PEA15) Form a Molecular Switch Governing Cellular Fate Depending on the Metabolic State

Total Page:16

File Type:pdf, Size:1020Kb

PEA15) Form a Molecular Switch Governing Cellular Fate Depending on the Metabolic State Mitochondrial hexokinase II (HKII) and phosphoprotein enriched in astrocytes (PEA15) form a molecular switch governing cellular fate depending on the metabolic state Philipp Mergenthalera,b,c,d,1,AnjaKahla,c, Anne Kamitza,c, Vincent van Laake, Katharina Stohlmanna,c, Susanne Thomsena,c, Heiko Klawittera,c, Ingo Przesdzinga,c, Lars Neeba,b,c, Dorette Freyera,c, Josef Prillerf, Tony J. Collinsg,h, Dirk Megowa,c, Ulrich Dirnagla,b,c, David W. Andrewsg,h,1, and Andreas Meisela,b,c,d aDepartment of Experimental Neurology, bDepartment of Neurology, cCenter for Stroke Research Berlin, dNeuroCure Clinical Research Center, eDepartment of Internal Medicine, Pulmonary Medicine, and Infectious Diseases, and fNeuropsychiatry and Laboratory of Molecular Psychiatry, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; and gDepartment of Biochemistry and Biomedical Sciences, hMcMaster Biophotonics Facility, McMaster University, Hamilton, ON, Canada L8N 3Z5 Edited by Douglas R. Green, St. Jude Children’s Research Hospital, Memphis, TN, and accepted by the Editorial Board December 14, 2011 (received for review May 24, 2011) The metabolic state of a cell is a key determinant in the decision to live elusive. We therefore aimed to investigate the complex functional and proliferate or to die. Consequently, balanced energy metabolism roles of mitochondrial HKs in the adaptive response to different and the regulation of apoptosis are critical for the development and states of metabolic deprivation during hypoxia and hypoglycemia. maintenance of differentiated organisms. Hypoxia occurs physiolog- ically during development or exercise and pathologically in vascular Results disease, tumorigenesis, and inflammation, interfering with homeo- HIF-1–Dependent Activation of HKII in Primary Neurons and Hypoxia static metabolism. Here, we show that the hypoxia-inducible factor Tolerance. First, we assessed whether mitochondrial hexokinases (HIF)-1–regulated glycolytic enzyme hexokinase II (HKII) acts as a mo- responded to a hypoxia-mimicking stimulus that activates HIF-1 lecular switch that determines cellular fate by regulating both cytopro- transcriptional activity by interfering with degradation of HIF-1α tection and induction of apoptosis based on the metabolic state. We (19). We treated cultured primary rat brain cortical neurons with CELL BIOLOGY provide evidence for a direct molecular interactor of HKII and show the iron chelator deferoxamine (DFO), thereby mimicking hypoxia that, together with phosphoprotein enriched in astrocytes (PEA15), (19). DFO treatment resulted in marked protection from neuronal HKII inhibits apoptosis after hypoxia. In contrast, HKII accelerates ap- cell death after oxygen-glucose deprivation (OGD), an established model of cerebral ischemia (Fig. 1). At specified time points (Fig. optosis in the absence of PEA15 and under glucose deprivation. HKII 1A), we extracted RNA or protein from sister cultures to assess both protects cells from death during hypoxia and functions as a sensor changes in gene expression for both HKI (Fig. 1B)andHKII(Fig. of glucose availability during normoxia, inducing apoptosis in response 1C). HKI is the predominating isoenzyme in the brain and HKII is to glucose depletion. Thus, HKII-mediated apoptosis may represent an typically expressed in insulin-sensitive tissues or in malignant tumors evolutionarily conserved altruistic mechanism to eliminate cells during (20). However, messenger RNA (mRNA) expression of HKII was metabolic stress to the advantage of a multicellular organism. increased at 12 h after DFO treatment, whereas HKI expression did not change over 48 h. Immunoblotting of proteins from these cul- cell death | endogenous tolerance | fluorescence lifetime imaging tures revealed a concordant increase in HKII protein content (Fig. microscopy-FRET | mitochondria | preconditioning 1D). Using