and expression levels are higher in the proximal tubules of obese/diabetic mice. Therefore, this project is focused on examining the role of DPP4 in the proximal tubule and kidney immune system in the setting of obesity/diabetes. The experiments are designed to address the central hypothesis that obesity- induced proximal tubule DPP4 activation incites a pro-inflammatory immune response leading to tubulointerstitial fibrosis and progression of kidney disease. To address this hypothesis, DPP4 will be activated via high sucrose/high fat (Western Diet) feeding in mice with or without DPP4 deficiency. In Aim 1, the role of Western diet in activation of DPP4 in the proximal tubule and tubular dysfunction will be studied by using proximal tubule specific deletion of DPP4. In Aim 2, the role of proximal tubule DPP4 in kidney immune system activation will be examined. In Aim 3, the role of proximal tubule DPP4-induced immune system activation in tubulointerstitial fibrosis and kidney disease progression will be studied. It is anticipated that results from this project will yield unique insights into the mechanisms of kidney injury and inflammation in obesity/diabetes with the ultimate goal of translating these findings into meaningful treatments. Emily Noble, Ph.D.

UNIVERSITY OF GEORGIA, ATHENS, GA

Melanin-concentrating hormone and the neural regulation of feeding

Project Summary/Abstract

Obesity and its associated comorbidities pose a major burden to public health, and existing therapeutics are only minimally effective or have major adverse consequences. The brain plays a central role in mediating energy balance. Melanin-concentrating hormone (MCH), a neuropeptide produced primarily in the lateral hypothalamic area (LHA) increases energy intake and body weight gain, and therefore there is recent interest in developing obesity therapeutics that block the hyperphagic effects of MCH. Preliminary data herein suggest that in addition to generally increasing food intake, MCH contributes to excessive feeding via conditioned appetitive mechanisms, such as food impulsivity and conditioned place preference for palatable food. Furthermore, these mechanisms can be attributed to specific nodes of MCH neuronal circuitry such as the ventral hippocampus (vHP), a region that contains high levels of MCH receptor but where the function of MCH with regard to feeding behavior has not previously been investigated. Through studying the MCH system and its contributions to the central regulation of ingestive behavior, the principal goal of the proposed 5-year research career development plan is to facilitate the applicant's transition from Research Assistant Professor to Tenure-track Assistant Professor with independent R01 funding. The proposed research will enable the applicant to master several virus- based neuroanatomical and chemogenetic techniques, rodent behavioral paradigms, high- resolution whole brain mapping with functional connectivity analyses. Using these techniques, she will determine the role of MCH to vHP neuronal circuitry in mediating feeding behavior, the relevance of this pathway to obesity, and will identify collateral projections, functional connections, and downstream targets. Specific Aim 1 utilizes dual virus chemogenetic techniques to selectively activate MCH neurons that project to the vHP in order to study the role of vHPà MCH neuronal circuitry in mediating several aspects of feeding behavior. Specific Aim 2 uses viral-based neural tracing, immunohistochemistry and hybridization to further characterize these neurons by their collateral projections and phenotypic gene/protein expression profiles. Aim 3 uses the novel combination of dual virus chemogenetics with whole brain perfusion mapping to determine downstream regions engaged by activating MCH neurons that project to the vHP. Additionally, experiments in Aim 3 will determine how MCH to vHP neuronal circuitry augments regional brain activity during food impulsivity. This novel combination of approaches allows for investigation of the interaction between activating specific nodes of a defined circuit and immediate behavioral effects. Results from these experiments will be generative as pilot data for an R01 submission by the applicant, will provide significant contributions to ingestive behavior research, and will overall provide the applicant with a unique set of skills for her independent research career. Amy OConnell, Ph.D., M.D.

UNIVERSITY OF FLORIDA, GAINESVILLE, FL

Evaluating canine diabetes as a novel means to understand the human disease

PROJECT SUMMARY

Type 1 diabetes (T1D) is a disease of increasing prevalence worldwide. Despite decades of research, the specific underlying causes and consistently effective methods to prevent or cure the disease have not been identified. Much of this research has involved rodent models of the disease, particularly the non- obese diabetic mouse; however it has become evident that this model has substantial limitations. A large animal model more reflective of the human disease could provide important translational discoveries in the areas of pathogenesis, prevention, and therapy of T1D. Canine diabetes is similar in many ways to human T1D; however, in-depth studies characterizing the phenotypic and etiopathologic characteristics are lacking. The genetic structure of dog breeds, living environment that is similar to humans, and disease heterogeneity also contribute to the attractiveness of the dog as a potential disease model. The overall objective of this proposal is to examine the etiopathogenesis of canine diabetes to determine the relevance of these animals as a naturally occurring model of human T1D. This objective will be evaluated via the following specific aims: 1) Define the phenotypic characteristics of canine diabetes from initial onset to established disease, 2) Evaluate the role of autoimmunity in the pathogenesis of canine diabetes, and 3) Identify potential biomarkers of the pre-diabetic state in dogs. To accomplish these aims, newly diagnosed diabetic dogs will be recruited to evaluate β cell function, metabolomic profiles, and to detect evidence of autoimmunity using novel methodology. Additionally, a cohort of non-diabetic dogs will be followed and monitored for the development of diabetes to search for biomarkers of a pre-diabetic state. The principle investigator, Dr. Allison O’Kell, is a veterinarian with board certification in small animal internal medicine and is an Assistant Professor at the University of Florida College of Veterinary Medicine. Together with her mentor’s laboratory, she has performed preliminary studies supportive of the aims of this proposal. This career development award will provide Dr. O’Kell with the additional training needed to achieve her goal of becoming an independent investigator, which includes training in the areas of: 1) prospective clinical study design and implementation, 2) performance, interpretation, and evaluation of various laboratory techniques and diagnostic tests, 3) statistical methods and interpretation, 4) collaboration with a multi- disciplinary and multi-institutional team, and 5) grant and manuscript writing. Dr. O’Kell has developed a comprehensive program including didactic training, practical and laboratory experience, and a strong mentoring and advisory team. The University of Florida Diabetes Institute and College of Veterinary Medicine together will provide a collegial and supportive environment with the equipment, laboratory space, and resources necessary to complete the research and training. At the culmination of this proposal, Dr. O’Kell will be poised to achieve independence as a researcher with continuing investigation in the field of diabetes. Allison O'Kell, M.S., D.V.M.

BOSTON CHILDREN'S HOSPITAL, BOSTON, MA

The Role of Wnt2b in Intestinal Health

Project Summary

Wnt signaling has been shown to be critical for intestinal health, and Wnts are required for intestinal stem cell (ISC) homeostasis. The roles of individuals Wnts in ISC regulation remain poorly defined. We have recently described a human congenital diarrheal disorder caused by deficiency of Wnt2b, marked by intestinal inflammation and loss of Lgr5+ISC, showing that Wnt2b is critical in the intestine. The goal of this proposed project is to clarify the need for Wnt2b in intestinal health. In Aim 1 we will investigate whether there is a developmental requirement for Wnt2b using mouse and human developmental models. In Aim 2 we will examine the impact of human Wnt2b on Lgr5+ISC homeostasis using a more mature enteroid model. We will also consider the mesenchymal versus epithelial sources of Wnt2b as components of Aims 1 and 2. This proposal is a five-year, mentored career development plan focused on expanding the investigator's knowledge of mucosal development and intestinal stem cell biology, as well as providing in depth instruction on genome editing and organogenesis techniques. The candidate is an MD/PhD trained physician scientist who is also an allergist/immunologist and neonatologist, and currently practices as a neonatologist at Boston Children's Hospital. The proposal will build on her strong immunology background by adding new expertise in stem cell biology and translational investigation through the tutelage of her co-mentors, Dr. Pankaj Agrawal and Dr. David Breault at the Manton Center for Orphan Disease Research and the Harvard Digestive Disease Center of Boston Children's Hospital. The proposed work will result in the candidate building niche expertise on regulators of intestinal development, upon which she plans to build her independent research career. Sarah Panzer, M.D.

UNIVERSITY OF WISCONSIN-MADISON, MADISON, WI

Improving long-term allograft survival in kidney transplantation by targeting B cell survival cytokines

PROJECT SUMMARY/ABSTRACT

The proposal presents a four-year career development program designed to provide Sarah Panzer, MD, an Assistant Professor at the University of Wisconsin-Madison, the career development and research experiences necessary to become an independent physician scientist to improve patient and allograft survival of kidney transplant recipients. Approximately 50% of transplanted kidneys fail within 10 years of transplantation. The major pathologic diagnosis in failing kidney allografts is transplant glomerulopathy (TG). While TG has largely been attributed to chronic antibody-mediated rejection (cABMR), the key mediators of this process remain unknown and no effective therapeutics exist. In a multitude of B cell mediated diseases, the B cell survival cytokines have been demonstrated to exacerbate disease activity and represent a viable therapeutic target. In kidney transplantation, preliminary studies have found elevated levels of B cell survival cytokines in the serum and kidney tissue of patients with cABMR. The hypothesis of this proposal is B cell survival cytokines play pivotal roles in promoting the development of TG and predict allograft failure. This proposal will test this hypothesis via 1) deficiency of B cell cytokines in an animal kidney transplant model to attenuate TG, 2) determination of the ability of intragraft B cell cytokines to predict allograft failure in prevalent TG patients, and 3) determination of B cell survival cytokines as a risk factor for incident TG in a prospective observational cohort of high-risk kidney transplant patients. These data will support future R01 funded study to investigate B cell survival cytokines in TG as a therapeutic target. As a junior faculty member at an institution with extensive infrastructure to support early stage investigators and a highly active transplant center, Dr. Panzer is in an ideal environment to complete the proposed research and pursue advanced training. Her career development plan includes both coursework and mentored research training in the areas of clinical study design, subject recruitment and retention, and survival analysis. To ensure success, she has identified committed, expert mentors and secured protected time for this work. This award addresses a significant gap in the field of kidney transplantation while affording the mentored research experience critical for Dr. Panzer to become a successful physician scientist leading a program to improve outcomes in kidney transplantation. Michael Patnode, Ph.D.

UNIVERSITY OF CALIFORNIA SANTA CRUZ, SANTA CRUZ, CA

Metabolic And Spatial Competition For Dietary Fiber Between Commensal And Pathogenic Gut Microbes

PROJECT SUMMARY

The microbial community that resides in the human intestine profoundly influences host metabolism, immune homeostasis, and the outcome of enteric infections. Dietary fiber is a promising tool for manipulating the gut microbiota to promote that provide beneficial functions to the host. Though, it is currently difficult to predict which gut bacterial species will respond to fiber-based dietary interventions, interspecies competition makes it possible to precisely target beneficial species of interest using a particular fiber type. Bacterial species with pathogenic potential, such as uropathogenic E. coli (UPEC), are present in the gut microbiota of asymptomatic individuals and these species have the capacity to expand in response to fiber. Exploiting competition between pathogens and their non- pathogenic relatives to reduce pathogen load in the gut will require detailed knowledge of the genes underlying these species’ overlapping nutrient harvesting strategies, including genes mediating adhesion to nutrient-rich diet-derived particles. The following aims will test the hypotheses that (i) expansion of commensal E. coli in the gut in response to dietary fiber can reduce the fitness of pathogenic E. coli, and that (ii) commensal and pathogenic bacterial species compete for adhesion to the same diet-derived surfaces in the intestinal lumen. In Aim1, I will identify dietary fibers that selectively increase the abundance of commensal E. coli in vivo. Preliminary studies have identified a widely consumed fiber that increases the abundance of commensal E. coli in a model microbial community. I will define the mechanism of action by colonizing these mice with an E. coli transposon mutant library and performing community-wide quantitative proteomics and forward genetic analyses. To model a gut reservoir of pathogenic E. coli, I will substitute UPEC for commensal E. coli in this community, and then administer commensal E. coli with or without fiber to identify interventions that reduce UPEC abundance. In Aim2, I will determine whether commensal and pathogenic microbes adhere to the same surfaces in the gut. A multiplex adhesion assay, using glycan-coated magnetic beads, identified dietary fibers that support adhesion of both UPEC and commensal E. coli. I will validate adhesive interactions in vivo by administering these particles to mice and measuring bacterial localization around beads in situ. Application of the bead-based adhesion assay to cecal microbiota of mice colonized with uncultured human fecal samples will identify additional E. coli strains, as well as uncharacterized gut microbes, that adhere to dietary glycans in vivo. This research will (i) provide insights into the ecological relationships that determine the outcome of dietary interventions designed to promote beneficial species at the expense of known pathogens and ii) provide candidate dietary components, bacterial strains, and microbial genetic targets for manipulating these relationships to enhance human health. Elliot Perens, Ph.D., M.D.

UNIVERSITY OF CALIFORNIA, SAN DIEGO, LA JOLLA, CA

Identification of genetic pathways that regulate kidney cell fate: Hand2 inhibits intermediate mesoderm development

PROJECT SUMMARY

The Principal Investigator seeks mentored training to investigate the genetic regulation of early kidney development using the zebrafish embryo. Research and training will be carried out in the laboratory of Dr. Deborah Yelon, with the support of the Division of Pediatric Nephrology at the University of California, San Diego and an advisory committee composed of specialists in kidney development and disease and in the genetic regulation of organ formation. The objective is to obtain training and pursue laboratory research that will lead to the Principal Investigator's development into an independent physician-scientist. The overall goal of the research is to elucidate the genetic pathways regulating early stages of kidney development in order to gain insight into the etiology of kidney birth defects and to instruct strategies in regenerative medicine. To do so, the PI will take advantage of the genetic and embryological benefits of using the zebrafish as a model organism. Like mammalian kidneys, zebrafish kidneys are derived from the intermediate mesoderm (IM). Work in multiple organisms, including zebrafish, has defined several conserved factors that promote IM formation. However, the mechanisms defining the boundaries that distinguish the IM from its neighboring territories are not yet understood. Recent studies by the PI suggest that the bHLH transcription factor Hand2 defines the lateral border of the IM by regulating cell fate decisions within the posterior mesoderm. While the IM and embryonic kidney are expanded in hand2 mutants, hand2 overexpression results in strong inhibition of IM and kidney development. Additionally, hand2 is expressed in a portion of the posterior mesoderm that lies laterally adjacent to the IM. Venous progenitors arise between these two territories, and hand2 promotes venous progenitor development while inhibiting IM formation at this interface. The lateral hand2-expressing territory also expresses osr1, a zinc-finger transcription factor previously implicated in promoting kidney formation, and genetic analyses suggest that hand2 and osr1 have functionally antagonistic roles during kidney development. Together, these data shed light on a previously unrecognized genetic network that regulates IM boundaries, suggesting a model in which hand2 functions in opposition to osr1 to control the allocation of progenitor cells to kidney and vein lineages. To test this model, two specific aims will be pursued. The first aim will define the role of hand2 in the inhibition of IM lineage specification, using high-resolution fate map analysis and cell autonomy studies. The second aim will identify genes in the pathways through which hand2 and osr1 control IM specification, using complementary candidate gene and whole transcriptome approaches to identify relevant partner and effector genes. These studies will elucidate the role of hand2 in IM formation and illuminate the pathways that define the boundaries of the IM by balancing formation of the kidney and vein lineages. Over the long term, detailed comprehension of these genetic pathways should improve diagnostic, prognostic, and therapeutic options for patients with kidney disease. Chethan Puttarajappa, M.D.

UNIVERSITY OF PITTSBURGH AT PITTSBURGH, PITTSBURGH, PA

Utility of virtual crossmatch in deceased donor kidney transplantation

PROJECT SUMMARY CANDIDATE:

This proposal is for a K08 Mentored Clinical Scientist Research Career Development Award for Dr. Puttarajappa, a Transplant Nephrologist at the University of Pittsburgh Medical Center. He has completed a Master of Science in Clinical Research and aims to develop into a health services researcher with skills to apply observational data, mathematical modeling and decision analysis to kidney transplantation (KT). Under the mentorship of experts at the University of Pittsburgh, he will pursue courses in survey research, epidemiology, health policy and mathematical modeling in health care. These will allow him to complete his proposed research aims and facilitate transition to an independent investigator. Dr. Puttarajappa will be guided by experts in KT outcomes research (Dr. Hariharan), mathematical modeling and decision analysis (Drs. Kenneth Smith and Mark Roberts), and survey methodology and donor research (Dr. Howard Degenholtz). RESEARCH: Broadly, his aims are to investigate the Utility of Virtual Crossmatch (VXM) in deceased donor KT (DDKT). The 2014 kidney allocation system (KAS) gave high priority to highly sensitized (i.e. higher calculated PRA) waitlisted candidates and allowed for wider sharing of kidney donor profile index >85 kidneys, resulting in increased shipping of organs, higher non-local KT surgeries and increased cold ischemia time (CIT). To assess donor-recipient compatibility and avoid immediate transplant rejection, a physical cell-based crossmatch (XM) is routinely performed prior to KT. VXM is a technique that compares donor human leukocyte antigen(HLA) typing and recipient HLA antibody profile to predict physical XM results. VXM has the potential to decrease CIT in DDKT, which will reduce delayed graft function, rejection, and improve graft and patient survival. Reducing CIT could also reduce organ discard and improve organ utilization. Specific AIMS for this proposal are 1) Evaluate current crossmatch practices in deceased donor KT among different donor and recipient subgroups, along with assessing perceptions of transplant professionals regarding VXM, 2) Explore if use of VXM is associated with lower CIT and DGF, and if this is different before and after the introduction of KAS and, and 3) Explore if routine application of VXM prior to DDKT has the potential to improve patient outcomes. These aims will be achieved using a combination of surveys, analysis of data from the United Network of Organ Sharing, and mathematical modeling. His research will identify situations where a VXM will be sufficient to proceed to transplantation without a physical XM. Additionally, findings on national crossmatch practice variation and perceptions of the transplant community towards VXM will help direct future research, changes to histocompatibility protocols and health policy. This award will allow Dr. Puttarajappa to achieve the skills necessary to conduct independent clinical research in KT while simultaneously yielding valuable information on the optimization of crossmatch strategies for DDKT. Mattia Quattrocelli, Ph.D.

CINCINNATI CHILDRENS HOSP MED CTR, CINCINNATI, OH

Glucocorticoid and circadian clock coregulation of insulin sensitivity and metabolism

PROJECT SUMMARY/ABSTRACT

Circadian rhythm plays a central role in metabolic homeostasis and nutrient utilization in nearly all organisms and virtually all tissues. Glucocorticoids are oscillatory regulators of metabolic function that act with cell and tissue-type specificity. Glucocorticoid steroids like prednisone are used to treat a wide range of inflammatory conditions, where their use is associated with prominent metabolic side effects. Chronic daily glucocorticoid intake promotes insulin resistance and obesity, and therefore novel approaches are needed to reverse these dysmetabolic effects. An important breakthrough in glucocorticoid-driven metabolic regulation stems from recently published discoveries that steroid dosing frequency, i.e. daily versus pulsatile weekly, promotes strikingly opposing effects on lean mass quality, exercise tolerance, and energy production. Contrary to daily dosing, weekly glucocorticoids exposure improves nutrient uptake and metabolism, boosting muscle growth and curtailing fat accrual. Specifically, I have uncovered that pulsatile glucocorticoids stimulate branched-chain amino acid oxidative metabolism and insulin sensitivity through a glucocorticoid receptor-responsive epigenomic program focusing on the transcriptional regulator Kruppel-like factor 15 (KLF15). Furthermore, pulsatile glucocorticoids also activate BMAL1 and its molecular cascades. Each of these components, the glucocorticoid receptor, KLF15 and BMAL1 are regulated by circadian oscillations in their metabolic effects. However, it is still unclear whether and how the circadian clock and glucocorticoid cascades interact to promote fuel utilization and favorable metabolic reprogramming, and whether environmental or genetic challenges to this interaction will affect metabolic physiology. To address this question, I propose to (i) dissect circadian regulation of glucocorticoid receptor activation and its effects on glucose and fatty acid utilization in metabolically active tissues like muscle, liver and fat, and (ii) investigate the epigenomic cross-regulation between BMAL1 and KLF15 in driving branched-chain amino acid metabolism and energy production. Experiments will follow a basic-to-translational path from mice models to human cells using a multidisciplinary approach encompassing epigenetic, molecular and metabolic studies. The overarching goal for this proposal is to provide new actionable knowledge of cross-regulation between glucocorticoids and circadian clock, with implications for the treatment of metabolic diseases like obesity and diabetes. Nataliya Razumilava, M.D.

