Signaling This Information Is Current As of September 26, 2021

Total Page:16

File Type:pdf, Size:1020Kb

Signaling This Information Is Current As of September 26, 2021 IL-2 Requirement for Human Plasma Cell Generation: Coupling Differentiation and Proliferation by Enhancing MAPK−ERK Signaling This information is current as of September 26, 2021. Simon Le Gallou, Gersende Caron, Céline Delaloy, Delphine Rossille, Karin Tarte and Thierry Fest J Immunol 2012; 189:161-173; Prepublished online 25 May 2012; doi: 10.4049/jimmunol.1200301 Downloaded from http://www.jimmunol.org/content/189/1/161 Supplementary http://www.jimmunol.org/content/suppl/2012/05/25/jimmunol.120030 http://www.jimmunol.org/ Material 1.DC1 References This article cites 55 articles, 29 of which you can access for free at: http://www.jimmunol.org/content/189/1/161.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 26, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology IL-2 Requirement for Human Plasma Cell Generation: Coupling Differentiation and Proliferation by Enhancing MAPK–ERK Signaling Simon Le Gallou,*,†,‡,1,2 Gersende Caron,*,†,‡,x,1 Ce´line Delaloy,*,†,‡,x,1 Delphine Rossille,x,{ Karin Tarte,*,†,‡,x and Thierry Fest*,†,‡,x Mature B cell differentiation involves a well-established transcription factor cascade. However, the temporal dynamics of cell sig- naling pathways regulating transcription factor network and coordinating cell proliferation and differentiation remain poorly de- fined. To gain insight into the molecular processes and extrinsic cues required for B cell differentiation, we set up a controlled primary culture system to differentiate human naive B cells into plasma cells (PCs). We identified T cell-produced IL-2 to be crit- ically involved in ERK1/2-triggered PC differentiation. IL-2 drove activated B cell differentiation toward PC independently of its Downloaded from proliferation and survival functions. Indeed, IL-2 potentiated ERK activation and subsequent BACH2 and IRF8 downregulation, sustaining BLIMP1 expression, the master regulator for PC differentiation. Inhibition of the MAPK–ERK pathway, unlike STAT5 signaling, impaired IL-2–induced PC differentiation and rescued the expression profile of BACH2 and IRF8. These results identify IL-2 as a crucial early input in mature B cell fate commitment. The Journal of Immunology, 2012, 189: 161–173. cell-dependent immune response is initiated in germinal the follicle (3–7). In GCs, B cells undergo terminal differentiation http://www.jimmunol.org/ centers (GCs) after seeding by a small number of rapidly and selection, which depend tightly on the light zone microenvi- T dividing Ag-responding B cells. These cells undergo a se- ronment (3, 4). The gene regulatory network that governs transi- ries of proliferation/selection steps to give rise to memory B cells tion between GC B cells and plasmablasts is well understood and or long-lived plasma cells (PCs) (1). The GC response initiates in the heavily controlled by cytokines, particularly those that reduce outer follicle where naive B cells (NBCs) encounter their specific BCL6 expression and induce BLIMP1, two mutually exclusive Ags (2). Subsequently, activated B cells relocate to the B zone–T transcriptional regulators. BLIMP1 orchestrates PC differentiation zone boundary where Ag-specific B and T cells interact with each by extinguishing the mature B cell gene expression program in- other and form long-lived pairs. In this pairing, B and T cells up- cluding BCL6, freeing factors like IRF4, and XBP1 (8, 9). Notably, regulate BCL6, proliferate (3, 4), and acquire a centroblastic or a in response to stimulation, B cells stochastically pursue various by guest on September 26, 2021 follicular Th (TFH) cell identity respectively before migrating inside distinct fates. Some cells undergo differentiation and isotype switching depending on cell division and cytokine environment (10–12). *INSERM, Unite´ Mixte de Recherche 917, Rennes F-35043, France; †Universite´ de Rennes 1, Unite´ Mixte de Recherche 917, Rennes F-35043, France; ‡Etablissement The T cell help provides distinct signals that are critical in de- Franc¸ais du Sang de Bretagne, Unite´ Mixte de Recherche 917, Rennes F-35043, termining B cell behavior. During initial T–B cognate interaction, France; xCentre Hospitalier Universitaire de Rennes, Poˆle Cellules & Tissus, Rennes { T cell potentially induces a large variety of inputs including F-35033, France; and INSERM, Unite´ Mixte de Recherche 936, Rennes F-35043, France CD40 and CD80/CD86 engagement (13, 14). However, T cell- 1S.L.G., G.C., and C.D. contributed equally to this work. derived signals seem not to be mandatory, as T cell-independent B cell clonal expansion, GC formation, and acquisition of GC 2Current address: INSERM U783, Universite´ Descartes, Paris, France. B cell phenotype may be observed in the absence of T cells (15). Received for publication January 24, 2012. Accepted for publication April 25, 2012. However, stable contacts of TFH cells with B cells are absolutely