isoenzyme-specific electrophoretic zymography, we further demonstrated a specific increase in HKII activity (factor of alanced energy metabolism and regulation of apoptosis are of average increase = 2.28 ± 0.54), but not HKI activity in response to Bvital importance to all organisms (1, 2). Therefore, energy DFO treatment (Fig. 1 E and F). At 48 h after DFO treatment, metabolism and regulation of apoptosis are interdependent (3). In neurons were protected from cell death, as indicated by decreased cells, metabolism and apoptosis converge at mitochondria, thereby lactate dehydrogenase (LDH) release 24 h after OGD (Fig. 1G). integrating pathways responsible for endogenous tolerance against We therefore hypothesized that up-regulation of HKII significantly substrate deprivation (4). All differentiated multicellular organ- contributes to hypoxia tolerance mediated by activation of HIF-1. isms have evolved strategies to promote survival when deprived of metabolic substrates, such as during hypoxia (5). Therefore, elu- Overexpression of HKII Protects Primary Neurons from Hypoxic Cell cidating the underlying molecular mechanisms by which metabo- Death. To study the functional relevance of the increase in HKII lism and apoptosis are coregulated may lead to novel therapeutic expression and activity, we investigated the effect of overexpressed strategies for both acute and chronic diseases. HKII in comparison with BclXL under hypoxic conditions. We The transcription factor hypoxia-inducible factor (HIF)-1 is a key chose BclXL as a positive control because of its strong antiapoptotic regulator in the adaptation to hypoxia and the resultant energy properties (21) and its role in preconditioning-induced neuro- depletion, orchestrating the cellular response to hypoxic conditions protection (22). Transient transfection typically targets only a sub- (5–7). Induction of HIF-1 leads to the transcriptional regulation of set of cells (∼30% of neurons in a culture). Therefore, we visualized a multitude of genes, ultimately resulting in a hypoxia-tolerant state transfected cells using fluorescence after transfection of plasmids of the cell (7). HIF-1 also links hypoxia and glycolysis (8) via complex and incompletely understood mechanisms. HIF-1 adapts cellular metabolism to hypoxic conditions during development (7) Author contributions: P.M., D.W.A., and A.M. designed research; P.M., A. Kahl, A. Kamitz, or exercise (9) and thereby prevents death of tumor cells and pri- V.v.L., K.S., S.T., H.K., I.P., L.N., D.F., J.P., and D.M. performed research; P.M., T.J.C., D.M., mary cells under various conditions of disease (5–7, 10). In addition, U.D., D.W.A., and A.M. analyzed data; and P.M., D.W.A., and A.M. wrote the paper. HIF-1 controls innate immunity by regulating glycolysis in cells of The authors declare no conflict of interest. the immune system (11, 12). Finally, by controlling the expression This article is a PNAS Direct Submission. D.R.G. is a guest editor invited by the Editorial of members of the glycolytic cascade, including hexokinase II Board. (HKII) (7, 8), HIF-1 contributes to a proliferative metabolism (13). 1To whom correspondence may be addressed. E-mail: [email protected] or Mitochondrial glycolytic hexokinase isoenzymes (HKI, HKII, or [email protected]. also HKIV) may mediate cytoprotection under various conditions This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. (14–18). However, the molecular mechanisms have remained 1073/pnas.1108225109/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1108225109 PNAS Early Edition | 1of6 Downloaded by guest on September 25, 2021 catalytic activity is required for HKII to sense the metabolic state of A 48h Day 8 protein, Day 0 the cell, thereby protecting cells from hypoxic death but inducing preconditioning HK activity, 72h plate neurons 150 mM DFO / no DFO OGD LDH cell death upon glucose starvation. HKII Regulates Apoptotic Signaling After Metabolic Disturbance but 0 6h 12h 18h 24h 48h Not After Genotoxic Injury. Next, we investigated whether HKII mRNA extraction would protect cells from