UNIVERSITY OF MICHIGAN AT ANN ARBOR, ANN ARBOR, MI

Mechanisms of extrahepatic biliary proliferation and regeneration

PROJECT SUMMARY/ABSTRACT

Cholangiopathies are incurable, progressive extrahepatic bile duct (EHBD) disorders characterized by injury- induced cholangiocyte hyperproliferation. Development of therapies for cholangiopathies requires better understanding of cellular and molecular mechanisms regulating EHBD proliferation during homeostasis and in response to injury. Molecular pathways that regulate cholangiocyte proliferation are currently not well understood. This application aims to answer a fundamental question about mechanisms of EHBD regeneration and explore strategies to modulate the proliferative potential of the biliary epithelium. The rationale for the proposed research is that defining cellular and molecular interactions underlying cholangiocyte responses to injury can offer novel therapeutic strategies for cholangiopathies and hepatobiliary regenerative medicine. Our preliminary data suggest that Hedgehog (HH) and WNT signaling play important roles in EHBD homeostasis and proliferation after injury. The overarching hypothesis for this proposal is that HH and WNT signaling regulate crosstalk between epithelial and stromal cells to promote EHBD proliferation after injury. In this proposal, we will use genetic mouse models and pharmacological treatments in vivo. Bile duct ligation will be used as an injury model. Human and mouse EHBD organoid (BDO) co-culture with primary mesenchymal cells will be used to directly study epithelial-stromal crosstalk and pathway interaction in vitro and enhance the translational component of this project. Aim 1 will focus on HH signaling and determine if the Indian HH ligand from cholangiocytes signals to GLI1+ HH-responsive fibroblasts to indirectly regulate cholangiocyte proliferation. Aim 2 will focus on WNT signaling and test if WNT from GLI1-expresing cells is critical for cholangiocyte proliferation. Aim 3 will focus on cell-cell and pathway interactions and determine if IHH from cholangiocytes directly regulates WNT production by fibroblasts to induce cholangiocyte proliferation. Under Aims 1 and 2 we will use transgenic reporter and loss-of-function mouse models, pharmacological inhibitors of HH and WNT signaling, in situ hybridization, and immunohistochemistry techniques to define the in vivo effects. Under Aim 3 we take a reductionist approach by using organoid co-culture models. It will also support the development of robust new tools (transgenic mice and organoid models) and skills (in situ hybridization and flow cytometry) to support an independent research program focused on fundamental signaling pathways regulating EHBD. This new K08 application will also promote the development of the PI into an independent NIH-funded investigator and support her long-term goal to understand fundamental mechanisms of EHBD biology to ultimately improve outcomes in patients with cholangiopathies. CASEY REBHOLZ, Ph.D., M.P.H., M.S.

JOHNS HOPKINS UNIVERSITY, BALTIMORE, MD

Metabolomics for Identifying Biomarkers of Dietary Intake & Kidney Disease Progression

PROJECT SUMMARY/ABSTRACT

Chronic kidney disease is associated with high rates of the morbidity and mortality, but few effective treatments exist. Diet is central to kidney disease and its management, and is a modifiable risk factor for kidney disease progression. Metabolomics can now quantify over 800 small molecules in an unbiased approach providing an opportunity to assess the proximal physiologic effect of diet. The specific aims of the research proposal are: 1) to study the relationship between components of dietary intake and kidney disease progression; 2) to quantify the metabolomic expression of dietary intake; and 3) to examine the relationship between metabolites that reflect dietary intake and kidney disease progression. The proposed research leverages three chronic kidney disease studies: 1) the Modification of Diet in Renal Disease (MDRD) study, a randomized clinical trial of dietary protein restriction (N=840); 2) the Chronic Renal Insufficiency Cohort (CRIC) study, a prospective cohort study (N=3,939); and 3) the German Chronic Kidney Disease (GCKD) study, a prospective cohort study (N=5,217). Extensive collaboration with leaders in these research studies will catalyze the proposed research. Funding is provided through the parent studies and other funded grants (NIDDK R01 led by Drs. Andrew Levey and Josef Coresh, Chronic Kidney Disease Biomarkers Consortium) to perform global metabolomic profiling. Casey M. Rebholz, PhD, MS, MPH is a tenure-track Assistant Professor at Johns Hopkins Bloomberg School of Public Health. She seeks a Mentored Research Scientist Development Award in order to obtain essential skills and mentored research experience to prepare for a future career as an independent investigator in the field of nutrition and chronic kidney disease. The research and career development proposal details a five-year plan consisting of in-depth training in metabolomics and chronic kidney disease; advanced coursework in nutrition, kidney disease, and metabolomics (lab methods and analytic techniques); primary mentorship by Dr. Josef Coresh, MD, PhD; co-mentorship by Dr. Lawrence J. Appel, MD, MPH, Dr. Morgan E. Grams, MD, PhD, and Dr. David R. Graham, PhD; and epidemiologic research on the risk of kidney disease associated with dietary intake. Immediate career goals include the mastery of statistical techniques for metabolomics data analysis and nutrition science through an academic curriculum integrated with the research plan. Long term, Dr. Rebholz aims to lead independent research programs investigating optimal diets for the prevention and treatment of chronic kidney disease. Proposed research will advance dietary assessment methodology and provide novel insights into kidney disease pathogenesis with the goal of guiding therapy through dietary interventions, to be tested in future grant proposals by Dr. Rebholz, for the ~13% of the U.S. population with chronic kidney disease. Elizabeth Rogers, M.S., M.D.

UNIVERSITY OF MINNESOTA, MINNEAPOLIS, MN

Optimizing Diabetes Care Quality for Low-Income Patients

This K23 award will establish the candidate, Dr. Elizabeth Rogers, as an independent physician- investigator with expertise in development, implementation, and evaluation of patient-centered primary care interventions that are strategically designed to address patient complexity – both medical and social – to optimize care for low-income patients with multiple chronic diseases. Most of the 29.1 million Americans with type 2 diabetes mellitus (T2DM) have at least one additional chronic condition, but existing management guidelines fail to account for patient complexity. Despite evidence of effective clinic-based interventions, diabetes care quality remains low, particularly in safety net settings serving patients with low incomes, who are disproportionately racial and ethnic minorities, and who carry a larger burden of chronic disease. Until patient complexity factors are meaningfully addressed with specific, pragmatic interventions, patients with comorbid T2DM will remain at risk for poor-quality care. To address this critical need, Dr. Rogers's K23 project will identify key clinic and patient influences on care quality by using a “cumulative complexity model” as a framework. This model focuses on patients' experiences of (1) “workload” (disease-specific work such as attending appointments and self-care, plus “life” demands such as job, family, and transportation) and (2) “capacity” (e.g. fatigue and physical symptoms, plus factors such as literacy, finances, and social support). The project will be conducted in urban Minnesota clinics that are certified Patient-Centered Medical Homes and part of the Hennepin County Medical Center's safety net care system. After stratifying clinics by diabetes care quality, researchers will administer clinician and staff surveys in 3 high- and 3 low-scoring clinics to identify clinic resources associated with optimal diabetes care quality (Aim 1). They will conduct surveys and in-depth interviews with “at-risk” patients from these sites to discern their perspectives on social vulnerabilities that impede their engagement in care and specific clinic resources that are helpful in overcoming these obstacles (Aim 2). They will apply these results using the multiphase optimization strategy (MOST, used to design effective and efficient interventions) to develop and pilot test a clinic-based, multi-component behavioral intervention – one engineered to reduce patient workload, augment patient capacity, and thereby optimize patient-centered care for complex patients with T2DM and depression (Aim 3). A robust training plan, closely aligned with the research plan, will support the development of Dr. Rogers's expertise in patient-centered outcomes research, mixed methodology, and clinic-based intervention development and evaluation. She will be supported by a multidisciplinary team of mentors with expertise in these areas as well as in diabetes, depression, and multimorbidity, and with strong links to community stakeholders. This proposal addresses NIDDK priorities by positioning Dr. Rogers to advance patient-centeredness and improve care quality for patients at risk of experiencing low quality of care, and to test practical and sustainable strategies for improving care delivery and health outcomes in diabetes. Rachel Ross, Ph.D., M.D.

ALBERT EINSTEIN COLLEGE OF MEDICINE, BRONX, NY

Dissecting a novel prefrontal cortical pathway regulating feeding behavior

PROJECT SUMMARY/ABSTRACT

Eating disorders, spanning obesity to anorexia, are highly prevalent and problematic in our society. They are difficult to treat, affect a large proportion of the population, and underlie many other causes of morbidity that have become increasingly costly to manage. Furthermore, feeding behavior is upset in multiple psychiatric disorders and weight gain is a common side effect of many psychotropic medications, but the pathophysiology of these effects is unknown. Therefore, the study of feeding behavior and the neurocircuitry of metabolism are of great interest to psychiatry. Using cutting-edge neuroscience tools to build on traditional genetic methods, the applicant proposes a functional mechanism for projections from satiety-associated neurons in the hypothalamus (POMC neurons) to medial prefrontal cortex melanocortin-4 receptor-expressing neurons (mPFCMC4R) in the pathophysiology of overeating related to obesity. In this mouse-based study, the applicant will examine a novel pathway from POMC neurons to the infralimbic region of the mPFC and investigate the relevance to feeding behavior. First, she will determine if mPFCMC4R neurons, and specifically those that receive projections from POMC neurons, play a role in executive function specific to food-related decisions, focusing on set-shifting and rule reversal (Aim 1). Next, she will perform the same behavioral assays while recording the activity of the mPFCMC4R neurons in awake, behaving mice (Aim 2). These studies will employ genetically engineered mice and viruses that together enable temporal and spatial control and study of specific subpopulations of neurons, and will be performed in both male and female mice. Together, these aims will show that the POMCà mPFCMC4R circuit is a key node in the development of overeating behavior, and consider sex-specific vulnerability to overeating at this unique site of integration of feeding behavior and executive function. Further, this work will provide new animal models for future research with dissociable behavior and physiologic outputs as well as potential therapeutic targets which are urgently needed for eating disorders. The applicant, Dr. Rachel Ross, is well qualified to execute the proposed experiments. She is committed to pursuing a scientific career in neuroscience of behavior and has proposed a comprehensive four-year plan to meet her goal of becoming an independent physician-scientist. Dr. Ross will be working under the primary mentorship of Dr. Kerry Ressler with co-mentorship from Dr. Bradford Lowell. She has enlisted a research advisory committee of internationally-recognized experts in neuroscience and psychiatry to support her. The Department of Psychiatry at McLean Hospital (and secondary site of the Division of Endocrinology at Beth Israel Deaconess Medical Center) is an ideal environment for completion of her scientific and career development objectives given the outstanding research community, tradition of scientific discovery, and ongoing dedication to trainee mentorship in this field. Amy Rumora, Ph.D.

UNIVERSITY OF MICHIGAN AT ANN ARBOR, ANN ARBOR, MI

Sphingolipid and Fatty Acid Biology in Prediabetes and Neuropathy

ABSTRACT

Prediabetes – a condition that precedes type 2 diabetes – affects more than 80 million Americans. Peripheral neuropathy is a diabetic complication that results in a loss of sensation in the limbs due to peripheral nerve damage, and it can develop in both type 2 diabetic as well as prediabetic patients. Despite the prevalence and severity of peripheral neuropathy, there are currently no disease-modifying options. Novel therapeutic targets are therefore of critical importance for future treatments. Dyslipidemia is characterized by increased levels of saturated fatty acids in the plasma, resulting in altered levels of sphingolipids. Ceramides are one type of sphingolipid that are lipotoxic to neurons. Accumulation of saturated fatty acids leads to mitochondrial dysfunction, and cellular apoptosis and may play a central role in the development of peripheral neuropathy. However, monounsaturated fatty acids protect neurons from ceramide-mediated toxicity in vitro, likely via the formation of intracellular lipid droplets. Reversal of ceramide accumulation using monounsaturated fatty acid supplementation may therefore be an effective strategy for normalizing ceramide profiles and preventing peripheral neuropathy. We hypothesize that peripheral neuropathy in prediabetes is the result of toxic ceramide accumulation in neurons in vivo. We further hypothesize that supplementing prediabetic mice with monounsaturated fatty acids will enhance lipid droplet synthesis in neurons to prevent ceramide accumulation and subsequent neurotoxicity. We will test these hypotheses by: 1) determining the ceramide profile of prediabetic mice with neuropathy in both plasma and peripheral nerves, 2) evaluating the efficacy of monounsaturated fatty acid supplementation on ceramide accumulation in prediabetic mice during neuropathy, and 3) determining the mechanisms by which ceramides damage the peripheral nervous system as well as determining the mechanisms by which monounsaturated fatty acid ameliorate this damage. Together, these studies will demonstrate that dyslipidemia plays a central role in the development of peripheral neuropathy and provide an immediate therapeutic target for future treatments. Jose Saenz, Ph.D., M.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

Establishing roles for the type I interferon/double-stranded RNA response and - specific transcripts in the progression to metaplasia in gastric epithelium

Project Summary/Abstract

This application proposes a five year research career development program that focuses on the host and microbial factors that contribute to gastric metaplasia. The comprehensive approach of exploring the host and bacterial determinants of the metaplastic milieu will enhance our overall understanding of the dynamic interplay that establishes a gastric pre-neoplastic state. The candidate is currently an Instructor in Medicine in the Division of Gastroenterology at the Washington University School of Medicine. This proposal is an extension of the candidate’s previous work demonstrating the ability of Helicobacter pylori to exploit metaplastic changes in the host to expand its niche. The proposed experiments will incorporate gastric epithelial biology expertise from the candidate’s mentor, Dr. Jason Mills, as well as Helicobacter pylori mutagenesis experience from the candidate’s co-mentor, Dr. Rick Peek. Together, the candidate will be uniquely positioned to acquire new skill sets and expand on previously developed techniques that will allow him to carve out a unique niche within the field and transition to an independent physician scientist. Gastric cancer remains one of the leading causes of cancer-related deaths worldwide. Chronic infection with the stomach-adapted bacterium, Helicobacter pylori, represents the most significant risk factor for the progression to gastric cancer. In the setting of chronic inflammation, loss of acid-secreting parietal cells from the gastric corpus stimulates a reorganization of the corpus gland, characterized by an increased proliferation of gastric progenitor cells and a reprogramming of post-mitotic chief cells at the gland base into a population of proliferative metaplastic cells, a process that we have termed paligenosis. We recently demonstrated that Helicobacter pylori exploits these metaplastic glandular changes to expand its colonization of the gastric corpus, which is known to confer added oncogenic risk. This proposal describes a dual approach toward identifying and characterizing the host and microbial factors that contribute to the establishment of the metaplastic milieu. From the host perspective, a microarray analysis identified multiple components of the type I IFN/dsRNA signaling pathway that were upregulated in two distinct models of gastric metaplasia. We will dissect the role of this highly conserved antiviral pathway in the previously uncharacterized context of gastric metaplasia. Similarly, we aim to demonstrate that the accumulation of endogenous dsRNA during paligenosis serves as an intra-cellular signal for the development of gastric metaplasia. From the microbial perspective, a newly developed bacterial RNAseq analysis found Helicobacter pylori-specific transcripts that were differentially expressed in the gastric corpus. Using an established pipeline of in vivo and ex vivo experiments, we will validate and characterize these genes in the context of Helicobacter pylori’s colonization of the gastric corpus and establishment of gastric metaplasia. Taken together, this proposal seeks to identify microbial prognostic indicators and areas of potential therapeutic intervention in the development of gastric metaplasia. Jami Saloman, Ph.D., B.A.

UNIVERSITY OF PITTSBURGH AT PITTSBURGH, PITTSBURGH, PA

Neuropathic vs. inflammatory pain in chronic pancreatitis: can unique biomarkers be identified to guide mechanistic approaches to pain treatment?

Abstract

Chronic pancreatitis (CP) is often accompanied by profoundly debilitating pain that is quite difficult to treat. There are no tools available in clinics to properly characterize the subtype of pain a patient is experiencing to choose a therapy most likely to benefit a patient’s pain symptoms. There are two peripheral mechanisms that could contribute, inflammatory pain (IP) and neuropathic pain (NP). A major driver of IP is neurogenic inflammation, inflammation resulting from increased neural activity that drives release of neuropeptides that recruit/activate immune cells. NP results from direct nerve injury (compression or lesion). Importantly, both types of pain are accompanied by unique changes in cytokine expression that can be used to identify mechanisms (IP and/or NP) contributing to a patient’s pain. The development of tools to differentiate IP and NP in the clinic could ultimately streamline CP pain management because there is evidence that certain therapies are far more efficacious for one type of pain versus the other. For example, opioids are more effective for IP while gabapentinoids are more useful for NP. This proposal is designed to compare a novel animal model in which pancreatitis pain is induced purely by IP (optogenetic) and a commonly used animal model of CP (chronic cerulein) in which pain is a result of both IP and NP. The goal is to identify cytokine profiles associated with the specific pain phenotypes and compare to legacy samples from a well-characterized clinical cohort of CP patients. We will also test whether patients show correlations between cytokine profiles and pain characteristics. The long-term plan is to use an iterative approach that uses patient data to refine animal models of CP that can then be used to develop pain-type specific therapies. In addition to incorporating new technologies (optogenetic) to the study of CP, this project will provide in depth training in handling and analyzing human data and will provide a foundation for future prospective human studies that will include survey tools specifically designed for diagnosis of inflammatory and neuropathic pain in humans with CP. Vijay Samineni, Ph.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

DEFINING THE NEURAL BASIS FOR CHRONIC BLADDER PAIN AND ITS ASSOCIATED COMORBID DISORDERS

Project Summary

The goal of this “Career Development Award” is to provide Dr. Samineni with a mentored neurourology research experience and to successfully bridge transition from mentored research to a career as an independent investigator as a neurourologist. Dr. Samineni’s primary goal in the next five years is to establish as a successful independent scientist conducting neurourology research that provides mechanistic insights into our understanding of chronic bladder pain and to obtain R01 level funding. The long-term goal of Dr. Samineni’s research is to independently lead a research group that investigates new avenues for relief of chronic bladder pain and identify new therapeutic opportunities through furthering our mechanistic understanding of chronic bladder pain. To achieve this goal, Dr. Samineni is proposing a project that provides him with significant training in neurourology. Training plan includes rigorous bench work under the primary supervision of Dr. Gereau and Dr. Andriole, attending urology focused grand rounds and didactic series, presentations at national meetings, and training in personnel management, grantsmanship and scientific writing. This training will enable Dr. Samineni to become an independent scientist in urology and allow him to be competitive in obtaining an independent NIH research grant (R01). The institutional environment for career development at Washington University and in the Department of Anesthesiology and Urology is exceptional. Dr. Evers (Chief of Anesthesiology) pledged his full department commitment. To achieve Dr. Samineni’s goal, he is proposing a project that provides him with significant training by examining how maladaptive plasticity in the central amygdala (CeA) circuit contributes to chronic bladder pain in IC/ BPS. Specifically, Dr. Samineni will examine necessity and sufficiency of these circuits in mediating chronic bladder pain. To test this, Dr. Samineni will use optogenetic techniques to control the activity of the CeA neurons and determine their relative contribution to bladder pain. This proposal combines Dr. Samineni’s prior expertise using rodent in vivo work, and pushes forward an innovative combination of genetics, electrophysiology and circuit dissecting tools. Successful completion of these studies will identify the maladaptive mechanisms in these circuits for processing chronic bladder pain and will provide new avenues for developing novel therapies and treatments that would be beneficial for the treatment of chronic bladder pain. Completion of this K01 award will train Dr. Samineni with comprehensive, multidisciplinary training in behavioral pharmacology, optogenetics and whole-cell slice electrophysiology. Furthermore, additional mentored training via the K01 mechanism will allow Dr. Samineni to bridge the gap in his training with regard to skills relevant to running an independent research program. Vanessa Scanlon, Ph.D.

YALE UNIVERSITY, NEW HAVEN, CT

The Role of Intrinsic and Extrinsic Factors in Megakaryocytic-Erythroid Progenitor Lineage Commitment

PROJECT SUMMARY

Candidate. Dr. Scanlon, a member of an underrepresented minority, is a third year postdoctoral fellow in the Department of Laboratory Medicine at Yale School of Medicine with degrees in Molecular Cell Biology, Diagnostic Genetic Sciences, and Biomedical Science. She is an experimental biologist looking to expand her ability to utilize algorithms and perform bioinformatic analyses with the long-term goal of becoming an independent investigator. Her previous training and work with intracellular signaling pathways controlling progenitor cell response to injury combined with her current work in hematopoiesis uniquely qualify her to conduct the proposed work. The proposed career development plan will build upon her previous training and enhance her path toward independent research. This plan includes experimental and didactic learning in image processing, bioinformatic analysis, algorithm optimization, and advanced statistical analyses to independently analyze massive quantities of data. Development in these areas will ready her for independent academic molecular and cell biology research in the current era. Mentor/Advisors and Environment. Dr. Scanlon's primary mentor, Diane Krause, MD, PhD, and imaging advisor, Dr. Joerg Bewersdorf, PhD will guide Dr. Scanlon through the proposed training and research activities. In addition to Dr. Krause's extensive experience in mentoring numerous successful academic researchers, and her expertise in hematopoietic stem and progenitor cells, and Dr. Bewersdorf expertise in high quality live imaging, Dr. Scanlon will also receive bioinformatic analysis support and learn cell-tracking algorithms through established collaborations with Dr. Masahiko Sato, developer of cell-tracking algorithms, and Mr. Rolando Garcia-Millian, a dedicated bioinformatic support specialist at Yale. Dr. Scanlon will meet regularly with her mentor, advisor, and collaborators to ensure her progress. The proposed career development plan utilizes the expertise and rich resources available at Yale to delineate additional training activities to facilitate Dr. Scanlon's research. Research. The molecular mechanisms underlying lineage commitment of hematopoietic stem and progenitor cells have implications in deriving blood cells in vitro for transfusion medicine, as well as elucidating aberrant pathways responsible for hematological disorders. Dr. Scanlon's recent postdoctoral work has focused on studying lineage commitment of Megakaryocytic-Erythroid Progenitors (MEPs). Previous work in the lab identified MYB (a transcription factor) to be important in controlling human MEP fate, however the mechanism remains elusive. She proposes to use live imaging to directly visualize MEPs undergoing lineage commitment, as well as transcriptomic and epigenomic approaches to tease apart the mechanism by which MYB regulates this process. Additionally, she will launch an independent line of studies investigating the effect of intercellular signaling between MEP and other marrow-residing cells on MEP fate. The results of these studies may shed light on more general rules of stem and progenitor fate decisions, as well as potentially help derive patient-specific platelets and red blood cells for transfusions. This career development award will be an instrumental step in Dr. Scanlon's trajectory towards an independent investigator and leader in hematopoiesis. Jarrad Scarlett, Ph.D., M.D.