apoptosis in general or only when cells B C were stressed by metabolic disturbances (e.g., hypoxia). Trans- 12 12 control control fected HeLa cells were submitted to hypoxic conditions for 6 h 10 150μM DFO 10 150μM DFO A B 8 N=7 8 N=7 (Fig. 3 ) with 24 h of reoxygenation or for 21 h (Fig. 3 ). Al- 6 6 ternatively, apoptosis was induced by treatment with 100 nM 4 4 actinomycin D (Fig. 3C)or75μM etoposide (Fig. 3D). Cell death 2 2 was analyzed in transfected cells using flow cytometry. Although HKI - fold induction 0 HKII - fold induction 0 0h 6h 12h 18h 24h 48h 0h 6h 12h 18h 24h 48h HKII rescued cells from apoptosis after hypoxia irrespective of severity (Fig. 3 A and B), it did not rescue cells after actinomycin D E F G D or etoposide treatment (Fig. 3 C and D). BclXL, used as positive 20 * 20 * fi N=7 N=7 control, mediated signi cant protection from cell death in both - DFO + DFO 15 15 models. These data demonstrate that HKII regulates apoptosis < application - DFO + DFO after metabolic disturbances but not after genotoxic injury. 10 HKII 10 5 Identification of Phosphoprotein Enriched in Astrocytes as Direct Actin HKI 5 % HK II activity of total HK activity neuronal cell death Interactor of HKII. To investigate the molecular mechanism that 0 [%LDH (OGD - control)] HKII 0 governs HKII-dependent regulation of apoptosis, we screened a mouse brain cDNA library for putative interactors of HKII using no DFO no DFO a membrane-based split-ubiquitin yeast two-hybrid system (Fig. 150 μM DFO 150 μM DFO 4A). Using this approach, we identified phosphoprotein enriched – in astrocytes (PEA15) as a candidate for interaction with HKII. To Fig. 1. Hypoxia tolerance mediated by HIF-1 dependent activation of HKII fi in primary neurons. (A) Diagram of experimental paradigm. Analysis of HKI investigate the regulation of PEA15, we rst measured its ex- (B, E, and F) and HKII (C–F) expression in response to DFO treatment. (C) HKII pression after hypoxia-mimicking activation of HIF-1. In contrast expression was induced 12 h after hypoxia-mimicking treatment. After 48 h, to HKII (Fig. 1), PEA15 mRNA was not regulated under these (D) HKII protein and (E and F) enzyme activity were also increased.
Recommended publications
  • PARSANA-DISSERTATION-2020.Pdf
    DECIPHERING TRANSCRIPTIONAL PATTERNS OF GENE REGULATION: A COMPUTATIONAL APPROACH by Princy Parsana A dissertation submitted to The Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland July, 2020 © 2020 Princy Parsana All rights reserved Abstract With rapid advancements in sequencing technology, we now have the ability to sequence the entire human genome, and to quantify expression of tens of thousands of genes from hundreds of individuals. This provides an extraordinary opportunity to learn phenotype relevant genomic patterns that can improve our understanding of molecular and cellular processes underlying a trait. The high dimensional nature of genomic data presents a range of computational and statistical challenges. This dissertation presents a compilation of projects that were driven by the motivation to efficiently capture gene regulatory patterns in the human transcriptome, while addressing statistical and computational challenges that accompany this data. We attempt to address two major difficulties in this domain: a) artifacts and noise in transcriptomic data, andb) limited statistical power. First, we present our work on investigating the effect of artifactual variation in gene expression data and its impact on trans-eQTL discovery. Here we performed an in-depth analysis of diverse pre-recorded covariates and latent confounders to understand their contribution to heterogeneity in gene expression measurements. Next, we discovered 673 trans-eQTLs across 16 human tissues using v6 data from the Genotype Tissue Expression (GTEx) project. Finally, we characterized two trait-associated trans-eQTLs; one in Skeletal Muscle and another in Thyroid. Second, we present a principal component based residualization method to correct gene expression measurements prior to reconstruction of co-expression networks.