UNIVERSITY OF WASHINGTON, SEATTLE, WA

Central and Peripheral Mechanisms of FGF1-Mediated Remission of Diabetic Hyperglycemia

Project Summary

This proposal delineates a 5-year program to provide training toward the development of an independent academic research career in the study of integrated central and peripheral regulation of glucose homeostasis. The candidate has been prepared for this pathway by completing MD and PhD degrees and clinical training in Pediatrics and Gastroenterology. He has been scientifically productive at all levels of training through graduate and postdoctoral work, contributing to 15 manuscripts (7 of them first or co-first author), and successfully competing for fellowships from the American Heart Association and the NIH. The proposed research will be conducted in the laboratory of Dr. Michael Schwartz, an expert in the field of hypothalamic regulation of energy balance and glucose homeostasis. It will be overseen by an expert mentoring committee with two members of the Endocrinology Division (Dr. Gregory Morton and Dr. Joshua Thaler) as well as an external advisor (Dr. David Wasserman, Vanderbilt University). The comprehensive training plan involves continued education in the use of isotopic techniques and analytical methods to study regulation of glucose homeostasis and metabolism from the gene to the whole organism level. The proposal focuses on evidence that members of the fibroblast growth factor (FGF) family play a key role in the regulation of glucose homeostasis by targeting hypothalamic glucoregulatory neurocircuits. Using sophisticated metabolic phenotyping, the candidate demonstrated that a single central injection of FGF1 induces sustained remission of diabetic hyperglycemia in rodent models of diabetes. The anti-diabetic effect is not secondary to weight loss, and is not associated with an increase in insulin sensitivity. Furthermore, the ability of icv FGF1 to induce diabetes remission is lost in animals with severe insulin deficiency and additional preliminary data suggest that relapse of diabetes in animals previously responsive to icv FGF1 is associated with progressive pancreatic β-cell dysfunction. This research will characterize the mechanisms by which icv FGF1 induces remission of diabetic hyperglycemia through 2 specific aims. In Aim 1, we will characterize the specific receptors, intracellular signaling cascades, and synaptic changes that mediate sustained remission of diabetic hyperglycemia in response to icv FGF1. Aim 2 focuses on the requirement of an intact basal insulin signal for FGF1 to induce remission of diabetic hyperglycemia and the extent to which FGF1-induced diabetes remission is attributable to increased basal insulin secretion due to preservation of pancreatic β-cell function. The applicant's combination of expertise in neuroanatomy, molecular biology, histochemistry, pharmacology and physiology of the neural regulation of metabolism uniquely qualify him to conduct the studies in this proposal. Outcomes from these studies are expected to provide a compelling rationale for future studies investigating the mechanisms of FGF1-mediated diabetes remission, exploring the translational potential for centrally- targeted FGF1 and related peptides as anti-diabetic agents. Sarah Schrauben, M.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

A Mixed Methods Study of Chronic Kidney Disease (CKD) Self-Management

Chronic kidney disease (CKD) is a growing epidemic, affecting close to 15% of the US population, and is gen- erally progressive, with potentially fatal complications of cardiovascular disease and end-stage kidney disease. Directed treatment of CKD is challenging because CKD is a syndrome with causes that are often not identifia- ble and a dearth of disease-specific interventions. However, there are some cross-cutting approaches that are broadly beneficial for people with CKD, and are recommended by current CKD management guidelines, which include advising patients to engage in self-management behaviors (e.g., monitor blood pressure, maintain physical activity, and avoid tobacco). To date, there has been limited research on self-management in pre- dialysis CKD. The few published studies demonstrate poor uptake of recommended self-management behav- iors, which highlights the need to 1) understand the reasons for substandard engagement in recommended behaviors, and 2) develop and evaluate new approaches to improve CKD self-management. Sarah Schrauben, MD, MSCE, a nephrologist, will target this understudied area in her proposed K23 Award. Dr. Schrauben is positioned to use this award to launch an independently funded research career in the rich environment at the University of Pennsylvania, as she has completed a Masters degree in clinical epidemiology, through which she gained experience in epidemiology, biostatistics, study design, and during her post-doctoral fellowship, conducted a preliminary study on self-management behaviors in a large CKD observational cohort, as well as participated in formal training in qualitative and mixed methods research. Her proposed K23 work will utilize in- depth interviews with CKD patients to understand the influences of participating in health behaviors and identi- fy relevant factors for the design and implementation of a self-management support intervention (Aim 1). In a large cohort of individuals with CKD, Dr. Schrauben will investigate potential predictors of recommended self- management behavior engagement to identify patient characteristics important for a self-management support intervention (Aim 2). Using this information, she will develop a patient-centered self-management support in- tervention harnessing health behavior theory and mobile health (mHealth) technology and then pilot its feasibil- ity (Aim 3). Through her proposed career development, she seeks to gain skills in 1) clinical trial design, im- plementation, and evaluation, 2) mixed research methods, 3) advanced statistical analyses, including the longi- tudinal data analyses from the pilot trial, while garnering expertise in 4) implementation science, and 5) health behavior research. To achieve these objectives, Dr. Schrauben has assembled a multi-disciplinary mentorship team led by national experts in renal epidemiology, biostatistics, clinical trials, mixed research methods, im- plementation science, health behavior research, and interventions leveraging mHealth. Upon completion of these K23 activities, Dr. Schrauben will be poised to conduct a RCT supported by a R01 grant, leveraging her training to improve self-management among persons with CKD. Andrew Schreiner, M.S., M.D.

MEDICAL UNIVERSITY OF SOUTH CAROLINA, CHARLESTON, SC

Improving the Diagnosis of Liver Disease in Primary Care Patients with Abnormal Liver Function

Abstract

Improving the Diagnosis of Liver Disease in Primary Care Patients with Abnormal Liver Function Tests Through Predictive Modeling Reducing diagnostic error has been identified by the Institute of Medicine as a top national priority. Diagnostic errors pervade all of healthcare, with the average individual experiencing one major error during their lifetime. Therefore, improving the diagnostic process and reducing diagnostic error is not only highly appropriate for all patients, but will play a crucial role in optimizing the quality and value of healthcare delivery in the United States.1 Liver disease, with complications including acute liver failure, cirrhosis, and liver cancer ranks as a leading cause of death in America and over recent years has had a significant climb in age-adjusted mortality, while death rates from heart disease and cancer have fallen.2 Despite the increasing preventability of liver- related conditions through early recognition and treatment, the toll of chronic and end stage liver disease continues to rise.3 The traditional diagnostic process, a synthesis of information gathered from history, physical exam, and laboratory testing, performs poorly in the detection of early liver disease.4,5 Instead, clinicians rely more heavily on laboratory studies, and liver function tests (LFTs) in particular.6 Abnormal LFTs are among the most frequently encountered findings in medicine.7,8 Currently, primary care clinicians currently lack the ability to consistently identify liver-related disease from these abnormalities.9-12 Preliminary data in primary care emphasize the immense scope of the problem; in studies from Europe, LFTs have been found elsewhere to be abnormal in nearly 1 in 5 people.13,14 In our preliminary studies, we have up to 40% of patients seen in an academic primary care clinic possessed at least one abnormal LFT. Further, these abnormal liver tests are inappropriately or inadequately followed-up. These data and our own experience indicate that primary care physicians (PCPs) lack the resources to reliably identify and accurately diagnose liver-related diseases amongst these many abnormal LFTs. In this proposal, the candidate and his mentorship team seek to harness inter-professional teamwork and information technology to reduce diagnostic error. They will identify clinical and demographic variables of patients with abnormal LFTs associated with specific liver-related diagnoses in primary care (Aim 1). Additionally, they will develop and validate a predictive model to identify patients with abnormal LFTs at risk for liver-related diagnoses (Aim 2). Lastly, they will create a decision support tool application to aid PCPs confronted with abnormal LFTs to promptly and accurately diagnose liver disease (Aim 3). Anna Seekatz, Ph.D.

CLEMSON UNIVERSITY, CLEMSON, SC

Critical functions of the gut microbiota that mediate recovery from recurrent Clostridium difficile infection

ABSTRACT

Recurrent Clostridium difficile infection (CDI) affects 20-30% of patients following successful treatment of an initial disease episode. It is hypothesized that the gut microbiota, the microbial community within the gastrointestinal tract, is involved in the development of and recovery from recurrence. Microbial- based treatments that promote the growth of `healthy' bacteria, such as fecal microbiota transplantation (FMT), have emerged as an effective treatment method for recurrent CDI. Nevertheless, there is a fundamental lack of knowledge of the microbes responsible for functions that contribute to colonization resistance against C. difficile. We have characterized a murine model of recurrent infection, and observed that murine fecal material from healthy mice is capable of mediating efficient clearance of C. difficile. Previous studies using germ-free mice have successfully established colonization resistance against C. difficile with human microbiota. Contrary to this, we observed that FMT from healthy humans did not resolve CDI in our model. While both FMT treatments changed the microbiota composition, we observed deficient levels of several metabolites. Our mouse model of recurrent CDI provides us with a method to understand how community structures assemble to provide colonization resistance against CDI, enabling us to differentiate the critical microbial features in colonization resistance against C. difficile. We hypothesize that the metabolic environment that limits C. difficile colonization and growth is dependent on the assembly of succinct microbiota communities. The scientific objective of this proposal is to elucidate the link between the structure and function of a microbial community capable of mediating resistance to or recovery from CDI. We aim to do this using the following specific aims: 1) define structural features in microbial communities that mediate colonization resistance to C. difficile and 2) identify metabolic pathways in microbial communities that resolve CDI. The training objective of this proposal is to complement the trainee's background in microbial ecology with bioinformatic and experimental skills that focus on functions of microbial communities to support transition of the candidate into an independent, translational investigator. Completion of these aims will result in more comprehensive knowledge of how microbes function to provide colonization resistance, aiding the critical need to prevent or treat CDI, an important healthcare-associated infection. In conjunction with the exceptional mentoring team and institutional environment, this proposal will provide the candidate with scientific skills and career mentorship to become an independent investigator in an inter- disciplinary field encompassing both microbial ecology and infectious disease. Nilay Sethi, Ph.D., M.D.

DANA-FARBER CANCER INST, BOSTON, MA

Integration of early genetic alterations and inflammation in gastroesophageal premalignancy

PROJECT SUMMARY

Chronic inflammation in the gastrointestinal tract promotes the development of premalignant lesions imbued with potential devastating consequences. Defining the precise molecular mediators that collaborate with inflammation to endorse premalignancy will inform the design of effective prevention strategies. Genomic annotation of premalignant gastroesophageal (GE) lesions revealed that TP53 mutations occur frequently and predict the progression to cancer. Based on these observations, we hypothesize that chronic inflammation provides a selective advantage for GE cells harboring TP53 mutations to generate premalignant disease. The objective of this mentored research career development proposal is to combine our recent findings in cancer genomics with novel mouse models of inflammation to derive new conceptual insights into the premalignant state. To this end, we have designed an experimental system that integrates TP53 alterations in GE cells with two modes of inflammation using a novel mouse model (Aim 1). By labeling TP53 mutant GE cells with fluorescent probes, we will be able to track the evolution of premalignant disease in the setting of inflammation. Furthermore, direct analysis of premalignant lesions from these studies will help elucidate specific mutations and/or pathways that enable a selective advantage for TP53 mutant GE cells. We will also utilize an in vitro system to systematically test the impact of disease-relevant inflammation- associated factors on cellular functions of TP53 altered GE cells (Aim 2). Our preliminary data showed that deletion of TP53 in premalignant GE cells stimulates the production of inflammatory cytokines, implicating a potential vicious feedback cycle. Using a complement of mouse models, organoid culture, and patient samples, we will investigate the functional significance of inflammation pathways induced by TP53 alterations in premalignant GE lesions (Aim 3). Overall, these studies hold tremendous promise for cancer prevention in GE premalignancy. I am a medical oncologist with a research background in cellular and molecular biology. My long-term goal is to become a tenure-track independent laboratory investigator with expertise in gastrointestinal diseases. I am dedicated to leading a basic and translational research laboratory that defines key functional mechanisms of premalignant gastrointestinal disease with an emphasis on inflammation, genomics, and therapeutics. During my proposed training period, I will perform mentored research in the laboratory of Dr. Adam Bass at the Dana-Farber Cancer Institute. I am fortunate to have an exceptional advisory committee to help guide my research and career development including Dr. William Kaelin, Dr. Benjamin Ebert, Dr. Timothy Wang, Dr. Anil Rustgi, and Dr. Kevin Haigis. Coupled with an outstanding institutional environment, training plan, and career development program, the proposed research will enable me to achieve my long-term career aspirations. Valentina Shakhnovich, M.D.

CHILDREN'S MERCY HOSP (KANSAS CITY, MO), KANSAS CITY, MO

Effect of Obesity on Pantoprazole Pharmacokinetics and in Children

PROJECT SUMMARY

Pediatric obesity has reached epidemic proportions, with >30% children meeting criteria for overweight/obese. The alarming obesity epidemic brings with it increasing need for pediatricians to treat chronic obesity-related comorbidities (e.g., GERD) that frequently require long-term medical management. Yet, guidelines are lacking for optimal dosing of medications in this population. The proposed investigation builds on my recently published findings, from two independent prospective investigations that demonstrate increased systemic exposure to the proton pump inhibitor (PPI) pantoprazole in obese vs. non-obese children, suggesting slower PPI drug clearance in obesity. Using intravenous pantoprazole as a model drug probe for the hepatic drug metabolizing pathway CYP2C19, I will test the hypothesis that hepatic adiposity underlies the observed reduction in pantoprazole clearance, and that weight-reduction reverses alterations in liver adiposity, hepatic drug clearance and drug effect. Understanding of the biologic and physiologic mechanisms underlying altered drug metabolism and clearance is the first step toward developing accurate predictive models for optimizing the dose selection of PPIs, and other drugs commonly prescribed to obese patients. Postdoctoral training in an NIH-funded pediatric clinical pharmacology program at Children's Mercy Kansas City (CMKC; T32HD069038) prepared me well for a research-focused career in pediatric therapeutics by providing didactic training in curve fitting and compartmental/noncompartmental pharmacokinetic analysis; however, clinical pharmacology training has limited exposure to quantitative systems pharmacology, a biomedical discipline that uses mathematical computer models to characterize interactions of biological systems, disease processes and pharmacology, to individualize drug therapeutics in a variety of circumstances. The K23 mechanism will enable me to build on my basic pharmacology skill-set and pursue this advanced training, essential for developing physiololgically-based pharmacokinetic and pharmacodynamic (PBPK/PD) models for simulating and predicting the drug doseàconcentrationàresponse relationship for children with gastrointestinal disorders, starting with PPI dosing for obese children, who are disproportionately affected by GERD. To test the validity of the PBPK/PD models that I develop, I will need to design, conduct and effectively lead prospective longitudinal clinical trials, a mentored-research opportunity afforded to me by this K23. To expand my models to other drugs commonly prescribed to children, I will also need to update my knowledge base of drug metabolizing enzymes beyond CYP2C19, as proposed in my Education Plan. As a pediatric gastroenterologist and clinical pharmacologist at CMH, with 75% protected research time, a mentoring team comprised of expert NIH-funded faculty, lead by pharmacogenomics expert J. Steven Leeder, PharmD, PhD, I have the requisite institutional support, pedigree and academic environment to accomplish the research and training goals described in this K23 application. Allison Shapiro, Ph.D., M.P.H.

UNIVERSITY OF COLORADO DENVER, Aurora, CO

The neural underpinnings of disinhibited eating behavior in adolescents with and without obesity

PROJECT SUMMARY

Dr. Shapiro is an excellent candidate for the NIDDK Mentored Research Scientist Development Award. Currently, she is transitioning from a postdoctoral research position in the NIMH-funded T32 program for Developmental Psychobiology, Psychopathology and Behavior (MH015442-39A1) to faculty in the Department of Psychiatry at the University of Colorado. She has a Master of Public Health in Epidemiology and Biostatistics, a Doctorate in Epidemiology, and an impressive publication and funding record. The proposed research plan has two specific aims: 1) Determine the extent to which an energy surplus stimulus alters neuronal response within, and functional connectivity between homeostatic and non-homeostatic brain regions in adolescents with and without obesity; and 2) Determine the degree to which neuronal connectivity between homeostatic and non-homeostatic brain regions is associated with disinhibited eating behaviors among adolescents with and without obesity. Dr. Shapiro has designed an innovative study to address these aims that will employ functional neuroimaging methodology (Aim 1) and a directly observed measure of disinhibited eating behavior (Aim 2). She will test the overarching hypothesis that altered brain function in and between homeostatic and non-homeostatic brain systems underlie disinhibited eating behaviors in adolescents, and that this is most pronounced in adolescents with obesity. The career development plan supports the research aims and Dr. Shapiro’s transition to independence with training in 1) Functional neuroimaging methodology and analysis; and 2) Eating behaviors in youth: theory, measurement and interpretation. Her mentors, Drs. Jason Tregellas (primary), Susan Johnson, Marc Cornier (co-mentor) are recognized experts in the fields of neuroimaging, eating behaviors, and obesity, respectively. Dr. Shapiro will also receive additional training with leading experts in the field of pediatric eating behaviors at the Pennsylvania State University (collaborators). The University of Colorado Denver, Anschutz Medical Campus will provide a rich training and research environment for Dr. Shapiro to accomplish her short- and long-term research and career goals. Specifically, the University of Colorado Brain Imaging Center, with its research- dedicated 3T MRI scanner (Director: Tregellas), will be a significant resource for Dr. Shapiro’s current and future research efforts. Dr. Shapiro also has the full backing of her department (Psychiatry) which will continue to provide her with protected research time, a start-up package with significant research funds, office space, and equipment. Furthermore, Dr. Shapiro will have access to The Children’s Eating Laboratory (Director: Johnson), a state-of-the-art laboratory for collecting observational data for eating behaviors in pediatric groups. This award will help to establish the foundation on which Dr. Shapiro will build her expertise as a leading scientist in the study of the neural underpinnings of pediatric eating behaviors and obesity risk. Andrea Shin, M.D.

INDIANA UNIV-PURDUE UNIV AT INDIANAPOLIS, INDIANAPOLIS, IN

Fecal microbiota, short chain fatty acids, bile acids, and colonic transit in Irritable Bowel Syndrome

PROJECT SUMMARY

The precise mechanisms by which the gut microbiome and contributes to irritable bowel syndrome (IBS) symptoms are unclear. However , it is recognized that microbial metabolites such as short chain fatty acids (SCFA) and bile acids exert important effects on gastrointestinal physiology. Thus, an enhanced understanding of the relationships between the gut microbiome, SCFAs, and bile acids will be essential to developing novel strategies for effective IBS treatment. This career development application is submitted on response to PA-18- 374 in which the candidate proposes a hypothesis-driven research strategy to (1) identify changes in the fecal microbiota that are associated with SCFA and bile acid profiles in IBS, (2) establish SCFAs as an actionable IBS biomarker, and (3) interrogate interactions between SCFA and bile acids in IBS. This proposal builds on preliminary data acquired through the support of an institutional KL2. The specific aims of this research strategy are to (1) identify shifts in the relative abundance of SCFA-producing bacteria that are associated with fecal SCFA levels, markers of SCFA production through inulin fermentation (residual fecal inulin after inulin challenge), and colonic transit in IBS with constipation (IBS-C), IBS with diarrhea (IBS-D), and controls and (2) identify shifts in the relative abundance of bile acid dehydroxylating bacteria that are associated with fecal bile acids and markers of SCFA production in IBS-C, IBS-D, and controls and test if bile acid profiles are associated with markers of SCFA production. To achieve these aims, the candidate will develop a prospective cohort of well- phenotyped IBS patients and matched-controls who will undergo (1) baseline assessments of their fecal microbiota, fecal SCFAs, fecal bile acids, and colonic transit, followed by (2) repeat assessments of fecal microbiota, fecal SCFAs, fecal bile acids, as well as measurement of fecal inulin after standardized dietary intervention with inulin supplementation. The proposed career development plan integrates in-depth mentoring from a multidisciplinary team of senior scientists, advanced coursework in bioinformatics and microbiome analysis, experiential learning through the conduct of the proposed research, and a highly supportive research environment. The mentorship team, which includes independent investigators with expertise in clinical and translational research in microbiome science (Nelson) and functional gastrointestinal disorders (Camilleri); data analysis and biostatistics (Xu); bioinformatics (Dong); and career development (Chalasani) will guide the candidate's research and career development. The superb institutional infrastructure for facilitating junior investigators and substantial institutional commitment greatly strengthen this application. At the conclusion of the program, the candidate will be well positioned to become an independent physician investigator studying novel microbial and metabolomics biomarkers and novel interventions in IBS. Jung-Im Shin, Ph.D., M.D.