    [Show full text]
  • Aneuploidy: Using Genetic Instability to Preserve a Haploid Genome?
    Health Science Campus FINAL APPROVAL OF DISSERTATION Doctor of Philosophy in Biomedical Science (Cancer Biology) Aneuploidy: Using genetic instability to preserve a haploid genome? Submitted by: Ramona Ramdath In partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biomedical Science Examination Committee Signature/Date Major Advisor: David Allison, M.D., Ph.D. Academic James Trempe, Ph.D. Advisory Committee: David Giovanucci, Ph.D. Randall Ruch, Ph.D. Ronald Mellgren, Ph.D. Senior Associate Dean College of Graduate Studies Michael S. Bisesi, Ph.D. Date of Defense: April 10, 2009 Aneuploidy: Using genetic instability to preserve a haploid genome? Ramona Ramdath University of Toledo, Health Science Campus 2009 Dedication I dedicate this dissertation to my grandfather who died of lung cancer two years ago, but who always instilled in us the value and importance of education. And to my mom and sister, both of whom have been pillars of support and stimulating conversations. To my sister, Rehanna, especially- I hope this inspires you to achieve all that you want to in life, academically and otherwise. ii Acknowledgements As we go through these academic journeys, there are so many along the way that make an impact not only on our work, but on our lives as well, and I would like to say a heartfelt thank you to all of those people: My Committee members- Dr. James Trempe, Dr. David Giovanucchi, Dr. Ronald Mellgren and Dr. Randall Ruch for their guidance, suggestions, support and confidence in me. My major advisor- Dr. David Allison, for his constructive criticism and positive reinforcement.
    [Show full text]
  • Substantial Conformational Change Mediated by Charge-Triad Residues of the Death Effector Domain in Protein-Protein Interactions
    Substantial Conformational Change Mediated by Charge-Triad Residues of the Death Effector Domain in Protein-Protein Interactions Edward C. Twomey1,2,3, Dana F. Cordasco1,2, Stephen D. Kozuch2, Yufeng Wei1* 1 Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey, United States of America, 2 Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey, United States of America, 3 Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, New York, United States of America Abstract Protein conformational changes are commonly associated with the formation of protein complexes. The non-catalytic death effector domains (DEDs) mediate protein-protein interactions in a variety of cellular processes, including apoptosis, proliferation and migration, and glucose metabolism. Here, using NMR residual dipolar coupling (RDC) data, we report a conformational change in the DED of the phosphoprotein enriched in astrocytes, 15 kDa (PEA-15) protein in the complex with a mitogen-activated protein (MAP) kinase, extracellular regulated kinase 2 (ERK2), which is essential in regulating ERK2 cellular distribution and function in cell proliferation and migration. The most significant conformational change in PEA-15 happens at helices a2, a3, and a4, which also possess the highest flexibility among the six-helix bundle of the DED. This crucial conformational change is modulated by the D/E-RxDL charge-triad motif, one of the prominent structural features of DEDs, together with a number of other electrostatic and hydrogen bonding interactions on the protein surface. Charge- triad motif promotes the optimal orientation of key residues and expands the binding interface to accommodate protein- protein interactions.
    [Show full text]
  • Role and Regulation of the P53-Homolog P73 in the Transformation of Normal Human Fibroblasts
    Role and regulation of the p53-homolog p73 in the transformation of normal human fibroblasts Dissertation zur Erlangung des naturwissenschaftlichen Doktorgrades der Bayerischen Julius-Maximilians-Universität Würzburg vorgelegt von Lars Hofmann aus Aschaffenburg Würzburg 2007 Eingereicht am Mitglieder der Promotionskommission: Vorsitzender: Prof. Dr. Dr. Martin J. Müller Gutachter: Prof. Dr. Michael P. Schön Gutachter : Prof. Dr. Georg Krohne Tag des Promotionskolloquiums: Doktorurkunde ausgehändigt am Erklärung Hiermit erkläre ich, dass ich die vorliegende Arbeit selbständig angefertigt und keine anderen als die angegebenen Hilfsmittel und Quellen verwendet habe. Diese Arbeit wurde weder in gleicher noch in ähnlicher Form in einem anderen Prüfungsverfahren vorgelegt. Ich habe früher, außer den mit dem Zulassungsgesuch urkundlichen Graden, keine weiteren akademischen Grade erworben und zu erwerben gesucht. Würzburg, Lars Hofmann Content SUMMARY ................................................................................................................ IV ZUSAMMENFASSUNG ............................................................................................. V 1. INTRODUCTION ................................................................................................. 1 1.1. Molecular basics of cancer .......................................................................................... 1 1.2. Early research on tumorigenesis ................................................................................. 3 1.3. Developing
    [Show full text]
  • Brief Genetics Report
    Brief Genetics Report Calsquestrin 1 (CASQ1) Gene Polymorphisms Under Chromosome 1q21 Linkage Peak Are Associated With Type 2 Diabetes in Northern European Caucasians Swapan Kumar Das,1 Winston Chu,1 Zhengxian Zhang,1 Sandra J. Hasstedt,2 and Steven C. Elbein1,3 Genome-wide scans in multiple populations have iden- tified chromosome 1q21-q24 as one susceptibility region for type 2 diabetes. To map the susceptibility genes, we onsiderable data support a genetic etiology for first placed a dense single nucleotide polymorphism type 2 diabetes, but multiple susceptibility loci (SNP) map across the linked region. We identified two on different chromosomes are likely involved SNPs that showed strong associations, and both mapped (1). We previously mapped one type 2 diabetes to within intron 2 of the calsequestrin 1 (CASQ1) gene. C susceptibility locus in extended North European Cauca- We tested the hypothesis that sequence variation in or near CASQ1 contributed to type 2 diabetes susceptibil- sian families to chromosome 1q21-q24 (2), a region that is ity in Northern European Caucasians by identifying now well replicated in Old Order Amish, Pima Indian, additional SNPs from the public database and by screen- Chinese, French, and British families (1). Recently, we ing the CASQ1 gene for additional variation. In addition completed a dense linkage map and identified two linkage to 15 known SNPs in this region, we found 8 new SNPs, peaks centered on locations 157 and 164 Mb (3). Addition- 3 of which were in exons. A single rare nonsynonymous ally, we have previously reported the association of poly- SNP in exon 11 (A348V) was not associated with type 2 morphisms of the liver pyruvate kinase enzyme with type diabetes.
    [Show full text]
  • Molecular Characterization of the Human PEA15 Gene on 1Q21–Q22 and Association with Type 2 Diabetes Mellitus in Pima Indians K
    Gene 241 (2000) 143–148 www.elsevier.com/locate/gene Molecular characterization of the human PEA15 gene on 1q21–q22 and association with type 2 diabetes mellitus in Pima Indians k Johanna K. Wolford *, Clifton Bogardus, Victoria Ossowski, Michal Prochazka Clinical Diabetes and Nutrition Section, Phoenix Epidemiology and Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National institutes of Health, 4212 N. 16th Street, Phoenix, AZ 85016, USA Received 24 June 1999; received in revised form 1 October 1999; accepted 8 October 1999 Received by K. Gardiner Abstract The PEA15 gene encoding a protein kinase C substrate is widely expressed, and its overexpression may contribute to impairment of glucose uptake. PEA15 is located within a region on human 1q linked with type 2 diabetes in both Pima Indians and Caucasians. To assess the potential contribution of genetic alterations within this locus to disease susceptibility in the Pimas, we have investigated its genomic sequences. The PEA15 locus is composed of four exons spanning approximately 10.2 kb of genomic DNA, flanked upstream by an potentially expressed Alu element, downstream by the H326 gene, and is located within 250 kb of KCNJ9. We also sequenced over 2 kb of the promoter region and identified various motifs analogous to known transcription factor binding sites. By analysis of 22 Pimas, including 13 diabetic subjects, we detected four single nucleotide polymorphisms (SNPs) in the non-coding regions of PEA15, including three frequent variants that were in allelic disequilibrium, and one variant found only in a single Pima. The three SNPs were not associated with type 2 diabetes mellitus in 50 affected and 50 control Pimas ( p=0.12–0.17), and we conclude that mutations in this gene probably do not contribute significantly to disease susceptibility in this Native American tribe.