JOHNS HOPKINS UNIVERSITY, BALTIMORE, MD

The Comparative Effectiveness and Safety of Second-line Therapy in Patients with Type 2 Diabetes

PROJECT SUMMARY/ABSTRACT

Jung-Im Shin, MD, PhD, is an Assistant Scientist in the Department of Epidemiology at the Johns Hopkins Bloomberg School of Public Health. She seeks a K01 Mentored Research Scientist Development Award in order to obtain essential skills and mentored research experience for an independent career as a research scientist in the field of pharmacoepidemiology and diabetes. The research proposal details a five-year plan consisting of in-depth training in novel causal inference methods, mentorship by Dr. Morgan Grams, MD, PhD, and Dr. Elizabeth Selvin, PhD, MPH, and pharmacoepidemiologic research in utilization, effectiveness, and safety of second-line therapy in patients with type 2 diabetes. The specific aims of the research agenda are to: 1) Describe the real-world epidemiology of second-line therapy among people with diabetes after metformin monotherapy; 2) Evaluate real-world effectiveness of 5 classes of second-line therapy for hyperglycemia management among similar people with diabetes after metformin monotherapy; and 3) Evaluate real-world risks of adverse events and benefits on long-term clinical outcomes of 5 classes of second-line therapy among similar people with diabetes after metformin monotherapy. Immediate career goals include the mastery of applying target trial emulation and estimating the effect of a dynamic treatment regime with Robin’s g-methods through an academic curriculum integrated with the research plan. Long-term, Dr. Shin aims to lead independent research programs investigating optimal treatment and decision-making to improve health outcomes of patients with type 2 diabetes. Completion of the proposed aims will provide vital information relevant to the approximately thirty million U.S. patients with type 2 diabetes. Type 2 diabetes is associated with high levels of morbidity and mortality. Although consensus exists around first-line therapy, there remains great uncertainty regarding the optimal choice of a second-line therapy, with inconsistent recommendations across guidelines. A better understanding of the risks and benefits of different classes of second-line antidiabetic drugs would greatly help clinical practice in patients with type 2 diabetes.

Robin Shook, Ph.D.

CHILDREN'S MERCY HOSP (KANSAS CITY, MO), KANSAS CITY, MO

Metabolism, appetite, and physical activity in adolescents

PROJECT SUMMARY/ABSTRACT

Childhood obesity is the most serious public health problem of the 21st century, given the prevalence, global reach, and widespread health, economic, and social consequences. While weight gain at the most basic level is due to a chronic energy surplus, there are a host of influences that act on the energy balance system that remain largely unknown. Thus, there is a critical need to identify previously unexamined factors that may influence the energy balance system in order to develop effective interventions for prevention and treatment. Our long-term goal is to quantify the dynamic relationships between various physiological and psychological components of the energy balance system. Our short- term goal is to examine linkages between physical activity, appetite control, and energy metabolism in adolescents. The status quo as it relates to appetite control is that body weight status and related peripheral signals are a key regulator of energy intake. In contrast to body weight playing a primary role in energy regulation, our working hypothesis is that high levels of regular moderate-to-vigorous physical activity (MVPA) result in a metabolic phenotype consisting of enhanced metabolic function and proper regulation of appetite, which prevents the development of obesity. We have reported that adults with low levels of MVPA have dysregulated appetite control, evidenced by higher levels of energy intake and elevated levels of dietary disinhibition. Additionally, primary mentor Thyfault has observed that rats with low fitness possess dysregulated control of appetite and reduced hepatic fat oxidation compared to rats with high fitness. Based on these findings in adults and rodents, in addition to supportive work by others, we hypothesize that low levels of MVPA create a metabolic phenotype, leading to dysregulated appetite control which predisposes individuals to weight gain. To test this hypothesis we will perform two studies focused on the links between physical activity and appetite. For Study 1, we will assess metabolic function and appetite control in male and female adolescents stratified by bodyweight (normal vs overweight/obese) and physical activity (sedentary vs. active) using a 2 x 2 cross-sectional study design (N=40). For Study 2, we will examine the influence of MVPA on the same outcomes by randomly assigning sedentary overweight/obese adolescents (N=44) to either a structured-exercise group or no treatment control for three months. The specific aims of the proposed studies are: 1) To examine the association between physical activity and appetite control in adolescents, with metabolic function serving as a mediator; and, 2) To test if increased physical activity through an exercise intervention influences metabolic function and appetite control. At the completion of the proposed studies, it is our expectation that we will have described the independent effects of physical activity and body weight on energy metabolism and appetite in adolescents. This study will have a significant positive impact on the understanding of energy balance in adolescents, and will inform public health interventions that specifically target mechanisms associated with weight gain in youth. Sarah Short, Ph.D.

VANDERBILT UNIVERSITY MEDICAL CENTER, NASHVILLE, TN

Antioxidant regulation of intestinal homeostasis and disease

Project Summary

This proposal details a 5-year training plan to aid the continued develop of Dr. Sarah Short, Ph.D. into an independent GI researcher. This research plan will focus on elucidating the role of glutathione peroxidase 1 (GPx1), a ubiquitously expressed selenoenzyme and potent , in inflammatory bowel disease (IBD) and colitis-associated dysplasia (CAD). Compelling preliminary data using Gpx1-/- mice indicates that unlike many whose loss exacerbates murine colitis, loss of GPx1 confers striking protection from dextran- sodium sulfate (DSS)-induced colitis. GPx1 deficiency also increases survival and stemness in 3D organoids and alters metabolism in cells, which may additionally promote regeneration and wound healing. GPx1 expression also modifies immune cell function, as Gpx1-/- bone marrow-derived macrophages have heightened response to “M2” stimuli and decreased migratory ability. Together, these results suggest that GPx1 augments inflammatory injury through alterations in both epithelial and immune cell function. Based on these finding, the hypothesis of this proposal is that GPx1 is detrimental in inflammatory bowel disease by altering stem cell function, redox homeostasis, and immune responses. Further, inhibiting GPx1 activity may be an effective therapeutic strategy. This hypothesis will be tested in two specific aims to determine how GPx1 contributes to intestinal epithelial cell homeostasis, oxidative stress, colitis, and colitis- associated dysplasia. The first aim will investigate epithelial function, capitalizing on Dr. Short’s over 10 years of experience in epithelial cell biology and barrier function. The second aim will complement epithelial-based studies by determining how GPx1 loss alters immune cell recruitment, differentiation, and function, and identify how these changes modify intestinal injury responses. In addition to being the logical “next step” experiments in defining GPx1 function, these experiments provide the perfect framework to further Dr. Short’s development in aspects of mucosal immunology which contribute to intestinal diseases, and will include new training in flow cytometry, chemokine analysis, bone marrow transplantation, and the T-cell transfer colitis model. Dr. Short’s career development will be further enhanced by regular discussions with primary mentor, Dr. Christopher Williams, and her mentoring committee consisting of Drs. Keith Wilson, Jeremy Goettel, and Sean Davies. All studies and training will take place at Vanderbilt, and the institution, Department of Medicine, and Gastroenterology Division are highly supportive of Dr. Short’s academic career and fully support her application. Dr. Short’s ultimate goal is to become an independent academic researcher focusing on mechanisms which regulate development and severity of IBD and colitis-associated cancer that can lead to improved therapeutic options for these patients. Interestingly, both specific aims proposed in this application will evaluate GPx1 as a therapeutic target using tiopronin, which is FDA-approved and well-tolerated. Together, these training experiences will ensure Dr. Short is poised to direct a well-rounded independent research program in IBD. SHAZIA MEHMOOD SIDDIQUE, M.S., M.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

Evaluation of variability in care and outcomes for patients with gastrointestinal bleeding

PROJECT SUMMARY

Gastrointestinal bleeding (GIB) is a major cause of inpatient hospitalizations in the United States and comprises a heterogeneous population with appreciable variability in patient outcomes amongst hospitals. Nationally, 30- day mortality rates are estimated at 7%; however, mortality and readmission rates vary widely across hospitals. Such variability suggests the potential to improve patient care and save lives by identifying modifiable factors that influence outcomes after hospitalization with GIB. Patient-related factors, hospital structure and processes of care play major roles in determining outcomes for many other diseases, but the impact of these factors on outcomes related to GIB has not been adequately defined. The objective of this proposal is to identify and understand sources of variation in care for patients with GIB, including patient-level factors, organization (i.e. structure) and processes of care (e.g. endoscopic and interventional radiology utilization) amongst hospitals. Our central hypothesis is that modifiable factors impact patient outcomes during hospitalizations for GIB, which include both structural characteristics and processes of care. Our proposal is to build a multi-level model of patient outcomes standardized across hospitals, accounting for patient factors and assess the impact of hospital factors, and then qualitatively explore additional processes of care, through the following interrelated specific aims: Aim 1- To develop a risk-standardized model for GIB outcomes across hospitals using Medicare inpatient, outpatient, and pharmaceutical claims data. To ensure generalizability and clinical applicability, model validation will be performed in a multipayer, multicenter database and using UPenn health system data which contains clinical variables. Aim 2 - to identify hospital factors (structures and processes of care) associated with GIB outcomes, using a combined dataset of the aforementioned Medicare data with American Hospital Association Annual Survey data. Aim 3 – to explore barriers and facilitators of processes of care for inpatient GIB among hospitals with better and worse patient outcomes, using qualitative methods. This mixed methods approach will allow for a comprehensive evaluation of drivers of variation in care for gastrointestinal bleeding. The expected outcome for the proposed research is that it will identify important modifiable factors in care associated with patient outcomes for those hospitalized with GIB. This can lead to the development and validation of quality metrics to improve patient care. To facilitate completion of the research and further the academic development of the applicant, the PI will enroll in coursework that builds on her Master of Science by focusing on statistical methods using clustered and longitudinal data, data analytics in confounding, and qualitative methodologies. This proposal has unequivocal and outstanding divisional and institutional support and exceptional mentorship of experienced faculty with a strong track record in epidemiology, health services research, and mentoring prior K-award grant recipients. These activities will allow the PI to develop the skills necessary to become a successful independent NIH-funded investigator and a leader in GI-related health services research.

Jocelyn Silvester, Ph.D., M.D.

BOSTON CHILDREN'S HOSPITAL, BOSTON, MA

Mechanisms and Markers of Small Intestinal Epithelial Injury and Villous Atrophy

PROJECT SUMMARY

Mucosal remodeling with villous atrophy is a generic response to small intestinal injury with many causes that require different treatments. While easily recognized on intestinal biopsy specimens, determining the specific cause of villous atrophy is more challenging and often leads to therapeutic delays. This Career Development Award is based upon the hypothesis that transcriptomics can both elucidate mechanisms of villous atrophy across diseases and provide improved tools for diagnosis and monitoring. Jocelyn A. Silvester, MD PhD is Instructor at Harvard Medical School and a subspecialist within the Celiac Disease Program at Boston Children's Hospital. She has gained substantial basic and clinical research experience during her doctoral and medical training and has demonstrated commitment to an academic career in patient oriented research. This Career Development Award will provide additional mentored training and research opportunities in bioinformatics and mucosal immunology for Dr Silvester to advance her quantitative research skills while addressing the current knowledge gap related to mechanisms of small intestinal epithelial injury and the lack of disease-specific biomarkers of villous atrophy. Dr Ciarán Kelly, an acknowledged expert in celiac disease and intestinal inflammation, and Dr Isaac Kohane, an expert in bioinformatics and quantitative biology, will serve as mentors. In addition an Advisory Committee comprised of experts across multiple relevant disciplines (Drs Leffler, Lencer, Goldsmith, and Fasano) will meet with the applicant and mentors regularly to assess progress. This award will aid Dr Silvester in establishing an independent research program as an academic physician- scientist through a structured training plan that includes formal course work in bioinformatics and statistics, as well as relevant clinical research in functional genomics. She will also receive additional training in quantitative interpretation of intestinal histology and grant writing to facilitate her goal of using insights from large-scale genomic data to improve diagnosis, treatment, and outcomes for patients with celiac disease and other enteropathies. The research at the foundation of this application aims to characterize the different mechanisms leading to villous atrophy and to identify a molecular signature of celiac disease activity in peripheral blood. Celiac disease will be used as a model to study the time course of changes occurring as villous atrophy recurs during gluten challenge in celiac patients who had recovered on a gluten-free diet. Additionally, comparative transcriptomic studies will be performed using intestinal tissue from patients with celiac disease, adult-onset autoimmune enteropathy and drug-induced (olmesartan) enteropathy. Finally, we will study peripheral blood mRNA markers of villous atrophy in participants in our ongoing Manitoba Celiac Disease Cohort study undergoing protocol biopsy 24 months after starting a gluten-free diet. The substantial research, educational and clinical resources of Boston Children's Hospital, Harvard Medical School and the Harvard Celiac Research Program are committed to the applicant to ensure successful attainment of the goals of this award. Siddharth Singh, M.S., M.D.

UNIVERSITY OF CALIFORNIA, SAN DIEGO, LA JOLLA, CA

Comparative Risks and Benefits of Pharmacological Therapies for Older Patients with Inflammatory Bowel Diseases

PROJECT SUMMARY/ABSTRACT

The prevalence of inflammatory bowel diseases (IBD) in older adults (≥65y) is rising, and there is limited evidence-based guidance on treating these older adults. In older patients with multi-morbidity, treatment decisions should factor in not only risk of disease complications, but also treatment complications and non-IBD, extra-intestinal complications. Hence, to inform optimal treatment approach, a comprehensive assessment of comparative effects of different therapies on all of these outcomes is warranted. Existing single-center observational studies are limited by small sample size, and missing data due to fragmented health care, whereas administrative claims-based studies are limited by lack of detailed clinical data; these limitations can be overcome by linking the two data sources. In this patient-oriented mentored career development award proposal, Dr. Siddharth Singh proposes to: (Aim #1.1) characterize disease burden and treatment patterns, (Aim #1.2) assess and predict risks of death, disease, treatment and extra-intestinal complications using machine-learning algorithms and (Aim #2.1) compare overall effectiveness and (Aim #2.2) safety of different treatment strategies in older patients with IBD. This will be studied using a highly innovative informatics-based approach in a multi-site, electronic medical record (EMR)-based cohort of older patients with IBD, linked to their corresponding Medicare claims. The EMR-based cohort will facilitate phenotyping and disease severity assessment, and linkage to Medicare claims will augment exposure and outcome ascertainment, overcoming challenges of fragmentation of healthcare and short follow-up. The central hypothesis is that, that older adults have systematically different risk profiles than younger adults, and using biologic monotherapy is a safer and more effective approach to treating IBD, as compared to using long-term corticosteroids alone, non-biologic immunomodulator monotherapy, and combination therapy of biologics and immunomodulators. The access to the advanced infrastructure of pSCANNER (Patient-centered Scalable National Network for Effectiveness Research), one of 13 PCORI-funded Clinical Data Research Networks, and entire Medicare database, in a highly supportive and conducive environment at UCSD, with cross- disciplinary mentorship by a collaborative and experienced team of mentors and advisors from diverse backgrounds (clinical informatics, comparative effectiveness research, IBD therapeutics) is a key strength of this application. Besides directly informing clinical practice on treatment approaches in older patients with IBD, this proposal will enhance the career of the candidate by providing unique skills in applied clinical informatics, privacy-preserving record linkage techniques, predictive analytics, and comparative effectiveness research, and create a multi-institutional, EMR- based cohort of well- characterized IBD patients, linked with Medicare claims. This will ultimately contribute to the candidate's long-term goal of establishing an independent IBD research career focusing on comparative effectiveness research and population health management using novel informatics-based approaches. Meghan Sise, M.S., M.D.

MASSACHUSETTS GENERAL HOSPITAL, BOSTON, MA

Effect of Hepatitis C Virus Eradication on Chronic Kidney Disease Progression

PROJECT SUMMARY/ABSTRACT

Hepatitis C Virus (HCV) infection is a common comorbidity in patients with chronic kidney disease (CKD) that leads to accelerated progression to end-stage renal disease. Because of the recent approval of all- oral, interferon (IFN)-free, direct-acting antiviral therapy (DAAs), HCV can now be cured in the majority who are treated. This proposal seeks to identify factors that determine CKD outcomes in patients treated with DAAs, and to investigate the effect of enhanced IFN activation that occurs after HCV eradication on kidney function. Aim 1 employs the Scalable Collaborative Infrastructure for a Learning Healthcare System (SCILHS) Network, an electronic health records network covering 12 healthcare systems, to examine a large, diverse cohort of 10,000 patients with HCV infection to (1) determine the effect of DAAs on eGFR decline over five years follow- up and (2) identify factors that predict which HCV- infected patients are likely to have progressive CKD despite treatment of HCV infection. Aim 2A proposes to recruit 40 patients with HCV and CKD undergoing DAA treatment to prospectively study the IFN-pathway to determine if monocyte chemoattractant protein 1 (MCP-1), a candidate marker of IFN- activation, predicts which patients will have progressive CKD and which will recover. Aim 2B examines patients who develop de novo immune-mediated kidney diseases (lupus-like immune complex glomerulonephritis and focal segmental glomerulosclerosis) after DAAs to determine if baseline and post-treatment IFN-stimulated genes and chemokines are increased in patients who develop autoimmunity. This K23 Mentored Patient-Oriented Research Career Development Award proposal seeks to explore the effect of HCV eradication and IFN activation on CKD progression, and to prepare Dr. Sise for a career as an independent translational researcher in academic medicine. Dr. Sise's clinical training is in internal medicine and nephrology, with prior Masters-level training in biostatistics and patient-oriented research. During the course of this career development award, she will be supported by her institution to devote 75% of her time to focus on developing this research plan and completing didactic and hands-on training in longitudinal data analysis and immunology through coursework and applied analytic experience. Dr. Sise will benefit from the guidance of her primary mentor, Dr. Raymond Chung, an international leader in basic, translational, and clinical studies of HCV and HIV, and her co- mentor Dr. Ravi Thadhani, an established clinical and translational CKD investigator. Her training will also be overseen by an advisory committee of senior scientists, Drs. Jules Dienstag, Steven Grinspoon, and Arthur Kim, with collective expertise in mechanisms of immune activation in HCV and HIV, biomarker research, and clinical trials. She will work in collaboration with Dr. Kenneth Mandl (SCILHS network PI), Dr. Sushrut Waikar (CKD biomarkers) and Dr. Yuchiao Chang (Statistician). Dr. Sise's goal is to become an independent clinician-investigator at an academic center with a research program focused on strategies to slow CKD progression. Brittany Smalls, Ph.D., M.H.S.

UNIVERSITY OF KENTUCKY, LEXINGTON, KY

Social Network Analysis and Social Support Intervention for Rural Dwelling Older Adults with T2DM

PROJECT SUMMARY/ABSTRACT

Approximately 29.1 million (9.3%) adults in the US have type 2 diabetes (T2DM).1 T2DM is associated with significant complications including premature coronary artery disease, kidney disease, stroke, and blindness.1 Estimates show that prevalence of T2DM is 17% higher in rural dwellers compared to their urban counterparts.2 For example, the prevalence of T2DM for adults in Kentucky's rural Appalachian counties is 13.6%,3 compared to the overall state prevalence of 10.6% 3. Appropriate self-care behaviors are necessary for optimal clinical outcomes in T2DM 4-6 and account for 90% of the variance in glycemic control 7. Yet, self-care among T2DM is notoriously poor in general and even worse in rural-dwellers. Without attention to improving self-care in this population, we are facing an epidemic of T2DM complications in rural areas of the US. The impact of living with T2DM in rural communities is compounded in the aging population. The prevalence of T2DM increases with age and an estimated 25% of older adults (≥65 years) have T2DM 8. Older adults living with T2DM have more difficulty than younger people adhering to self-care regimens and social/environment factors account for up to 85% of their self-care nonadherence.6,9-13 Notable social/environment factors include competing social demands, lack of social support (SS), and stress.14-17 Social support plays a particularly significant role in older adults with T2DM. The extent and type of SS available in rural communities can positively or negatively impact self-care in those diagnosed with T2DM. 18-25 To better understand the effects of the social environment in communities where older rural dwelling adults reside and to lay the foundation for interventions, social network analysis (SNA) will be used to determine social support network structures. By using SNA, social support structures can be developed and relationships between individuals within a social network can be evaluated. 26 The overall objective of this study is to improve T2DM self-care regimens in older rural dwelling adults by training influential community members to be community health workers and provide social support strategies within an older adults' existing social network that promotes T2DM self-care. The proposed study seeks to: Aim 1—Use social network analysis to identify key players in a rural community to serve as community health workers for those living with T2DM. Aim 2—Use mixed methods to evaluate the impact of social support on T2DM self-care and clinical outcomes and evaluate social networks in rural dwelling older adults with T2DM. Aim 3—Test the feasibility and preliminary effectiveness of a 6-week intervention administered by community health workers with the support of members of older adults' individual social network. ANJU SREELATHA, Ph.D.

UT SOUTHWESTERN MEDICAL CENTER, DALLAS, TX

Novel mechanisms of redox signaling in acute kidney injury

PROJECT SUMMARY

Acute kidney injury resulting from ischemia reperfusion or toxin induced damage is correlated with mitochondrial dysfunction and increased ROS levels. The damaging effects of ROS on DNA, protein and lipids can be ameliorated by cellular antioxidants that detoxify the ROS. I have identified a novel mechanism of cellular detoxification in the mitochondria by a predicted pseudokinase, Selenoprotein O (SelO). Preliminary studies using E. coli and S. cerevisiae demonstrates that SelO catalyzes the transfer of AMP from ATP to multiple substrates involved in redox homeostasis to dampen oxidative damage and prevent cell death. Due to the phyletic spread and conservation from bacteria to humans, we postulate that SelO will play a role in oxidative stress response in mammalian systems that are heavily dependent on the mitochondria, such as the kidney. We will test this hypothesis by identifying the molecular targets and pathways mediated by SelO, and characterizing the functional importance of SelO in the rodent cisplatin-induced acute kidney injury model. A deep understanding and technical expertise in the fields of oxidative stress and nephrology are essential to the successful completion of this proposal. This career development award will allow me to strengthen my scientific skill set by training in kidney isolation, and phenotypic characterization in combination with biochemical assays to evaluate mitochondrial health. These studies will define a novel paradigm of signaling in the mitochondria, which can be usurped to identify therapeutic targets to prevent oxidative damage in a plethora of pathologies including acute kidney injury. Shylaja Srinivasan, M.D.