    [Show full text]
  • The Human Gene Connectome As a Map of Short Cuts for Morbid Allele Discovery
    The human gene connectome as a map of short cuts for morbid allele discovery Yuval Itana,1, Shen-Ying Zhanga,b, Guillaume Vogta,b, Avinash Abhyankara, Melina Hermana, Patrick Nitschkec, Dror Friedd, Lluis Quintana-Murcie, Laurent Abela,b, and Jean-Laurent Casanovaa,b,f aSt. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065; bLaboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris Descartes University, Institut National de la Santé et de la Recherche Médicale U980, Necker Medical School, 75015 Paris, France; cPlateforme Bioinformatique, Université Paris Descartes, 75116 Paris, France; dDepartment of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; eUnit of Human Evolutionary Genetics, Centre National de la Recherche Scientifique, Unité de Recherche Associée 3012, Institut Pasteur, F-75015 Paris, France; and fPediatric Immunology-Hematology Unit, Necker Hospital for Sick Children, 75015 Paris, France Edited* by Bruce Beutler, University of Texas Southwestern Medical Center, Dallas, TX, and approved February 15, 2013 (received for review October 19, 2012) High-throughput genomic data reveal thousands of gene variants to detect a single mutated gene, with the other polymorphic genes per patient, and it is often difficult to determine which of these being of less interest. This goes some way to explaining why, variants underlies disease in a given individual. However, at the despite the abundance of NGS data, the discovery of disease- population level, there may be some degree of phenotypic homo- causing alleles from such data remains somewhat limited. geneity, with alterations of specific physiological pathways under- We developed the human gene connectome (HGC) to over- come this problem.
    [Show full text]
  • Identification of Gastric Cancer–Related Genes Using a Cdna Microarray Containing Novel Expressed Sequence Tags Expressed in Gastric Cancer Cells
    Vol. 11, 473–482, January 15, 2005 Clinical Cancer Research 473 Identification of Gastric Cancer–Related Genes Using a cDNA Microarray Containing Novel Expressed Sequence Tags Expressed in Gastric Cancer Cells Jeong-Min Kim,1,5 Ho-Yong Sohn,4 overexpressed in z68% of tissues and the MT2A gene Sun Young Yoon,1 Jung-Hwa Oh,1 Jin Ok Yang,1 was down-expressed in 72% of the tissues. Western blotting and immunohistochemical analyses for CDC20 and SKB1 Joo Heon Kim,2 Kyu Sang Song,3 Seung-Moo Rho,2 1 1 5 showed overexpression and localization changes of the Hyan Sook Yoo, Yong Sung Kim, Jong-Guk Kim, corresponding protein in human gastric cancer tissues. 1 and Nam-Soon Kim Conclusions: Novel genes that are related to human 1Genome Research Center, Korea Research Institute of Bioscience and gastric cancer were identified using cDNA microarray Biotechnology; 2Department of Pathology, Eulji University School of 3 developed in our laboratory. In particular, CDC20 and Medicine; and Department of Pathology, College of Medicine, MT2A represent a potential biomarker of human gastric Chungnam National University, Daejeon, Korea; 4Department of Food and Nutrition, Andong National University, Andong, Korea; and cancer. These newly identified genes should provide a 5Department of Microbiology, College of Natural Sciences, Kyungpook valuable resource for understanding the molecular mecha- National University, Daegu, Korea nism associated with tumorigenesis of gastric carcinogenesis and for the discovery of potential diagnostic markers of gastric cancer. ABSTRACT Purpose: Gastric cancer is one of the most frequently INTRODUCTION diagnosed malignancies in the world, especially in Korea and Japan.