UNIVERSITY OF CALIFORNIA, SAN FRANCISCO, SAN FRANCISCO, CA

A comprehensive approach to understanding the determinants of metformin pharmacokinetics and pharmacodynamics in youth with type 2 diabetes

PROJECT SUMMARY

The prevalence of type 2 diabetes (T2D) is increasing in youth with current management strategies ineffective at maintaining durability of glycemic control. Metformin is the cornerstone of treatment of T2D in youth, yet youth with T2D have high treatment failure rates on metformin therapy. The reasons for treatment failure remain unknown, and need to be comprehensively understood in order to optimize dosing strategies for this safe and cost-effective therapy. To address this unmet medical need, Shylaja Srinivasan MD proposes to build on her successful patient-oriented genetic research through this career development award with the overall objective to use an integrated approach to understand the determinants of metformin response in youth with T2D. With the assistance of a multi-disciplinary mentoring team, she will evaluate the pharmacological, clinical and physiological determinants of metformin response in youth with T2D. Dr. Srinivasan proposes the following aims: (1) develop a population PK model to describe the pharmacokinetics (PK) of metformin in youth with T2D; (2) characterize the variance in long-term response to metformin, integrating comprehensive clinical, pharmacological and genetic data with disease progression in youth with T2D and develop a clinically usable precision dosing tool; and (3) evaluate the precision dosing tool for metformin in a pilot study of youth with T2D in a real clinical setting. To advance these studies, she has access to large datasets and samples from pediatric patients with T2D. In Aim 1, Dr. Srinivasan will develop a population PK model using PK data from 60 youth with T2D recruited from UCSF. In Aim 2, she will develop a longitudinal model to characterize the association between dose, predicted metformin PK (Aim 1), clinical factors, genetic factors if any and metformin response using samples already collected from 699 youth with T2D from the Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) study. She will then utilize these results to develop a clinically usable precision dosing tool. In Aim 3, she will evaluate the precision dosing tool in a pilot study of 20 youth from UCSF, comparing precision dosing with conventional dosing for metformin in a real clinical setting. Results from this proposal will inform an R01 in which precision dosing algorithms for metformin will be tested in a large randomized control trial and will be expanded to include other drug classes. Dr. Srinivasan's long- term career goal is to become an independent clinical researcher and to leverage pharmacological and genetic methods to improve therapeutic strategies for youth with T2D. In this career development award, her training goals are to gain experience in clinical pharmacology and quantitative pharmacokinetics; become proficient in advanced biostatistical methods; gain experience in the conduct of pediatric clinical trials; and engage in career development activities in preparation for R01 funding. She will accomplish these goals with the advice and mentoring from a world-class multi-disciplinary mentoring team, participation in relevant didactic coursework and hands-on research experience, all necessary for future independent research success. Rachel Stine, Ph.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

PRDM16 regulation of metabolism in the intestinal stem cell niche

PROJECT SUMMARY

Long term objectives and training aims: With this award, Dr. Rachel Stine will receive the support, mentorship and training required to reach her ultimate goal of becoming an independent investigator focused on the metabolic control of stem cells within the intestinal niche. This research is an excellent fit for the mission of the NIDDK as it relates to both digestive and metabolic disorders. The University of Pennsylvania offers all of the scientific resources required to complete this proposal, as well as two exemplary research programs focused on metabolic studies and intestinal biology respectively. Dr. Stine has assembled a group of renowned scientists to serve as her mentors, advisory committee and collaborators. She has developed a training plan to enhance her publication record, to secure independent funding in the form of project grants and to apply for and secure an independent position by the completion of this award. Dr. Stine’s distinctive research program seeks to answer basic questions about stem cell biology in the intestine, and will ultimately provide insight into how alterations in metabolic control of stem cells and their differentiating daughters can lead to a disease state. Dr. Stine will master techniques essential to her success in this proposal and her future independent research; integrate and expand her expertise in metabolism and intestinal biology through classes, mentorship and interactions within the broader scientific community; and build skills to successfully secure an independent position and start a new laboratory. Background and research aims: Intestinal stem cells have the capacity to rapidly divide and replenish the intestinal lining every few days. Preliminary studies completed by Dr. Stine show that deletion of the transcription factor PRDM16 in an adult mouse causes severe intestinal wasting within five days and death shortly after. RNAseq following Prdm16 deletion shows downregulation of metabolic genes in the intestinal crypt, particularly members of the fatty acid oxidation (FAO) pathway. Intriguingly, pharmacological inhibition of FAO blocks budding and growth of intestinal enteroids, specifically in the proximal small intestine where PRDM16 is highly expressed. Both PRDM16-deficiency and pharmacological inhibition of FAO can be rescued by supplementation with acetate, which can replenish pools of acetyl-CoA. Aim 1 will determine which intestinal progenitor cell populations require high levels of PRDM16 and FAO, allowing for more targeted analysis into how these pathways regulate intestinal differentiation. This aim will also explore whether mechanisms identified in mice are applicable to a human system. Aim 2 focuses on why FAO specifically is required for acetyl-CoA production, even in the presence of other nutrients. Because acetyl-CoA facilitates acetylation of histones, histone profiling as well as genetic perturbations in the acetyl-CoA pathway will be used to explore these mechanisms. This proposal will determine how metabolic changes in intestinal stem and progenitor cells translate to changes in cell behavior and provide fundamental insights into the role metabolism in this system. Sana Syed, M.D.

BETH ISRAEL DEACONESS MEDICAL CENTER, BOSTON, MA

A Novel Class of Lipids as Potential Therapeutic Agents to Promote Pancreatic Beta cell Survival and Prevent Type 1 Diabetes.

Project Summary/Abstract:

Research: Type 1 Diabetes (T1D) is an autoimmune disease characterized by loss of insulin-producing islet β- cells and insulin deficiency. Despite several clinical trials no intervention has been found yet which has long- term effects, and can be utilized safely over years without adverse effects. Therefore, developing therapeutic targets which are safe, and act directly on islets to attenuate inflammatory responses and improve β-cell function in humans with T1D are of utmost importance. We recently discovered a novel class of mammalian lipids called Palmitic Acid Hydroxy Stearic Acids (PAHSAs) that are highly anti-inflammatory and anti-diabetic. PAHSAs also potently reduce the incidence of T1D in non obese diabetes (NOD) mice, a model of autoimmune T1D. This project will investigate the mechanisms for these beneficial effects of PAHSAs in T1D. Specific Aim1: To determine the immune mechanisms by which PAHSAs delay onset and reduce incidence of T1D. Specific Aim2: To investigate the cellular mechanisms by which PAHSAs directly promote β-cell survival and function. This research will provide novel insights into how PAHSAs modulate immune function and promote islet β-cell survival/function which may be used develop novel therapies for T1D treatment in humans. Candidate: I have a long- standing interest in studying the signaling mechanisms responsible for islet β-cell death and dysfunction. My doctoral training with Dr. Anjan Kowluru focused on the role of small G-proteins and reactive oxygen species in insulin secretion under physiological and pathophysiological states. My postdoctoral training with Dr. Barbara B Kahn has been geared towards identifying novel therapeutic approaches to enhance β-cell function and improve glucose-insulin homeostasis. My tenure in Dr. Kahn’s lab has been extremely productive and I have authored and co-authored 8 papers with one first author paper in Cell, a second in under revision at Cell Metabolism. During the grant period, I will expand my knowledge of islet autoimmunity by learning immunological techniques and signaling pathways involved in β-cell survival/function. This K01 career development award will provide me with protected research time to undergo the final and critical training phase of my career prior which will equip me to successfully transition to independence. Environment: The Division of Endocrinology, Metabolism and Diabetes at the BIDMC is an ideal training site for basic scientists and has a distinguished record of training young scientists towards a path of independence. Graduates from Division have gone on to make transformative discoveries that continue to shape the future of diabetes and metabolism research. The research proposed in this award will be carried under the mentorship of Dr. Kahn at BIDMC and Dr. Mathis at the HMS with guidance of a scientific advisory committee (Drs. Hao Wu, Gokhan Hotamisligil, and Susan Bonner-Weir) who are committed to my development as an independent scientist. I will meet weekly with Dr. Kahn, monthly with Dr. Mathis, and quarterly with Advisory Committee members to discuss results, experimental design, future directions, and for the preparation of my R01 grant. Ismail Syed, Ph.D.

UNIVERSITY OF VIRGINIA, CHARLOTTESVILLE, VA

Computational Characterization of Environmental Enteropathy

PROJECT SUMMARY/ABSTRACT

Undernutrition afflicts 20% of children < 5 years of age in low- and middle-income countries (LMICs) and is a major risk factor for mortality. Linear growth failure (or stunting) in children is tightly linked to irreversible physical and cognitive deficits, with profound implications for development. A common cause of stunting in LMICs is Environmental Enteropathy (EE) which has also been linked to decreased oral vaccine immunogenicity. To date, there are no universally accepted, clear diagnostic algorithms or non-invasive biomarkers for EE making this a critical priority. In this K23 Mentored Career Development Award application, Dr. Sana Syed, a Pediatric Gastroenterologist with advanced training in Nutrition at the University of Virginia, proposes to 1) Develop and validate a Deep Learning Net to identify morphological features of EE versus celiac and healthy small intestinal tissue, 2) correlate the Deep Learning Net identified distinguishing EE intestinal tissue findings with clinical phenotype, measures of gut barrier and absorption, and bile acid deconjugation, and 3) Use a Deep Learning Net computational approach to identify distinguishing multiomic patterns of EE versus celiac disease. This work will be carried out in the context of an ongoing birth cohort study of environmental enteropathy in Pakistan (SEEM). Dr. Syed proposes a career development plan which includes mentorship, fieldwork, coursework, publications, and clinical time that will situate her as an independent physician-scientist with expertise in translational research employing computational `omics and image approaches to elucidate biologic mechanisms of stunting pathways and in identification of novel and effective therapies for EE. Konrad Szymanski, M.D.

INDIANA UNIV-PURDUE UNIV AT INDIANAPOLIS, INDIANAPOLIS, IN

A patient-centered approach to urinary incontinence and quality of life in childrenand adolescents with spina bifida

Most people with spina bifida (SB) suffer from urinary incontinence (UI), necessitating life-long urological care. Health professionals have assumed that continence, achieved by medical or surgical management, leads to better health-related quality of life (HRQOL) in people with SB. However, studies have failed to demonstrate this association. Because SB results in a multisystem disability, UI may not be the only, or even primary, determinant of HRQOL in people with SB. To respond to individuals’ preferences and needs, clinicians must focus on patients’ individual health goals across several dimensions (e.g., symptoms; physical and social function) and determine how well they are met. Only by identifying patients interested in UI treatments, and helping them define their therapeutic goals, will we be able to determine if improving UI increases HRQOL. My long-term goal is to use my clinical and research expertise to optimize children’s SB care, which is currently disconnected from patient-centered goals and evidence-based practice. My short-term goal is to develop a tool (MyGoal) to improve how we approach UI decisions with SB patients. This tool would offer many advantages, including simplifying decision making by helping patients articulate what is important to them, framing the discussion, and aligning treatments toward those goals. Finally, this approach should allow for effective shared decision making (SDM) by letting patients select health outcomes of highest priority, and clinicians determine treatments most likely to achieve them. By using the tool to achieve goal setting and attainment, we hypothesize that patients will experience greater SDM, satisfaction with patient- provider communication, and HRQOL. Therefore, the specific aims of this career development award are to: (1) Engage children with SB to better understand their experience with UI in the context of other competing care issues; (2) Employ a mixed- methods approach to develop and refine the MyGoal tool to identify SB patients interested in addressing UI and establish their continence goals; and (3) Employ a ‘before-after’ prospective study design at a single site to measure the impact of the MyGoal tool on SDM, children’s satisfaction with patient-provider communication, and their HRQOL. The work proposed in this application will have a significant impact by improving SB patient care through the use of patient- centered tools. In the future, we anticipate applying for R03-level funding to develop the MyGoal tool for adults. We also anticipate applying for R01-level funding to evaluate interventions targeting patient- centered goals in SB across several healthcare systems and to monitor renal function prospectively, using the patient cohort from the K23 award. This application will serve to advance my expertise in qualitative research, including design-thinking methods, patient communication and SDM, patient- reported outcome evaluation, and cutting-edge quantitative study design and analysis. Receiving this award is essential to my career development, as it will provide me with further practical research experience and didactic training, within a team of experienced and encouraging mentors in a patient- centered research environment. Tze-Woei Tan, M.P.H., M.D.

UNIVERSITY OF ARIZONA, TUCSON, AZ

Determinants in Limb Preservation in Hispanics and Native Americans

ABSTRACT

The complications of diabetic foot ulcerations (DFUs) are severe and costly to patients and society. DFUs are quite common: 1 in 4 adults with diabetes will develop a DFU, and 2 out of 3 treated DFUs will recur. Every year, 40,000 individuals lose a leg owing to DFUs. Compared to other groups, Hispanics and Native Americans (NAs) experience a disparate rate of diabetes, foot ulceration, and lower extremity amputation. To date, our understanding of the factors leading to racial and ethnic disparities in major amputations (ankle and above) is limited. Knowledge of the differences in access to care, barriers to limb salvage (amputation prevention), patients' preference in revascularization (improving arterial blood flow for wound healing), and factors related to major amputation are particularly essential to the development of targeted interventions. The goal of this K23 proposal is to address the critical knowledge gap regarding disparities in lower extremity amputations among Hispanics and NAs— vulnerable and often underserved populations. This K23 proposal will leverage the unique and diverse patient populations in the Southwestern Academic Limb Salvage Alliance (SALSA) and the NIH-supported Precision Medicine Initiative Cohort within the All of Us Research Program (AoURP), to determine patient-centered factors that influence the care of the Hispanic and NA populations with DFUs. The objectives of the study include Aim 1: Evaluate the racial and ethnic disparities in healthcare access and utilization among adults with diabetes and DFUs using the cross-sectional survey data of the AoURP; Aim 2: Determine the patient-reported barriers to DFU care and perceived risk factors related to major amputations using focus groups; and Aim 3: Prospectively identify the clinical factors and non-clinical variables (individual, community, and health system-level) related to major amputations among 219 Hispanics and NAs with DFUs. The Classification and Regression Tree (CART) will be used to identify vulnerable subgroups with DFUs that might benefit from early identification and aggressive treatment. The proposed study is significant, as it focuses on the traditionally understudied Hispanic and NA populations. The proposed study is innovative in that it uses the standard Wound, Ischemia, and Foot Infection (WIFi) classification system to stratify the severity of the ulcer presentation. The proposed, rigorous training plan and highly experienced mentorship team composed of national experts in health disparities research, mixed methods research, DFUs and limb salvage, and implementation science will prepare the PI to independently conduct research aimed at designing, conducting, and evaluating culturally competent interventions. Finally, the findings of this study will address a critical knowledge gap regarding disparities in amputations among these vulnerable and underserved groups that continue to make up an increasing portion of the Southwest's population, with the overarching goal of reducing lower extremity amputations among at-risk minority populations. Molly Tanenbaum, Ph.D.

STANFORD UNIVERSITY, STANFORD, CA

ONBOARD: OvercomiNg Barriers & Obstacles to Adopting Diabetes Devices

Project Summary

Type 1 diabetes (T1D) is a burdensome chronic disease that requires a demanding self-management regimen to optimize glycemic control and prevent short- and long-term complications. Advances in insulin pumps and continuous glucose monitoring (CGM) technologies have enabled improved glycemic control and reduced risk of complications and have laid the groundwork for the arrival of closed loop systems – the first of which became commercially available in 2017. Closed loop systems, which integrate an insulin pump and CGM to partially automate insulin delivery, represent a major paradigm shift in diabetes care such that people with T1D will offload some of the diabetes management burden to an automated system, resulting in tighter glucose control and better quality of life. However, a minority of adults (18-50) with T1D currently use CGM – a core component of closed loop - and a concerning proportion quit CGM within the first year, which does not bode well for the adoption of closed loop. Low uptake and high discontinuation of CGM among adults with T1D signal the need for well-designed, tailored interventions that provide education, support and problem-solving skills to empower device users to work through physical, social, and other barriers to maximize benefit from advanced diabetes technologies. In this application, Molly Tanenbaum, PhD, proposes a series of studies with an overarching goal of developing advanced behavioral research skills while refining a comprehensive behavioral intervention package to equip adults with T1D with the resources and skills to maximize benefit and minimize daily interference from CGM, with application for closed loop system adoption. The specific aims in this career development award proposal are: 1) to deliver, iteratively refine a behavioral intervention package, ONBOARD (OvercomiNg Barriers & Obstacles to Adopting Diabetes Devices), in a small sample of adults with T1D using CGM, and then conduct a randomized controlled trial of ONBOARD comparing device use, health and psychosocial outcomes after adults with T1D receive the intervention versus CGM-only; and 2) to use qualitative interview data to adapt ONBOARD for adults initiating closed loop systems. Results from these studies will provide preliminary data to inform a larger trial of ONBOARD for CGM and closed loop adoption and sustained use in a future R01 application. Dr. Tanenbaum’s long-term career objective is to be an independent investigator with a programmatic line of research focused on optimizing use of emergent diabetes technology to improve health and quality of life outcomes for adults with T1D. The mentoring, coursework, and career development activities proposed in this 5-year training award will provide Dr. Tanenbaum with the advanced statistical training and experience conducting a randomized controlled trial – skills necessary for a successful career as an independent behavioral diabetes researcher. Elliot Tapper, M.D.

UNIVERSITY OF MICHIGAN AT ANN ARBOR, ANN ARBOR, MI

Optimizing Measures for Clinical Trials in Covert Hepatic Encephalopathy

Optimizing Measures for Clinical Trials in Covert Hepatic Encephalopathy PI: Elliot B. Tapper, MD Among all complications of cirrhosis, none is more devastating than hepatic encephalopathy (HE). Although it will afflict up to 40% of patients with cirrhosis, HE has few dedicated researchers. A spectrum of neuropsychiatric disability, HE ranges in severity from covert (deficits in executive function) to overt (confusion, even coma). Even covert HE – the earliest stage – is independently associated with repeated hospitalization, falls, motor vehicle accidents and death. Unfortunately, strategies to prevent the development HE are limited. Furthermore, investigations of potential therapies are complicated by limited knowledge of both the clinical predictors of HE and impact on patient well-being and daily functions. This grant proposes to close these gaps. The training program aims to prepare the applicant for a career vested in identifying risk factors for HE, and in designing interventions to prevent HE and to improve patient well-being. A master's degree in clinical investigation with targeted additional courses will provide training in clinical trial design, calibration of patient reported measures for clinical trials, and longitudinal data analysis to develop clinical predictive models. The mentorship team assembled is diverse and talented, including Dr. Anna Lok (a clinical trialist and content expert), Dr. Jasmohan Bajaj (an authority on HE and clinical trials), Dr. Noelle Carlozzi (a neuropsychologist with expertise in mixed- methodology), and Dr. (a trialist with a focus on interventions for frail patients). In research aim 1, we will determine the most important domains of health related quality of life (HRQOL) for patients with covert HE. In Aim 2, we will analyze a cohort of 300 cirrhosis patients without HE followed for up to 4 years to determine the modifiable risk factors for HE and to develop predictive models for HE and falls. In Aim 3, we will launch a pilot intervention aimed at the improvement of HRQOL and prevention of falls in patients with covert HE using a web-based platform that co-ordinates a combination of text-message, email and interactive-voice response to deliver structured encouragement to meet nutritional and exercise goals aimed at improving sarcopenia. By the conclusion of the K23, the applicant will be positioned for academic success with a niche in an important complication of cirrhosis and providing necessary preliminary data for an R01 supporting a larger, randomized trial. See Wan Tham, M.D.