    [Show full text]
  • PEA-15 Inhibits Tumorigenesis in an MDA-MB-468 Triple-Negative
    Published OnlineFirst March 9, 2010; DOI: 10.1158/1078-0432.CCR-09-1456 Published Online First on March 9, 2010 as 10.1158/1078-0432.CCR-09-1456 Cancer Therapy: Preclinical Clinical Cancer Research PEA-15 Inhibits Tumorigenesis in an MDA-MB-468 Triple-Negative Breast Cancer Xenograft Model through Increased Cytoplasmic Localization of Activated Extracellular Signal-Regulated Kinase Chandra Bartholomeusz1,2, Ana M. Gonzalez-Angulo2,3, Anna Kazansky1,2, Savitri Krishnamurthy4, Ping Liu5, Linda X. H. Yuan1,2, Fumiyuki Yamasaki1,2, Shuying Liu3, Naoki Hayashi1,2, Dongwei Zhang1,2, Francisco J. Esteva1,2, Gabriel N. Hortobagyi1,2, and Naoto T. Ueno1,2,6 Abstract Purpose: To determine the role of PEA-15 in breast cancer. Experimental Design: A reverse-phase protein array was used to measure PEA-15 expression levels in 320 human breast cancers; these levels were correlated with clinical and tumor characteristics. PEA-15 was overexpressed by an adenovirus vector or by stably expressing PEA-15 in different breast cancer cell lines. The effects on breast cancer cell survival and on the downstream apoptotic signaling pathway were mea- sured in terms of cell proliferation (trypan blue for cell viability, bromodeoxyuridine incorporation for DNA synthesis), anchorage-independent growth (soft agar colony formation), and apoptosis (fluores- cence-activated cell sorter analysis). The preclinical efficacy of Ad.PEA-15 given intratumorally was eval- uated in nude mice bearing tumors from s.c. implanted human MDA-MB-468 triple-negative breast cancer cells. Results: In human breast cancers, low levels of PEA-15 expression correlated with high nuclear grade (P < 0.0001) and with negative hormone receptor status (P = 0.0004).
    [Show full text]
  • Multiscale Genomic Analysis of The
    University of Tennessee Health Science Center UTHSC Digital Commons Theses and Dissertations (ETD) College of Graduate Health Sciences 5-2009 Multiscale Genomic Analysis of the Corticolimbic System: Uncovering the Molecular and Anatomic Substrates of Anxiety-Related Behavior Khyobeni Mozhui University of Tennessee Health Science Center Follow this and additional works at: https://dc.uthsc.edu/dissertations Part of the Mental and Social Health Commons, Nervous System Commons, and the Neurosciences Commons Recommended Citation Mozhui, Khyobeni , "Multiscale Genomic Analysis of the Corticolimbic System: Uncovering the Molecular and Anatomic Substrates of Anxiety-Related Behavior" (2009). Theses and Dissertations (ETD). Paper 180. http://dx.doi.org/10.21007/etd.cghs.2009.0219. This Dissertation is brought to you for free and open access by the College of Graduate Health Sciences at UTHSC Digital Commons. It has been accepted for inclusion in Theses and Dissertations (ETD) by an authorized administrator of UTHSC Digital Commons. For more information, please contact [email protected]. Multiscale Genomic Analysis of the Corticolimbic System: Uncovering the Molecular and Anatomic Substrates of Anxiety-Related Behavior Document Type Dissertation Degree Name Doctor of Philosophy (PhD) Program Anatomy and Neurobiology Research Advisor Robert W. Williams, Ph.D. Committee John D. Boughter, Ph.D. Eldon E. Geisert, Ph.D. Kristin M. Hamre, Ph.D. Jeffery D. Steketee, Ph.D. DOI 10.21007/etd.cghs.2009.0219 This dissertation is available at UTHSC Digital
    [Show full text]
  • Transcriptome Analysis of Peripheral Whole Blood Identifies Crucial