SEATTLE CHILDREN'S HOSPITAL, SEATTLE, WA

Sleep deficiency and autonomic function in pediatric chronic abdominal pain

Project Summary/Abstract

Childhood chronic abdominal pain is highly prevalent, and associated with significantly poorer physical and psychosocial functioning, and increased school absenteeism. Moreover, these children are at a greater risk of becoming adults with chronic abdominal pain, overlapping pain syndromes and psychosocial dysfunction. Chronic pain has become a public health crisis, with annual costs exceeding $600 billion. At present, the mechanisms underlying chronic abdominal pain are not well understood, limiting the development of interventions. There is now compelling evidence that sleep deficiency may be a precursor of pain development. Additionally, sleep deficiency is associated autonomic dysfunction and affective disorders that are commonly reported by individuals with chronic abdominal pain, highlighting shared central neurobiological processes. Understanding the role of sleep deficiency as a potential mechanism of chronic abdominal pain will expand avenues for treatment. The long term goal of this research program is to develop novel strategies to interrupt the trajectories of chronic abdominal pain. The objectives of this application are to elucidate the longitudinal impact of sleep deficiency on the course of abdominal pain in adolescents, and to delineate the roles of autonomic and psychological function in this relationship. The K23 candidate is an Assistant Professor in the Department of Anesthesiology and Pain Medicine at the University of Washington, and a pediatric pain physician and anesthesiologist at Seattle Children's Hospital. This proposal outlines a well-integrated research and training plan for mentored, patient oriented career development supported by an interdisciplinary team of mentors with expertise in adolescent development, psychophysiological research, longitudinal modeling, and clinical research applications. This award will provide the candidate with specialized instruction in the evaluation of sleep and cardiophysiological biomarkers in pediatric abdominal pain populations, and expertise in the conduct of longitudinal studies. The central aims are to compare sleep deficiency, pain processing, autonomic and psychological function in adolescents with chronic abdominal pain compared to healthy adolescents (Study 1); and to evaluate these longitudinal trajectories and inter-relationships for adolescents with chronic abdominal pain compared to healthy adolescents over 6 months (Study 2). Assessments will include subjective evaluation of pain, autonomic and psychological function, in addition to home-based actigraphy monitoring of sleep patterns that will be paired with laboratory assessment of psychophysical pain tasks and heart rate variability. Data generated from this research will directly inform a planned R01 proposal to develop tailored interventions targeting sleep deficiency and autonomic function as modifiable risk factors for treatment. Completion of the proposed training will prepare the candidate for a successful independent research career in pediatric chronic abdominal pain medicine. Michael Thompson, Ph.D., M.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

Impact of Maternal Obesity on Offspring Non-alcoholic Fatty Liver Disease: Role of Bile Acid Homeostasis and Microbiome

PROJECT SUMMARY/ABSTRACT

The primary goal of this proposal is to develop the principal investigator, Dr. Michael Thompson, into an independent physician scientist in the field of hepatology research. Michael has previously received PhD training in cellular and molecular pathology with a focus on liver disease. At the current time, he has completed clinical training in Pediatric Endocrinology and has designed a 5 year career development plan to provide additional training in bile acid metabolism and microbiome research. At the end of this career development award, he will become an independent investigator with his own lab program evaluating the developmental origins of liver disease. Dr. Nicholas Davidson, Chief of Gastroenterology at Washington University, will mentor the PI. Dr. Davidson is a well-known leader in intestinal and hepatic bile acid metabolism research and an experienced mentor. Dr. Phil Tarr, Chief of Pediatric Gastroenterology at Washington University, will serve as co-mentor for the PI. Dr. Tarr is a recognized leader in microbiome research which is a primary focus of this proposal. The PI will take advantage of the abundant basic science and clinical resources available at Washington University to develop his own clinically relevant basic research program. Obesity and its complications affect 78 million adults and 13 million children with an estimated economic impact of $2.0 trillion per year. Growing evidence supports that in utero and perinatal events drive risk for insulin resistance and obesity associated complications such as nonalcoholic fatty liver disease (NAFLD) in the offspring. Our preliminary findings indicate that alterations in bile acid homeostasis are associated with this increased risk. This proposal will focus on defining the mechanisms behind the observed alterations in bile acid homeostasis. I will utilize an established model of maternal high fat/high sugar diet exposure to test this hypothesis. In the first aim, I will evaluate cholesterol absorption, bile acid excretion, and bile acid metabolism/transport to define which are contributing to the increased bile acid pool size and composition. In the second aim, I will define whether vertical transmission of the microbiome occurs across generations and whether changes in the microbiome impact bile acid metabolism and metabolic liver disease in the offspring. In the third aim, I will evaluate the efficacy of targeting the bile acid pool size or pool composition as a preventative approach for metabolic liver disease offspring of obese dams. Specifically, I will treat offspring with a bile acid sequestrant (cholestyramine) or a hydrophilic bile acid (UDCA) prior to feeding a western diet, after which I will evaluate insulin resistance and steatosis. Identification of potentially pathogenic alterations in bile acid pool composition, metabolism, and/or transport will support further hypothesis driven research design to identify the mechanism of increased risk for disease progression. Once a mechanistic link is proven between altered bile acid metabolism in offspring and risk for NAFLD progression, this information will be used to design bile acid based preventative therapies to prevent disease progression in at risk patients. Jena Tronieri, Ph.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

Improving Weight Loss in Early Non-responders to Behavioral Treatment

PROJECT SUMMARY/ABSTRACT

My long-term career goal is to be an independent investigator who develops effective obesity treatments and matches these treatments to patients' characteristics. Through my training in clinical psychology, I gained skills in the development and delivery of behavioral treatments (BTs) for obesity and in conducting randomized controlled trials (RCTs) to test their efficacy. As a postdoctoral fellow, I was then introduced to obesity pharmacotherapy, but still have limited experience in this area. In order to successfully develop a career in precision medicine for obesity, I will need additional training in behavioral, biological, and pharmacological influences on body weight. The proposed K23 award will allow me to devote 95% effort to filling critical gaps in my training and to conducting research that will prepare me to be an independent investigator in obesity treatment. My training plan will prepare me for an independent research career by increasing my knowledge in three key areas: 1) behavioral phenotypes associated with obesity and methods for their objective measurement; 2) neuroendocrine mechanisms of energy balance regulation that may impact weight loss; and 3) the use of pharmacologic agents to improve weight loss. I will accomplish these objectives by receiving guidance from mentors, engaging in hands-on research training, and completing relevant coursework and seminars. My research project will complement these training goals by examining behavioral phenotypes and neuroendocrine biomarkers as predictors of early weight loss and by testing whether medication enhances weight loss in patients with a suboptimal early response to BT alone. Participants will complete an initial assessment of behavioral and biological characteristics, followed by 4 weeks of BT. Those who lose < 2.0% of initial weight during the run-in will then be randomly assigned to an additional 24 weeks of: 1) BT plus placebo; or 2) BT plus medication (liraglutide 3.0 mg). I believe that low satiety will predict poor early weight loss with BT and that pharmacotherapy will enhance 24-week weight loss for patients with suboptimal response to BT. This project could shape best practice recommendations for obesity treatment and ultimately result in algorithms for matching treatments to patient characteristics. The environment at the Center for Weight and Eating Disorders at the University of Pennsylvania is well-equipped with staff, resources, and infrastructure to support the proposed project. I will also access state-of-the-art facilities, coursework, and seminars at Penn to complete my research and training goals. My mentorship team will be led by Dr. Thomas Wadden (Professor of Psychology) and includes training from Dr. Tanja Kral (Associate Professor of Nursing) in behavioral phenotypes of obesity and Dr. Matthew Hayes (Associate Professor of Nutritional Neuroscience) in mechanisms of energy balance. This comprehensive, interdisciplinary mentored approach will allow me to acquire knowledge in areas essential to my career goals, implement the proposed research plan, and develop a successful R01 application prior to the end of the K23 award. Eva Tseng, M.P.H., M.D.

JOHNS HOPKINS UNIVERSITY, BALTIMORE, MD

Design and Implementation of An Intervention to Improve Prediabetes Management in Primary Care

PROJECT SUMMARY/ABSTRACT

Diabetes prevention is crucial for reducing the public health burden of diabetes in the United States. 84 million U.S. adults have prediabetes but 90% are undiagnosed. The NIH has prioritized finding efficient ways to translate outcomes achieved in diabetes prevention trials into routine healthcare practice. The National Diabetes Prevention Program recognizes lifestyle change programs meeting specific metrics that help prevent or delay diabetes. However, an effective model for engaging health systems and primary care providers in facilitating screening, testing and referral to these programs is lacking. There is currently no systematic approach to prediabetes management in the primary care setting, but it is essential for addressing the burden of diabetes. To address this need, we propose the following interrelated Specific Aims: 1) To conduct a retrospective cohort study to describe longitudinal primary care provider practices around the diagnosis and treatment of prediabetes using a linked claims and electronic medical record (EMR) dataset, 2) To conduct key informant interviews with stakeholders relevant to diabetes prevention, including leadership from health care system/clinics, insurance plan leaders, providers and patients, to identify barriers and facilitators to implementation, 3) To design and pilot test the diabetes prevention clinical pathway within a primary care clinic in Baltimore, MD, and evaluate its effectiveness on provider screening and intervention, and patient engagement in diabetes prevention over a period of 12 months. Dr. Eva Tseng is an Assistant Professor in the Division of General Internal Medicine and a clinical researcher at The Johns Hopkins University School of Medicine. Her long-term career goal is to become an independent clinician-investigator conducting pragmatic intervention studies with a focus on improving the prevention of diabetes in primary care. The proposed career development plan includes didactic and research training activities that will substantially build her skills in developing and implementing an intervention. These will include training in implementation science, clinical trial methods, quantitative data analytic methods for large claims/EMR databases, as well as qualitative data collection and analytic methods. This training will occur in the rich training environment of the Johns Hopkins Medical Institutions, including the Bloomberg School of Public Health and the Welch Center for Prevention, Epidemiology, and Clinical Research. She will receive guidance from her outstanding team of mentors and advisors with expertise in the methodologies needed for conducting the planned research, and a track record of mentoring and funding. The award and protected time will allow Dr. Tseng to build an independent NIH-funded research career and become a leader in pragmatic approaches to improve the prevention of diabetes in primary care with potential applications to other chronic diseases. Ebele Umeukeje, M.P.H., M.D.

VANDERBILT UNIVERSITY MEDICAL CENTER, NASHVILLE, TN

Patient-centered Approach To Improving Dialysis Adherence in African Americans

Dr. Ebele Umeukeje is a Nephrologist and a Health Services Researcher in the Nephrology Division at Vanderbilt University. Her long-term career goal is to become a successful Independent Physician Scientist with expertise in kidney disease disparities research focused on improving clinical outcomes in vulnerable kidney disease patients. She is specifically interested in reducing the excessive hospitalization and re- hospitalization in end-stage renal disease (ESRD) by intervening upon dialysis treatment non- adherence. Her short-term career goals, with support from this K23, are to gain 1) advanced skills in the development of novel culturally sensitive, multi-level strategies to eliminate disparities by enhancing patient engagement in self-care and 2) practical experience in patient-centered adherence and disparities research, including the responsible conduct of research. Excessive hospitalization and re- hospitalization in ESRD has significant public health relevance to patients, providers and health systems due to its costs both financially and to patients' morbidity and mortality, and emerging evidence shows dialysis treatment adherence is an important modifiable contributor. Compared to whites, African American ESRD patients have a four-fold higher ESRD prevalence, an unacceptably high re- hospitalization rate and higher rates of dialysis treatment non-adherence. The goals of this proposal are to 1) identify novel patient and provider-level determinants of dialysis treatment non-adherence; 2) develop culturally sensitive interventions to improve dialysis treatment adherence; and 3) provide the expert mentorship and experience necessary to facilitate Dr. Umeukeje's successful transition to an independent Physician Scientist with expertise in improving adherence, reducing disparities and optimizing outcomes in ESRD. Ryan Ungaro, M.S., M.D.

ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, NEW YORK, NY

Assessing Predictors of Response to Anti-Tumor Necrosis Alpha Therapy in Early Crohns Disease

This project will evaluate predictive biomarkers of response to anti-tumor necrosis factor alpha (anti- TNF) therapy in early Crohn's disease (CD) patients. Candidate: The primary objective of this application is to support Dr. Ryan Ungaro's career development into an independent patient-oriented investigator in the field of personalized medicine for inflammatory bowel disease (IBD) patients. Dr. Ungaro's career goal is to become an independent researcher and leader in the application of predictive biomarkers and models to select the best treatment and optimize care for recently diagnosed IBD patients. Dr. Ungaro's proposed training activities are in four areas: 1) advanced biostatistical analysis, 2) predictive biomarker analysis, 3) computational genomics, and 4) principles of immune monitoring. To achieve this, he has assembled a mentoring and advisory team led by Dr. Judy Cho, Director of the Charles Bronfman Institute of Personalized Medicine and an expert in translational genomics, and Dr. Bruce Sands, Chief of the Division of Gastroenterology, who has expertise in clinical and translational investigation of IBD therapeutics, having driven much of the pioneering research in anti-TNFs. Environment: The Icahn School of Medicine at Mount Sinai has a strong tradition of outstanding research and is one of the top 20 medical schools in NIH funding. The Mount Sinai Division of Gastroenterology is consistently considered one of the top 10 divisions in the country by US News and World Report and is an international leader in IBD research and clinical care. Research: CD is a chronic, progressive, inflammatory condition affecting the gastrointestinal tract. Anti-TNF medications have vastly improved the treatment of CD patients, however a significant number of individuals do not respond to these agents which are very costly and have potentially fatal side effects. New classes of medications for CD are being released bringing the opportunity for personalized medicine. Clinicians will need the tools to help decide which medication will work best for each individual CD patient. This will be particularly important in recently diagnosed patients since effective early treatment can decrease long-term complications. Therefore our specific aims are to (1) determine the association of peripheral blood proteomic markers with response to anti- TNF (2) assess the association of intestinal tissue gene expression markers with anti-TNF response and (3) explore the mucosal immune architecture and predictive capacity for anti-TNF response of single cell RNA sequencing (scRNASeq) of intestinal biopsies. We will study recently diagnosed CD patients (within 2 years of diagnosis) from 3 prospective cohorts with biosamples: a population-based IBD inception cohort, a multi-center cohort of recently diagnosed pediatric CD patients, and a single-center biorepository of IBD patients linked to health records data. In addition, a cohort of recently diagnosed CD patients will be recruited for scRNASeq analysis. The general approaches and skills developed during this award can also be applied to new targeted medications and form the basis for future research on biomarkers of treatment response in early IBD. Mary Ellen Vajravelu, M.S., M.D.

CHILDREN'S HOSP OF PHILADELPHIA, PHILADELPHIA, PA

Behavioral Economics for Activity Motivation in Adolescents and Young Adults with Prediabetes and Type 2 Diabetes

Project Summary/Abstract

Obese adolescents and young adults (AYA) with type 2 diabetes (T2D) are significantly less active than non- diabetic peers, despite the many disease-related benefits of physical activity. Because in-person physical activity interventions for obese AYA are challenging to implement and have high attrition rates, there is an urgent need to engineer targeted physical activity interventions that are effective and scalable for obese AYA with prediabetes and T2D. Aligned with NIDDK priorities, the long-term objective of the proposed research is to develop an intervention that reduces the risk of disease progression in youth with prediabetes and T2D. The applicant will take full advantage of the rich scientific environment of the Children’s Hospital of Philadelphia and University of Pennsylvania to achieve her research and training goals, supported by an expert multidisciplinary mentorship and advisory team. This K23 will support the PI’s training in mixed methods research, behavioral economics, mHealth, physical activity interventions, and biobehavioral clinical trial design and analysis. The goal of the proposed research is to develop an effective mobile health (mHealth) intervention to increase moderate to vigorous physical activity (MVPA) in AYA with prediabetes or T2D, harnessing insights from behavioral economics and leveraging a highly efficient framework to determine effective intervention components. Aim 1 will determine attitudes, beliefs, and norms about, as well as barriers and facilitators to, engaging in daily MVPA by conducting semi-structured interviews of AYA with prediabetes or T2D. In Aim 2, theory-based and empirically-informed text messages to promote MVPA will be developed through adaptation of adherence- promoting text message banks developed by the PI’s mentors, using focus groups with patient stakeholders and insights from Aim 1. Aim 3 will determine intervention component settings that effectively increase MVPA and will consist of a 12-week factorial trial (Behavioral Economics for Activity Motivation) of 4 intervention components targeting theoretical mediators of MVPA: 1) text messages targeting Theory of Planned Behavior concepts of attitudes and norms, vs. simple reminders; 2) ramping-up vs. fixed step count goals, targeting perceived behavioral control; 3) financial incentives allocated up-front and lost for unmet goals vs. earned, targeting loss aversion; and 4) daily vs. weekly goals, targeting hyperbolic discounting (preference for smaller-sooner over larger-later rewards). The primary outcome is change in MVPA. Secondary outcomes include steps/day, proportion achieving 60 minutes MVPA/day, body mass index, hemoglobin A1c, and fasting insulin and lipids. The PI and mentors hypothesize that effective component settings will include text messages targeting attitudes/norms, ramped goal, loss-framed incentives and daily goals. Through this K23, the PI will be poised to compete successfully for R01-level funding to conduct a randomized controlled trial of the optimized intervention, helping to launch her career as an independent physician-scientist with expertise in health behavior change in AYA with prediabetes and T2D. Ravy Vajravelu, M.S., M.D.

UNIVERSITY OF PENNSYLVANIA, PHILADELPHIA, PA

Evaluation of multiple medication exposures concurrently using a novel algorithm

PROJECT SUMMARY

The development of large observational health databases (OHD) has expanded the data available for analysis by pharmacoepidemiology research. The efficiency of these studies may be improved by simultaneously studying the association of multiple medications with a disease of interest. Unfortunately, prior research has demonstrated that it is difficult to distinguish true-positive from false- positive results when studying multiple exposures simultaneously, thus limiting the conclusions drawn from these types of studies and representing a major gap in the field. The objective of this proposal, which is the first step in achieving the applicant's long-term goal of improving the diagnosis and treatment of gastrointestinal diseases using insights derived from OHD, is to evaluate and validate medication class enrichment analysis (MCEA), a novel set-based signal-to-noise enrichment algorithm developed by the applicant to analyze multiple exposures from OHD with high sensitivity and specificity. The central hypothesis of this proposal is that MCEA has equal sensitivity and greater specificity compared to logistic regression, the most widely used analytic method for OHD, for identifying true associations between medications and clinical outcomes. The applicant will complete the following two interrelated specific aims to test the hypothesis: Aim 1 – to calculate the sensitivity and specificity of medication class enrichment analysis (MCEA) and logistic regression (LR) for identifying medication associations with Clostridium difficile infection (CDI) and Aim 2 – to calculate the sensitivity and specificity of MCEA and LR for identifying medication associations with gastrointestinal hemorrhage (GIH). The rationale for these aims is that by reproducing known medication-disease associations without false positives, MCEA can be used to identify novel pharmacologic associations with gastrointestinal diseases in future studies. The expected outcome for the proposed research is that it will demonstrate MCEA as a valid method for pharmacoepidemiology research, opening new research opportunities for the study of multi-exposure OHD. These new research opportunities may lead to more rapid identification of potential pharmacologic causes of emerging diseases and discovery of unanticipated beneficial medication effects, allowing such medications to be repurposed for new indications. To attain the expected outcome, the applicant will complete additional coursework that builds on his Master of Science in Clinical Epidemiology to learn computational biology, machine learning, and econometrics techniques. With the support of this grant and his institution, he will also directly apply these techniques to pharmacoepidemiology applications under the close mentorship of a carefully selected team of faculty with extensive experience in gastroenterology, pharmacoepidemiology, medical informatics, and mentoring prior K-award grant recipients. Through these activities, the applicant will develop the skills necessary to obtain NIH R01-level funding and become a leader in developing novel techniques for application to the epidemiologic study of gastrointestinal diseases.

Jason Van Batavia, M.D.

CHILDREN'S HOSP OF PHILADELPHIA, PHILADELPHIA, PA

Defining the role of CRH-expressing neurons in Barrington's Nucleus (BN) and Characterizing the other neuronal subpopulations in BN

PROJECT SUMMARY/ABSTRACT

The candidate for this K08 Mentored Clinical Scientist Research Career Development Award aims to become an expert and leader in the field of neuro-urology, specifically on furthering our understanding of the brain-bladder control of volitional voiding. Lower urinary tract symptoms (LUTS) and disorders of voiding are among the most common reasons for referral to the urologist and affect ~20% of school- aged children and >40% of adults over 40 years old. Despite this prevalence, the specific neuronal populations that control voiding remain largely unknown which limits therapeutic options. Recent advances in neuroscience, genomics and bioinformatics will now allow study of specific neuronal subpopulations and characterization of the cellular heterogeneity inherent to all brain and brainstem nuclei. The proposed research in this grant will be performed in primary mentor Dr. Steven Zderic’s laboratory using mice. Aim 1 will determine the effects of long-term activation of one specific neuronal subpopulation in the pontine micturition center (i.e., Barrington’s nucleus, BN) on voiding function. These specific neurons express corticotropin-releasing hormone (CRH) and these neurons in preliminary studies were shown to inhibit voiding, thus activation of these neurons is expected to cause infrequent voiding and lead to bladder wall remodeling from chronic holding of urine. Aim 2 will identify the time and temporal pattern of firing of these CRH-expressing BN neurons in relation to volitional voiding using fiber photometry in freely moving mice. Aim 3 will utilize single cell RNA-sequencing techniques to identify and characterize the neuronal subpopulations that reside in and around BN. The first two aims will help define the role of BN CRH-neurons and potentially create a novel murine model of bladder wall remodeling, while aim 3 will lead to the identification of novel neuronal subpopulations for future study which could potentially lead to new therapeutic targets for LUTS and voiding disorders. The candidate brings to this proposal his experience studying voiding disorders both from a clinical and basic science standpoint. This career development proposal aims to provide the candidate with required additional training that leverages the resource-rich environment of the Children’s Hospital of Philadelphia and University of Pennsylvania. The candidate will be guided by an experienced mentoring team at CHOP [Dr. Zderic – Direct of Pediatric Urology Research; Dr. Seema Bhatnager – Associate Professor, neuroscientist; and Dr. Deanne Taylor – Director of Bioinformatics]. An Advisory Committee of internationally recognized leaders will provide content expertise and career counseling to ensure the candidate has a successful transition to independence as a surgeon-scientist. All together this research and training plan will provide the candidate with a foundation in advanced neuroscience, genomic, and bioinformatics necessary for achieving his long-term goals of understanding how specific brain and brainstem neuronal populations control voiding and finding ways to translate this knowledge into clinical improvement for patients with LUTS and voiding disorders. Michelle Van Name, M.D.