    Zheng et al. BMC Medical Genomics (2020) 13:136 https://doi.org/10.1186/s12920-020-00785-y RESEARCH ARTICLE Open Access Transcriptome analysis of peripheral whole blood identifies crucial lncRNAs implicated in childhood asthma Peiyan Zheng1†, Chen Huang2†, Dongliang Leng2, Baoqing Sun1* and Xiaohua Douglas Zhang2,3* Abstract Background: Asthma is a chronic disorder of both adults and children affecting more than 300 million people heath worldwide. Diagnose and treatment for asthma, particularly in childhood asthma have always remained a great challenge because of its complex pathogenesis and multiple triggers, such as allergen, viral infection, tobacco smoke, dust, etc. It is thereby great significant to deeply investigate the transcriptome changes in asthmatic children before and after desensitization treatment, in order that we could identify potential and key mRNAs and lncRNAs which might be considered as useful RNA molecules for observing and supervising desensitization therapy for asthma, which might guide the diagnose and therapy in childhood asthma. Methods: In the present study, we performed a systematic transcriptome analysis based on the deep RNA sequencing of ten asthmatic children before and after desensitization treatment, including identification of lncRNAs using a stringent filtering pipeline, differential expression analysis and network analysis, etc. Results: First, a large number of lncRNAs were identified and characterized. Then differential expression analysis revealed 39 mRNAs and 15 lncRNAs significantly differentially expressed which involved in two biological processes and pathways. A co-expressed network analysis figured out a desensitization-treatment-related module which contains 27 mRNAs and 21 lncRNAs using WGCNA R package. Module analysis disclosed 17 genes associated to asthma at distinct level.
    [Show full text]
  • Anti-PEA15 Antibody (ARG66865)
    Product datasheet [email protected] ARG66865 Package: 100 μg anti-PEA15 antibody Store at: -20°C Summary Product Description Rabbit Polyclonal antibody recognizes PEA15 Tested Reactivity Hu Predict Reactivity Ms, Rat Tested Application IHC-P, WB Host Rabbit Clonality Polyclonal Isotype IgG Target Name PEA15 Antigen Species Human Immunogen Synthetic peptide corresponding to aa. 70-119 of Human PEA15. Conjugation Un-conjugated Alternate Names MAT1H; MAT1; Astrocytic phosphoprotein PEA-15; HUMMAT1H; 15 kDa phosphoprotein enriched in astrocytes; PED; PEA-15; Phosphoprotein enriched in diabetes; HMAT1 Application Instructions Application table Application Dilution IHC-P 1:100 - 1:300 WB 1:500 - 1:2000 Application Note * The dilutions indicate recommended starting dilutions and the optimal dilutions or concentrations should be determined by the scientist. Positive Control Jurkat Calculated Mw 15 kDa Observed Size ~ 15 kDa Properties Form Liquid Purification Affinity purification with immunogen. Buffer PBS, 0.02% Sodium azide, 50% Glycerol and 0.5% BSA. Preservative 0.02% Sodium azide Stabilizer 50% Glycerol and 0.5% BSA Concentration 1 mg/ml www.arigobio.com 1/2 Storage instruction For continuous use, store undiluted antibody at 2-8°C for up to a week. For long-term storage, aliquot and store at -20°C. Storage in frost free freezers is not recommended. Avoid repeated freeze/thaw cycles. Suggest spin the vial prior to opening. The antibody solution should be gently mixed before use. Note For laboratory research only, not for drug, diagnostic or other use. Bioinformation Gene Symbol PEA15 Gene Full Name phosphoprotein enriched in astrocytes 15 Background This gene encodes a death effector domain-containing protein that functions as a negative regulator of apoptosis.
    [Show full text]