YALE UNIVERSITY, NEW HAVEN, CT

Effect of adiposity on hepatic and peripheral insulin resistance and hepatic steatosis in pubertal adolescents with type 1 diabetes

PROJECT SUMMARY/ABSTRACT

The purpose of this research study is to elucidate how the insulin resistance of obesity affects glucose and fat metabolism and hepatic fat deposition in pubertal adolescents with type 1 diabetes (T1D). With improved glycemic control and increased rates of obesity nationally, obesity is now prevalent in youth with T1D. Understanding how obesity affects metabolism in T1D has implications for treatment decisions. This knowledge will provide insights into potential adjunctive therapy interventions that may be applied in the future to improve insulin resistance and mitigate weight gain, both which would vastly thus improve management of T1D patients who have obesity. The specific aims of this project are to assess the effects of adiposity and puberty on resistance to insulin's ability to suppress hepatic glucose production, stimulate peripheral glucose metabolism, and suppress rates of lipolysis in pubertal youth with T1D and to examine the role of fatty liver disease on insulin resistance in adolescents with obesity and T1D. The first aim will be conducted using the stepped, hyperinsulinemic- euglycemic glucose clamp technique with stable isotope tracer infusions to assess changes in endogenous glucose production, peripheral glucose uptake, and glycerol turnover in response to graded increases in rates of insulin infusion. The second aim will assess the impact of fatty liver disease and fat distribution on the insulin resistance of obesity in adolescents with T1D using abdominal MRI. The clamp and MRI studies will be carried out in 36 obese and 36 lean pubertal adolescents with T1D and the clamp study will also include 36 lean young adults with T1D. Carrying out the proposed program of research and analyzing the results, under the guidance of my outstanding mentorship team, will further my development into an independent pediatric physician-scientist. I will gain expertise in sophisticated measures of glucose and fat metabolism, as well as advanced skills in all aspects of study design, implementation, and evaluation and complete coursework in isotope tracers, biostatistics, and grant writing. The academic environment at Yale School of Medicine is rich in intellectual stimulation and houses ample resources critical to the completion of this proposal. Results obtained will reveal intricacies of metabolism in youth with T1D and become essential components of the NIDDK mission to “improve people's health and quality of life” by “disseminat[ing] science-based information on diabetes.” Furthermore, the results obtained will be critical in the preparation of my initial R01 application to evaluate the impact of adjunctive medications to decrease weight and insulin dose while improving glycemic control and outcomes in youth with obesity and T1D. Silvia Vilarinho, M.D.

YALE UNIVERSITY, NEW HAVEN, CT

Deciphering the Genetic Basis of Portal Hypertension

PROJECT’S SUMMARY/ABSTRACT

Idiopathic liver disease remains a major challenge in both pediatric and adult hepatology, representing a high unmet medical need. Idiopathic portal hypertension (IPH), the focus of this proposal, is a prototype of a poorly understood liver disease. Portal hypertension is a silent clinical syndrome defined by portal venous system pressure that is at least 5 mmHg higher than the pressure in the inferior vena cava, but its complications represent the leading causes of liver-related death in the general population. IPH is diagnosed when a liver biopsy excludes cirrhosis, and all other known causes of portal hypertension have been ruled out. IPH has been reported in infancy and childhood, and in some families more than one individual is affected. Unexplained childhood phenotypes are excellent candidates to uncover mutations in genes not previously implicated in disease. Advances in human genetics and genomics through next generation sequencing have created tremendous opportunities for exploring how the plays a role in disease. We demonstrated the utility of whole-exome sequencing (WES) in the diagnosis of pediatric liver diseases of unknown cause. Moreover, using WES, we identified a new Mendelian form of IPH due to a recurrent recessive mutation (p.N46S) in DGUOK, which encodes deoxyguanosine kinase. Interestingly, treatment of HIV-infected patients with the nucleoside analog didanosine causes non-cirrhotic portal hypertension in a subset of these patients and lowers DGUOK levels in vitro. Furthermore, we recently uncovered a novel bile acid synthesis disorder due to ACOX2 deficiency in a patient with a variant of IPH defined by incomplete septal cirrhosis; and our preliminary data shows that homozygous loss of function mutations in GIMAP5, encoding GTPase, IMAP family member 5, cause IPH. Based on these findings, we hypothesize that delineation of the genetic architecture of IPH will identify the genes and mechanisms underpinning inherited portal hypertension and these genes will also be relevant to a fraction of patients with common forms of portal hypertension. We will investigate this premise through the following three specific aims: (1) identify additional genes and pathways underlying unrecognized Mendelian forms of IPH; (2) determine the molecular mechanism(s) linking mutated genes (e.g. GIMAP5, ACOX2) to liver phenotype; (3) investigate the contribution of genetic variation in IPH-causing genes in drug-related non-cirrhotic portal hypertension in the general population. The study of rare Mendelian forms of common diseases, such as coronary artery disease, hyperlipidemia and hypertension, revealed to be highly informative regarding the general mechanisms of these conditions and development of new therapeutics. Thus, by investigating the genes implicated in rare cases of familial IPH, we expect to advance our understanding of portal hypertension pathogenesis, define new diagnostic tests for personalized management, identify novel therapeutic targets, and uncover novel pathways relevant to portal hypertension due to common liver diseases. Bo Wang, Ph.D.

UNIVERSITY OF ILLINOIS AT URBANA-CHAMPAIGN, CHAMPAIGN, IL

The Role of Lpcat3 and Phospholipid Remodeling in Intestinal Homeostasis

PROJECT SUMMARY

Intestinal homeostasis is controlled by a strict balance between cell proliferation in the crypts and cell shedding from the villi. Dysregulation of intestinal homeostasis is known to cause severe intestinal pathologies, including inflammatory bowel diseases (IBD) and gastrointestinal cancer. Although much progress has been made towards understanding how this complex epithelial system maintains homeostasis, whether and how lipid metabolism may influence the epithelial cells along the crypt-villus axis during homeostatic and diseased states has been largely unexplored. Recent studies from our lab has identified critical functions of lysophosphatidylcholine acyltransferase 3 (Lpcat3), a phospholipid (PL) remodeling enzyme, in intestine. Loss of Lpcat3 in intestine selectively reduces polyunsaturated phosphatidylcholine (PC) in membranes, leading to decreased membrane fluidity and curvature, and impaired lipid absorption. Our following studies discovered that Lpcat3 and PL remodeling also play important roles in intestinal stem cell (ISC) proliferation and the maintenance of homeostasis. However, the mechanisms by which Lpcat3 affects intestinal homeostasis are not clear. The primary aim of this proposal is to understand how Lpcat3 and phospholipid remodeling regulate ISC proliferation and intestinal homeostasis. Aim 1 will examine if Lpcat3 deficiency affects proliferation and differentiation of ISCs, and test if different PC species regulate crypt proliferation using ex vivo crypt organoid culture. Aim 2 will unravel the mechanisms by which loss of Lpcat3 promotes crypt proliferation. Aim 3 will determine if PC remodeling and cholesterol metabolism may contribute to intestinal tumorigenesis. The candidate has a background in lipid metabolism and cancer biology. His long term scientific goal is to unravel the molecular mechanisms underlying lipid metabolism and human diseases. To further prepare himself for his long-term research goal, he plans to seek training that will complement his existing technical skills and further develop his professional skills. UCLA has a highly collaborative environment ideal to this project and for him to achieve these goals. His mentor, Dr. Peter Tontonoz, is a highly respected scientist with expertise in the areas of nuclear receptors, inflammation and lipid metabolism. The applicant also has an advisory committee that consists of Dr. Martin G Martín, an accomplished gastrointestinal biologist and expert in intestinal stem cell biology, Dr. Steve Bensinger, an internationally recognized expert in lipid metabolism/mass spectrometry, and Dr. Stephen Young, a pioneer and expert in lipid metabolism in intestine. His mentor team has a detailed plan to facilitate his research progress and scientific career development. In summary, his educational and research experience together with a strong and supportive mentoring team make him an ideal candidate for this research project and the K01 award. Chia-shi Wang, M.S., M.D.

EMORY UNIVERSITY, ATLANTA, GA

A Novel Mobile Application for Childhood Nephrotic Syndrome Management

Project Summary/Abstract

This proposal is a multidisciplinary research and training plan to rigorously evaluate and refine a user- centered mobile application (app) for childhood nephrotic syndrome management and enable the applicant, Chia-shi Wang, to obtain the skills needed for an independent research career in kidney disease intervention development and implementation. The broad objectives of the research proposal are to improve self- management and clinical outcomes of childhood nephrotic syndrome, one of the most common chronic kidney diseases in children. The disease is characterized by a relapsing-remitting pattern in the majority of patients and has high rates of complications and treatment side effects. Vigilant home care is central to disease management, yet many families find it difficult to adhere with the demands. Our interdisciplinary team has built a novel mobile app (UrApp) to assist families with self- management. UrApp has excellent technical validity and was well-received by users in preliminary usability testing. In this proposal, we will conduct a pilot randomized trial in three pediatric sites among caregivers of children with newly diagnosed nephrotic syndrome to evaluate the efficacy of UrApp in improving behavioral and biologic outcomes (Aim 1). Concurrently, we will perform process evaluation to determine influences on the effectiveness of UrApp (Aim 2). Lastly, we will engage caregiver stakeholders to use the results of the clinical trial and process evaluation to refine UrApp and plan a future user-centered, full-scale clinical trial (Aim 3). The anticipated outcome is the development of a novel, pragmatic, technology-based intervention that is ready for full-scale testing in a user-centered clinical trial. This proposal is made possible by the applicant’s exceptional research environment. Emory’s Division of Pediatric Nephrology is one of the largest pediatric nephrology programs in the U.S. with many successful, extensive research collaborations with other pediatric institutions, ensuring the feasibility of the current proposal. A strong alliance between Emory University, Children’s Healthcare of Atlanta, and Georgia Institute of Technology enabled the developed of UrApp and provides multidisciplinary mentorship in clinical trial research, behavior science, implementation science, and biostatistics. The mentorship, along with the courses available at Emory, will provide exceptional training for the applicant. In addition, the research activities will provide Chia-shi Wang with hands-on training in qualitative research, mixed-methods analysis, process evaluation methodology, and stakeholder engagement methodology. These new skills will augment the applicant’s background in clinical practice, clinical trials research, and quantitative research with expertise in behavioral and implementation sciences. The successful completion of the proposal will therefore provide Chia-shi Wang with a distinctive skillset to enable an independent research career and produce one of the first evidence-based disease management tools for the childhood nephrotic syndrome population. Chunmei Wang, Ph.D.

BAYLOR COLLEGE OF MEDICINE, HOUSTON, TX

A TAP63 TRANSCRIPTIONAL AXIS CONTRIBUTES TO THE SEXUAL DIMORPHISM IN POMC NEURON FUNCTIONS AND ENERGY HOMEOSTASIS

Obesity is recognized as a major health issue due to its high prevalence and strong association with diabetes and other metabolic disorders. Female animals are more resistant to obesity than males, but the mechanisms for this sexual dimorphism remain elusive. In my previous studies, I first screened body weight-regulatory neural populations and found that pro-opiomelanocortin (POMC) neurons in female mice fire more rapidly than male POMC neurons, and female mouse brains express higher POMC transcripts. Further, these sex differences in POMC neurons were associated with higher expression levels of TAp63 (a transcription factor) and SRC1 (steroid receptor coactivator-1, a transcriptional coactivator) in female POMC neurons than in male counterparts. Pilot studies showed that TAp63 can activate POMC gene expression in cultured cells. Further, SRC1 mRNA was significantly reduced by TAp63 deletion in female mice, suggesting that SRC1 is a transcription target of TAp63. Importantly, deletion of TAp63 or SRC1 only in POMC neurons in mice regulates body weight in a sexually dimorphic fashion. Together, I developed a hypothesis that an estrogen-TAp63-SRC1 transcriptional axis contributes to the sexual dimorphism in POMC neuron functions and energy homeostasis. I will generate mice that lack TAp63 or SRC1 in POMC neurons, and characterize energy homeostasis and POMC neuron functions (activity and gene expression profile) among male mice, female mice with or without intact ovary (OVX- V), and female without intact ovary but with estrogen supplement (OVX-E). I will also examine whether estrogen stimulates TAp63 expression and whether TAp63 stimulates SRC1 expression. The proposed studies represent logical extensions to my previous work, and will advance our understanding about the fundamental biology for sex differences in body weight control, which may facilitate the development of gender-specific therapeutic strategies for obesity and associated metabolic diseases. In addition, this project will provide an ideal training opportunity to prepare me for an independent research career focusing on transcriptional mechanisms in the hypothalamus and their roles in metabolic control. Jeremy R Wang, Ph.D.

UNIV OF NORTH CAROLINA CHAPEL HILL, CHAPEL HILL, NC

Advanced sequencing as a novel diagnostic tool to discover strain-level variation and function of mucosal-adherent bacteria contributing to IBD

Metagenomic analysis of the gut microbiome continues to provide critical insights into the function of microbiota in inflammatory bowel diseases (IBD). In contrast to luminal and fecal samples, the mucosa- associated microbiome is thought to be more directly relevant to host immune response and disease state. However, 16S profiling does not permit low-level taxonomic inference or characterization of functional potential and mucosa-associated microbiota are not amenable to traditional whole- metagenome sequencing due to prohibitively high host DNA. There is a critical need to develop novel sequencing and analysis methods that enable unbiased metagenomic sequencing of tissue-associated microbiota in complex host-microbiome systems. I will use a novel host-depleted metagenome sequencing approach to define the compositional and functional differences between mucosal, luminal, and fecal microbiota, and between healthy and disease states in unprecedented detail. My long-term goal is to establish an independent research program in computational genomics for human disease and personalized medicine focused on the gut microbiome and IBD. The central hypothesis of this proposal is that metagenomic sequencing of mucosa-associated microbiota will identify location-specific, species- and strain-level composition and functional variation associated with intestinal inflammation and human disease pathogenesis. I recently developed a novel sequencing and informatics protocol that interfaces with existing nanopore sequencing technology to enable dynamic selection and identification of species or genes from a metagenomic sample. This approach can be used to dynamically filter out DNA sequences belonging to previously observed microbial species or contaminating host genome. I will apply this method to perform the first effective high-depth shotgun sequencing of mucosa-associated microbiota in the ileum and colon of Il10-/- and wild-type mice. Using these data, I will compare host-depleted deep sequencing to traditional short-read shotgun sequencing and 16S rRNA sequencing for assaying composition and function of adherent communities. I will identify relative differences in taxonomic and genic abundances associated with colitis in a mouse model, including species- and strain-level variants that are not captured by existing approaches. I will also use this approach to determine whether adherent-invasive Escherichia coli (AIEC) selectively colonize the mucosal surface relative to the lumen in germ-free Il10-/- mice, supporting their role as causal pro-inflammatory agent in a mouse model of colitis. Lastly, I will assess variation in the mucosa- associated microbiome in colon biopsy samples from IBD and non-IBD patients to characterize disease behavioral phenotypes, potentially leading to novel diagnostic and therapeutic tools. Liheng Wang, Ph.D.

COLUMBIA UNIVERSITY HEALTH SCIENCES, NEW YORK, NY

TOX4 integrates hormone signaling on hepatic glucose and lipid metabolism

Project Summary

One in every 10 Americans is diagnosed with diabetes, and 90% of them have type 2 diabetes (T2D) due to insulin resistance and beta cell failure. Prevention and treatment of T2D remain an unmet medical need. The liver produces glucose during fasting to maintain normal blood glucose levels. This process is dysregulated in T2D, when resistance to or lack of insulin increases glucose production, resulting in hyperglycemia and its complications. Insulin regulation of hepatocyte glucose production (HGP) requires rapid changes in gene expression. FoxO1 plays an essential role in the transcriptional regulation of HGP. FoxO1 ablation impairs, but does not completely abolish the regulation of glucose production by insulin, likely due to incomplete suppression of phosphoenolpyruvate carboxykinase (Pck1) gene expression. These data highlight the heterogeneity of transcription factors (TFs) in regulating glucose production. The PI has recently identified TOX High Mobility Group Box Family Member 4 (TOX4) as a novel insulin- regulated TF, using DNA affinity purification from the Pck1 promoter and mass spectrometry (MS) in primary hepatocytes and liver samples. However, the metabolic function of TOX4 is unknown. In preliminary data, she shows that TOX4 is a transcriptional activator of Pck1 and glucose-6-phosphatase (G6pc). Knockdown of Tox4 in primary hepatocytes reduces glucose production by repressing these two genes. Moreover, in vivo KD of Tox4 in liver using adenovirus-encoded shRNA significantly inhibits gluconeogenesis and improves glucose tolerance and insulin sensitivity in C57BL/6J mice, while simultaneously promoting lipogenesis. These changes are more pronounced in diet-induced-obese (DIO) and diabetic db/db mice. Therefore, she hypothesizes that TOX4 regulates glucose and lipid metabolism in response to hormones. In this application, she will generate mice to ablate TOX4 in hepatocytes and critically investigate the function of TOX4 in hepatic glucose and lipid metabolism (Aim 1). She will further examine the mechanisms underlying hepatic TOX4 activation and regulation of its downstream targets (Aim 2). Successful completion of this application will illustrate the role of a new TF in regulating glucose and lipid metabolism in the liver in response to insulin. More importantly, these proposed studies will determine whether TOX4 is a potential therapeutic target in the treatment of insulin resistance and T2D. Jaycob Warfel, Ph.D.

LSU PENNINGTON BIOMEDICAL RESEARCH CTR, BATON ROUGE, LA

HuR as a Regulator of Skeletal Muscle Metabolism

PROJECT SUMMARY/ABSTRACT

The ability to switch between metabolic substrates based on their availability, metabolic flexibility, requires a complex network of molecular interactions for efficient function. We have found that one aspect of this network is the control of mRNA transcript levels by HuR. HuR is an RNA-binding protein that regulates levels of RNA transcripts and their translation within cells. Though HuR has been extensively studied for its roles in proliferation, inflammation, and apoptosis, its action as a metabolic sensor is relatively unknown. Our preliminary data indicate that skeletal muscle specific HuR knockout mice show increased insulin resistance and adiposity relative to control animals. Interestingly, these mice show increased levels of HuR target transcripts, while also showing decreased translated protein levels for these transcripts. The poor metabolic prognosis resulting from down regulation of HuR in skeletal muscle also complements data we have collected for metabolically unhealthy male and female human skeletal muscle samples. The proposed aims will therefore test the hypothesis that delivery of HuR target transcripts to the ribosome promotes metabolic flexibility by influencing the mTOR signaling pathway. Very important is the new methodological techniques that Dr. Warfel will learn in order to develop his career as an independent investigator focused on RNA biology and metabolism. Methods in this proposal include transgenic mouse model generation and phenotyping, use of metabolic chambers and radioactive metabolite assays, bioinformatics and molecular biology. The training in these techniques will also provide the preliminary data necessary to develop future projects aimed at understanding the contribution of RNA binding proteins to cellular metabolic homeostasis, and to consider new potential avenues in the treatment of insulin resistance and obesity. This is Dr. Warfel’s long-term research expectation, and he has assembled an outstanding team of mentors and collaborators with extensive experience in areas relevant to this proposal who will provide guidance to ensure that Dr. Warfel achieves independence. Zong Wei, Ph.D.

MAYO CLINIC ARIZONA, SCOTTSDALE, AZ

A role of chromatin remodeling complexes in beta cell dysfunction

Project Summary/Abstract

cell dysfunction, induced by inflammation and other types of stress, is critical in pathogenesis of type 2 diabetes (T2D). Accordingly, strategies designed to protect cells from dysfunction could be attractive for combating diabetes. The candidate’s previous study identified that a ligand-dependent switch between vitamin D receptor (VDR)-associated bromodomain readers BRD9 and BRD7 regulates the anti- inflammatory response in cells. A combination of VDR activation and BRD9 inhibition promoted VDR- BRD7 association, suppressed inflammation in cells, and improved glucose homeostasis in multiple mouse T2D models. BRD9 and BRD7 belong to BAF and PBAF, respectively, two overlapping but functionally distinctive SWI/SNF chromatin remodeling complexes. Therefore, chromatin remodeling complexes may play essential roles in cell dysfunction in T2D. The hypothesis of this proposal is that the balance between BAF and PBAF complex, modulated by BRD9/BRD7, controls the cell epigenome and metabolic homeostasis. To investigate the gene-regulatory mechanisms associated with cell dysfunction, the studies proposed here will combine genomic, molecular and genetic approaches to pinpoint the role of SWI/SNF complexes in regulating the cell epigenome and function in T2D, in following specific aims: Aim 1: determine SWI/SNF complex balance in cells exposed to inflammatory cues by charting the genome distribution of key BAF and PBAF complex components; Aim 2: characterize SWI/SNF-mediated chromatin accessibility dynamics in shaping cell super enhancer activity; Aim 3: use cell specific knock-out mouse models to determine the roles and downstream targets of BRD9/BAF in vivo. Collectively, these studies may reveal how a fine-tuned balance between two competing chromatin remodeling complexes controls cell function in T2D, and may lead to novel strategies for the development of next generation anti-diabetic therapies directly targeting cell dysfunction. The candidate’s goal for the next three years is to develop an independent research program in the area of diabetes, nuclear receptors, and epigenetics, and to transition into an academic faculty position. He has extensive background training in transcription, epigenetics and metabolism. In the laboratory of Dr. Ronald Evans at the Salk Institute he continues to gain experience to understand transcriptional regulation and how it controls cell physiology. Dr. Mark Huising, an islet biologist, will serve as co- mentor and provide additional training in cell physiology and imaging. The candidate will continue to collaborate with his co-mentor, Dr. Diana Hargreaves, to acquire expertise in biochemical and genomic assays related to chromatin remodeling. The candidate’s long-term career goal is to become a leading independent biomedical researcher at an academic institution, investigating the molecular basis of diabetes and inflammation. This award will also allow him to develop his own research niche and distinguish himself from his current mentor. Spencer Willet, Ph.D.

WASHINGTON UNIVERSITY, SAINT LOUIS, MO

The role of the Hippo Pathway in gastric homeostasis and metaplasia

PROJECT SUMMARY

In this K01 application, I will get mentorship and protected research time to grow my skill repertoire to pursue becoming a principal investigator in the field of gastric epithelial biology. My career development will be nurtured through weekly meetings with Dr. Mills and Dr. DiPaolo, through convening an experienced senior advisory committee, and opportunities for professional development through WashU and the NIH. Skill development focus will include, but are not limited to, manuscript/grant preparation, seminars and courses described in the career development section, and presentations at international conferences (DDW, FASEB, etc.). Training includes coursework in biostatistics, workshops on bioinformatics, continued training in bioethics, and participation diverse seminar series that will include me giving presentations, interacting with visiting outside speakers, and attending journal clubs. The overarching hypothesis of this project is the following: The Hippo pathway is a central regulator of gastric epithelial homeostasis and response to injury. We propose to address these Specific Aims with regards to the role of the Hippo pathway in the gastric epithelium: 1) To address the sufficiency/necessity of the Hippo pathway in maintaining normal, mature gastric epithelium cell identity during homeostasis and following injury and the molecular pathways the interact with the Hippo pathway in driving acute dedifferentiation phenotypes (gene expression, cell shape, cell proliferation). 2) To describe the role of the Hippo pathway in gastric organoid growth and stemness. We will also address discovery of novel regulated and direct targets of YAP1/TAZ in this in vitro metaplasia model. Jessie Wong, Ph.D.

STANFORD UNIVERSITY, STANFORD, CA

Family Matters: Optimizing Family-Based Interventions for Adolescents with Type 1 Diabetes

Project Summary/Abstract

Adolescents with type 1 diabetes (T1D) often experience deteriorations in glycemic control that can persist into adulthood. Although family-based interventions are empirically supported to offset this adolescent risk, such behavioral health services are rarely included in diabetes care. This K23 proposal seeks to bridge this gap between research and practice by optimizing family-based programs to provide the most efficacious, judicious, and streamlined intervention possible to adolescents with T1D. The proposed study aims to 1) pilot and evaluate specific family-based intervention components, and 2) identify the factors that explain and influence intervention effects. First, the candidate will use qualitative methods to pilot 3 intervention modules to assess feasibility and acceptability. Each module corresponds to one of 3 critical family processes: parental monitoring, parental involvement, and parent-adolescent conflict, each of which are empirically linked to adolescent diabetes management and glycemic control and are common family-based intervention targets. Next, the candidate will conduct an experimental trial of the intervention modules to evaluate their relative efficacy as well as combined impact on adolescent outcomes. Finally, the candidate will conduct advanced statistical analyses to verify the specific family processes (including both general and diabetes-specific) that explain (mediate) program effects as well as the contextual factors that influence (moderate) intervention effects. This proposal will yield an optimized family-based intervention as well as provide critical empirical information about the causal pathways between family functioning, mental health, and glycemic control among adolescents with T1D. The K23 award offers the training, mentorship, protected time, and resources necessary to complete the proposed study and support Dr. Wong’s progress toward becoming an independent research scientist in pediatric diabetes. Karen Woo, Ph.D., M.D.

UNIVERSITY OF CALIFORNIA LOS ANGELES, LOS ANGELES, CA

Outcomes of Vascular Dialysis Access in the Elderly

Project Summary/Abstract

This application proposes a career development and research plan aimed to develop me, Karen Woo, MD, MS into an independent clinician/scientist specializing in vascular surgery outcomes research and comparative effectiveness research. I am a board-certified vascular surgeon with a clinical and research interest in dialysis vascular access. I have prior clinical research experience in the subject of dialysis fistula maturation, patency and management of complications. I have created a unique collaboration with the Schaeffer Center for Health Policy and Economics at the University of Southern California which has extensive experience with outcomes research and comparative effectiveness research and is especially well suited to serve as a training site. The career development plan will consist of advanced coursework in biostatistics, clinical trial design, multidisciplinary conferences and project-based learning. The proposal will be conducted under the direction of the mentorship committee. The primary mentor will be Dana Goldman, PhD, who holds the Leonard D. Schaeffer Director's Chair at the Schaeffer Center and is also Professor of Public Policy, Pharmacy and Economics in the Sol Price School of Public Policy and the School of Pharmacy. The advisory committee is comprised of Michael Allon, MD, Professor of Medicine at the University of Alabama, Robert J. Hye, MD, Chief of Vascular Surgery at Kaiser Permanente Hospital, San Diego, Clifford Y. Ko, MD, Professor of Surgery at UCLA and Peter F. Lawrence, MD, Professor of Surgery at UCLA. In the United States, the Fistula First Breakthrough Initiative, the National Kidney Foundation Dialysis Outcomes Quality Initiative Guidelines and the Center for Medicare Services End Stage Renal Disease Quality Incentive Program all encourage the creation of fistulas over grafts for hemodialysis vascular access. However, the elderly are the fastest growing segment of the end stage renal disease population and recently, evidence has emerged suggesting that fistulas may not provide the same benefits over grafts in the elderly population as in the younger population. The research described in this proposal utilizes outcomes research techniques to evaluate short term outcomes of fistulas vs grafts for dialysis vascular access in the elderly compared to non-elderly and determine factors which may aid in patient selection to optimize outcomes. Kyle Wood, M.D.

UNIVERSITY OF ALABAMA AT BIRMINGHAM, BIRMINGHAM, AL

Obesity and Endogenous Oxalate Synthesis

Project Summary/Abstract

This proposal will enable advancement of my research career under the guidance, experience, and tutelage of successful and effective mentors. In addition, my research knowledge will be augmented by training in human studies, animal work and the complex metabolic analytical skills. This proposal will give me the tools necessary to become an independent investigator. Finally, the proposed study provides me the opportunity to continue my current research endeavors, specifically looking at kidney stone disease, endogenous oxalate synthesis, and the role of obesity. The prevalence of kidney stone disease linearly increased in the U.S. over the last several decades, now afflicting 10-15% of the population. The etiology of kidney stone disease is multifactorial involving lifestyle factors, genetics, diet, and environment. Multiple medical comorbidities have been linked to kidney stone disease including obesity. One component of the most common type of stone composition is oxalate, an end product of metabolism. Small increases in urinary oxalate can increase calcium oxalate crystal formation and thus stone disease. Multiple epidemiological studies have demonstrated a positive correlation between obesity and urinary oxalate excretion. Yet, little is known about the underlying reason for this increase in urinary oxalate. Urinary oxalate levels are affected by both a dietary and endogenous component and each is felt to contribute equally. Endogenous oxalate synthesis has been previously thought to occur primarily in the liver and its major source is glyoxylate. The biochemical pathways involved in oxalate production are poorly understood despite extensive research. The central hypothesis is that the increase in urinary oxalate seen in obesity is derived from increased endogenous production. Further, it is proposed that obesity influences the metabolic processes within the liver, resulting in increased oxalate synthesis. The hypotheses will be tested by pursuing two specific aims: 1) Evaluating oxalate synthesis in a lean and a diet-induced obese animal model, 2) To demonstrate that obese humans have increased endogenous oxalate synthesis on controlled diets. I will apply our laboratories expertise in controlled dietary studies in both humans and mice, and utilize complex analytical technologies, including mass spectrometry based assays. The proposed study may provide new insights regarding the role of obesity and fat distribution on endogenous oxalate production and thus calcium oxalate kidney stone disease. It will facilitate my transition into an independent and productive NIH funded investigator. Bridget Young, Ph.D.

UNIVERSITY OF ROCHESTER, ROCHESTER, NY

Developmental infant effects of exposure to high doses of oral insulin in human milk

PROJECT SUMMARY/ABSTRACT

1 This proposal utilizes cutting-edge mechanistic approaches to investigate the clinically meaningful but hard-to- 2 study question of HOW insulin in human milk affects infant development. This research question is particularly 3 relevant to the growing and under-studied population of infants receiving breast milk from obese and/or insulin 4 resistant (IR) mothers - who produce milk with significantly greater insulin concentrations. Animal models show 5 that oral insulin delivered to the infant via breast milk has far-reaching effects on normal offspring development 6 and maturation. This proposal documents these effects in human infants and investigates how effects may be 7 altered in the context of maternal IR - and thus sustained infant exposure to high concentrations of oral insulin. 8 Specifically, we examine systemic effects regulated by infant pancreatic function as well as local intestinal 9 effects. Importantly, we will document both acute effects during the neonatal period when organs are still 10 immature AND effects of more “chronic” exposure. We will recruit 64 exclusively breastfed infants of both 11 normal weight/normoglycemic (NW; n=28) and IR (n=36) mothers and study them during the neonatal period 12 (2-4 weeks) and after more “chronic” exposure to mother’s milk (5 months). Our aims are: 13 Aim 1 – Cross-Sectional Study – Investigate differences in metabolic development and intestinal maturation 14 between infants exclusively breastfed by NW vs IR mothers at two critical time points: 2-4 weeks and 5 15 months. We hypothesize that infants breastfed by IR mothers will exhibit: an altered metabolomic profile 16 indicating differences in carbohydrate handling and insulin signaling (H1.1); an altered microbial composition in 17 the intestinal microbiome (H1.2); increased relative transcript abundance of insulin-target genes in exfoliated 18 intestinal cells, indicating more active insulin signaling at the level of the enterocyte (H1.3); and decreased 19 intestinal permeability (a functional measure of intestinal maturation) at 2-4 weeks (H1.4). 20 Aim 2 – Glycemic Study - Determine if consumption of human milk from an IR mother (with high concentrations 21 of insulin) is correlated with differences in endogenous pancreatic response at 2-4 weeks. We hypothesize that 22 receipt of human milk with high insulin will result in a lower endogenous pancreatic insulin response (H2.1). 23 Aim 3 – Glucose Tolerance Study - Determine if chronic consumption of human milk from an IR mother (with 24 high concentrations of insulin) is correlated with pancreatic response to a glucose challenge at 5 months. We 25 hypothesize that chronic exposure to human milk from an IR mother will under- stimulate pancreatic β-cells and 26 result in a dampened endogenous insulin response to a standard glucose challenge at 5 months (H3.1). 27 Our long term objective is to fully understand the impact of oral insulin in the development and postnatal 28 programming of the breastfed infant. As the prevalence of insulin resistance rises among mothers – and as 29 these women’s offspring have increased risk of developing metabolic disease themselves – this work has far- 30 reaching implications regarding how we counsel women and care for their infants to minimize disease risk. Noelle Younge, M.D.

DUKE UNIVERSITY, DURHAM, NC

The Microbiome and Metabolism in Postnatal Growth Failure

ABSTRACT

The purpose of this Mentored Patient-Oriented Research Career Development Award (K23) is to provide Dr. Noelle Younge with the mentorship, training, and research experience necessary to become an independent physician-scientist and leader in understanding the preterm infant microbiome and its impact on early life growth failure. To facilitate her transition to independence, Dr. Younge and her mentors have developed a comprehensive research and career development plan that includes mentorship from an exceptional team of scientists with proven track records of successful mentorship and extensive expertise in microbial regulation of host metabolism and physiology; intensive didactic training; and a research plan that is purposefully designed to provide experiential learning in advanced research methods to study the microbiome and metabolome of preterm infants. Postnatal growth failure is a pervasive problem among preterm infants that is often refractory to modern nutritional management and portends poor long-term neurodevelopmental outcomes. Novel approaches to reduce the burden of early life growth failure are needed to improve the health and developmental outcomes of preterm infants. Recent work from Dr. Younge and her mentors demonstrates that extremely preterm infants with postnatal growth failure have disrupted maturation of the intestinal microbiota and host metabolome. In the research plan outlined in this K23 proposal, Dr. Younge will expand upon this work by 1) identifying metabolic functions of the microbiome that differentiate preterm infants with growth failure from preterm infants with appropriate postnatal growth, and 2) investigating the nature of the relationships between the microbiota, metabolism, and growth under controlled conditions in a gnotobiotic mouse model. This work will produce novel insights into the pathogenesis of postnatal growth failure in preterm infants and will serve as the foundation for Dr. Younge to build an independent research program conducting clinical and preclinical studies directed toward improving the health and development of preterm infants. In completing these aims, Dr. Younge will develop a unique and specialized skill set in the integration of multi- omics data and translational research that will facilitate her transition to an independent research career. Melody Zeng, Ph.D.

WEILL MEDICAL COLL OF CORNELL UNIV, NEW YORK, NY

Role of gut microbiota-induced IgG in enteric host defense

Project Summary/Abstract

The gut microbiota has a plethora of important functions that promote human health. The impact of maternal gut microbiota on neonatal health, however, remains largely unknown. Sepsis is an important cause of morbidity and mortality among newborn infants, particularly in preterm infants. Our preliminary studies have unraveled a previously unknown function of the gut microbiota to induce antigen-specific IgG antibodies under homeostatic conditions, which can rapidly mediate clearance of pathogens bearing conserved IgG antigens during systemic infection. Importantly, we found robust levels of commensal-specific IgG antibodies in both mouse and human breast milk. Therefore, our central hypothesis is that 1) maternal commensal IgG confers critical protection to the neonate against infection; 2) maternal with IgG-inducing beneficial gut symbiotic bacteria, or IgG-inducing antigens, may be a novel approach to treat neonatal sepsis, which can be caused by dissemination of gut bacteria. This hypothesis will be tested through three specific aims: 1) determine the role of maternal gut microbiota-induced IgG in neonatal enteric infection, 2) identify and characterize gut symbiotic bacteria that induce homeostatic antigen-specific IgG antibodies, and 3) determine whether maternal immunization with IgG-inducing bacteria or antigens confers protection in the neonate. The objective of the current application is to harness gut microbiota-induced IgG to develop therapeutics for neonatal sepsis, and for the PI to learn new skills such as developing neonatal infection models and vaccine nanoparticles, which are important for her to develop an independent research program with a focus on the gut microbiota and neonatal health. The research is significant because these studies will unravel potentially novel and effective approaches to treat neonatal sepsis. The distinguished mentoring team will allow the PI to complete the proposed studies with new skills and transition to independence in a new field of study. In addition, the excellent research environment of the University of Michigan will provide exceptional support for the proposed studies and the PI's career development. Dong Zhou, Ph.D., M.D.

UNIVERSITY OF CONNECTICUT SCH OF MED/DNT, FARMINGTON, CT

Early and Transient Activation of Fibroblast Promotes Tubule Repair after Acute Kidney Injury.

PROJECT SUMMARY/ABSTRACT

Career Development Plan My primary career goal is to become a successful, independent investigator and leader in the field of kidney disease. To achieve my career goal, I have assembled an advisory committee from a multi-disciplinary group of established researchers at the University of Pittsburgh (Pitt). These researchers are experts in the fields of bioinformatics, pathology, cell biology, immunology, and nephrology. My career development plan includes personal mentoring, focused coursework, practical research experience, and professional training. Pitt is one of the nation’s most distinguished, comprehensive universities and a major center of biomedical research national wide. It is committed to fostering the careers of research faculty and maintains a strong and well- established health sciences research program. All these factors establish a positive environment in my career development towards independence. Research Plan Acute kidney injury (AKI) is an abrupt or rapid decline in renal filtration that happens within a few hours or a few days. Most of the work in the field focuses on renal tubule damage, but research on repair of the tubules and what process promotes surviving tubular epitheliums to dedifferentiate is lacking. Cellular events involved in the early phases of AKI and the triggers or sources responsible for tubule dedifferentiation remain unclear. As the cell neighbor to renal tubules, we believe activated fibroblasts play a main role in inducing renal tubule repair after AKI. Our recent preliminary studies show that multiple fibroblast phenotypes were activated as early as 1 hour and reach peak at 12 hours after AKI, which is far earlier than tubular epithelium proliferation. We previously recognized that in chronic kidney disease (CKD), a tubule-derived novel growth factor, Sonic Hedgehog (Shh), specifically targets interstitial fibroblast, driving renal fibrosis through epithelial- mesenchymal communication (EMC). In our AKI mouse model, Shh was also directly secreted by renal tubules and was upregulated as early as 1 hour in injured kidneys. To our surprise, compared to its role in CKD, Shh plays a completely opposite role in AKI; it has a protective effect in AKI. Pharmacological inhibition of Shh suppressed fibroblast activity and aggravated AKI. In cultured fibroblasts, Shh causes transient fibroblast activation and secretion of hepatocyte growth factor (HGF), which we reported to have a renoprotective role in AKI. Therefore, our central hypothesis is that renal tubule-derived Shh induces early and transient fibroblast activation to promote AKI repair through a Shh-HGF feedback loop. We will test this hypothesis in two specific aims: 1) Determine the mechanism of Shh-mediated EMC in promoting renal repair after AKI. 2) Determine the roles of the Shh-HGF feedback loop in renal repair after AKI. Fully understanding the early stages of AKI pathogenesis will be very beneficial in determining AKI prognosis and designing novel future therapeutic strategies. Yeyi Zhu, Ph.D.

KAISER FOUNDATION RESEARCH INSTITUTE, OAKLAND, CA

Fetal Programming of Growth and Obesity: A Metabolomics Approach

PROJECT SUMMARY/ABSTRACT

This K01 Career Development Award proposes a multidisciplinary 4-year training program to provide the candidate, Dr. Yeyi Zhu, with the experience and resources necessary to launch a successful career as an independent investigator to elucidate early origins of obesity and related comorbidities. The childhood obesity epidemic remains an urgent public health priority. Early prevention is critical to stemming the tide of obesity. Yet, our ability to identify promising prevention targets has been impeded by the difficulty in measuring the holistic metabolic status by conventional tools. Several human studies, including from Dr. Zhu's previous work, indicate that individual exposures including dietary factors in pregnancy may program obesity risk in later life. However, the underlying metabolic pathways are elusive. To fill this important knowledge gap, Dr. Zhu will use a novel holistic framework that leverages both targeted and untargeted metabolomics approach, the state-of-the-art electronic health records (EHR) data, and bioinformatics analytics to investigate the mechanisms by which the in-utero environment may infer risk of fetal growth extremes [small for gestational age (SGA) or large for gestational age (LGA)] and altered infant growth trajectories and excess adiposity (weight-for-length or body-mass-index-for-age z-score ≥85th percentile). This application takes advantage of the Pregnancy Environment and Lifestyle Study within Kaiser Permanente Northern California, with unique, robust resources of fasting serum specimens collection in early to mid-pregnancy, anthropometric measurements, multi-domain survey data (e.g., diet, physical activity, psychosocial assessments), and EHR data throughout the gestation and offspring infancy. The specific aims, to be examined in a sample of 150 LGA, 150 SGA, and 150 appropriate for gestational age births, are to: examine the associations between candidate (branch-chain amino acids, myo-inositol, trimethylamine N-oxide, acylcarnitines, and fatty acids) and untargeted metabolites in early to mid-pregnancy with fetal growth extremes (Aim 1) and infant growth trajectories and excess adiposity from 0-2 years (Aim 2), and explore metabolomic signatures for dietary factors in utero (Aim 3). Study findings may elucidate the underlying metabolic pathways and potential upstream preventive targets related to modifiable in-utero exposures, such as maternal dietary factors, to mitigate childhood obesity. Dr. Zhu is well suited to perform this research: 1) she has a solid foundation in epidemiology, nutrition, and biostatistics; 2) this K01 will significantly broaden her repertoire of advanced training in metabolomics, bioinformatics, and related biological interpretation; and 3) she has leveraged a carefully coordinated set of resources including a multidisciplinary mentorship committee, coursework, and applied learning closely aligned with the training objectives and specific aims. This K01 is essential to advance her long-term career objective of becoming an independent investigator with expertise in omics and bioinformatics to study early origins of cardiometabolic disease, with the ultimate goal to inform individualized care for upstream prevention of obesity and its comorbidities.

Bernadette Zwaans, Ph.D., M.S.

WILLIAM BEAUMONT HOSPITAL RESEARCH INST, ROYAL OAK, MI

Cancer Survivorship Research and Training in Radiation Cystitis

Cancer survivors are at risk for developing radiation cystitis (RC) after radiation therapy. RC is a debilitating bladder and may be life-threatening. Current therapies are inadequate and can have severe life changing side-effects. The absence of reliable treatment and early diagnostic markers is in part due to limited comprehension of the histological and molecular changes associated with the progression of this condition. I hypothesize that radiation-induced damage to the bladder vasculature drives the chronic inflammation, fibrosis and hematuria associated with RC. This hypothesis will be addressed through three specific aims: 1. Determine the effect of radiation therapy on bladder vasculature function, 2. Assess and target the inflammation in an animal model of RC, and 3. Validate urinary candidate biomarkers for early detection of RC. The study design includes assessing functional changes of endothelial cells and determining changes in ICAM-1 expression and its role in immune cell recruitment in response to irradiation. Non-irradiated and cyclophosphamide treated cells will serve as negative and positive controls respectively. Using a mouse model of RC, a timeline of molecular and histological bladder changes in response to radiation will be determined. Non-irradiated bladders and cyclophosphamide-induced cystitis in mice will be used as negative and positive controls for these studies respectively. Finally urine samples from RC patients with a history of prostate or cervical cancer will be used to develop a biomarker for RC. Controls consist of age-matched male and female urine samples. As a cancer survivor and a scientist, my ultimate career goal is to improve the quality of life of cancer survivors through my research. I plan on achieving this goal by becoming an independent translation investigator, focusing my research on urological complications from cancer therapies, and implementing my research findings into the clinic. Through my past research track record, I have extensive experience in basic science techniques in vascular and cancer biology. The additional necessary skills to successfully complete this research and training proposal will be acquired through mentorship and coursework in translational research, radiation, and biostatistics. In addition, I will be receiving practical training in viral infections and pathology. I will be receiving continuous mentorship from Drs. Chancellor and Lamb. Drs. Kanai and Wilson will serve as co-mentors. My mentors have a history of successful mentorship of NIH scientists and we have a superb and supportive training environment here at Beaumont Research Institute and Oakland University William Beaumont School of Medicine. The outcome of our studies can significantly improve the quality of life of many cancer survivors that are suffering with severe bladder complications due to radiation therapy. These studies could lead to finding effective therapy for RC as well as a urine biomarker for early detection of RC. This research study has great potential to improve the quality of life of cancer survivors suffering from this debilitating condition.