This work was supported by an internal grant from the Hematology and Immunology Laboratory, Poˆle Cellules & Tissus, Centre Hospitalier Universitaire de Rennes. S.L.G. required to sustain GC maturation, centrocyte selection, and PC was supported by a research grant from La Ligue Contre le Cancer/Re´gion Bretagne, generation (5, 16). TFH cells are not statically providing a single and C.D. was supported by an internal grant from the Poˆle Cellules & Tissus, Centre stimulus to B cells, but instead at some point they promote high- Hospitalier Universitaire de Rennes. affinity GC B cells to become either PCs or memory B cells. This S.L.G. performed research, analyzed data, and established the B cell differentiation model; G.C. designed experiments and performed research; C.D. performed research could be an early or late input that might function with a con- and analyzed cell signaling; D.R. performed microarray analyses; K.T. contributed to comitant CD40 signal. First, T cells drive NBC proliferation and study design; and T.F. designed and supervised research, raised funds, and wrote the inhibit PC differentiation (17). At a later stage, T cells promote paper. FH a reinforcement of BCL6 repression signals in high-affinity B cells Microarray data presented in this article have been submitted to the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/) under accession no. GSE36975. (18, 19). Besides CD40L, TFH cells produce IL-21, which acts directly on GC B cells maximizing BCL6 expression leading to Address correspondence and reprint requests to Prof. Thierry Fest, INSERM U917, Faculte´ de Me´decine, 2 Avenue du Professeur Le´on Bernard, CS 34317, 35043 cell survival and proliferation (20–23). In contrast, IL-21 induces Rennes Cedex, France. E-mail address: [email protected] PC differentiation through a STAT3-dependent BLIMP1 induction The online version of this article contains supplemental material. (21, 24, 25). Numerous other T cell-derived cytokines enhance PC + Abbreviations used in this article: FC, fold change; GC, germinal center; GEP, gene differentiation such as IL-2 produced by memory CD4 T cells expression profiling; GL, gene list; HCA, hierarchical clustering analysis; NBC, (26). Recently in mice, ERKs have been shown to be necessary to naive B cell; PC, plasma cell; qRT-PCR, quantitative RT-PCR; T , follicular Th. FH trigger PC generation, mediating the cytokine-induced production Copyright Ó 2012 by The American Association of Immunologists, Inc. 0022-1767/12/$16.00 of BLIMP1 (27). Altogether, these findings reinforce the well-known www.jimmunol.org/cgi/doi/10.4049/jimmunol.1200301 162 IL-2–ENHANCED ERK1/2 SIGNALING AND PC DIFFERENTIATION assumption that a combination of a BCR signal and T cell help is Apoptosis and proliferation assays required to initiate PC differentiation (28). Apoptosis and proliferation were analyzed using a PE-conjugated anti-active To explore events that govern human NBC differentiation, we caspase-3 apoptosis kit (BD Biosciences) and an FITC-conjugated anti-BrdU designed an in vitro two-step culture model combining BCR signal, kit (BD Biosciences), respectively, according to the manufacturer’s instruc- TLR activation, and T cell help in the form of CD40L and tions. For BrdU staining, B cells were incubated with BrdU (10 mM) during cytokines. Unswitched human naive precursors differentiated into 45 min followed by CD38 staining. Cells were then fixed, permeabilized, and DNAse treated before subsequent staining and FACS analysis. CD20+CD38+ and CD20loCD38hi cells characterized by distinct cell fates. We explored one by one factors used in our model and Ig secretion and Western blotting found that T cell-produced IL-2 was critical for human NBC For Ig secretion assay, differentiated and nondifferentiated B cells were commitment to PCs. IL-2 activated the ERK pathway at a threshold isolated and reseeded with IL-2, IL-4, and IL-10 at 1 3 106 cells/ml for 18 h. level that triggered, beyond the induction of cell cycle progression, IgG, IgA, and IgM secretion was assessed by ELISA using a goat anti- PC generation. BACH2 and IRF8 expression was downregulated human Ig for coating and secondary alkaline phosphatase-coupled Abs specific for g, a,orm chain, respectively (all from Jackson Immuno- through the MEK–ERK signaling pathway in IL-2–primed B cells. Research Laboratories). SDS-PAGE and Western blotting were performed Therefore, IL-2 reinforced the mutual repression between BCL6 according to standard procedures and Abs listed in Table I.
Recommended publications
  • Epigenetic Reprogramming Sensitizes CML Stem Cells to Combined EZH2 and Tyrosine
    Published OnlineFirst September 14, 2016; DOI: 10.1158/2159-8290.CD-16-0263 RESEARCH BRIEF Epigenetic Reprogramming Sensitizes CML Stem Cells to Combined EZH2 and Tyrosine Kinase Inhibition Mary T. Scott 1 , 2 , Koorosh Korfi 1 , 2 , Peter Saffrey 1 , Lisa E.M. Hopcroft 2 , Ross Kinstrie 1 , Francesca Pellicano 2 , Carla Guenther 1 , 2 , Paolo Gallipoli 2 , Michelle Cruz 1 , Karen Dunn 2 , Heather G. Jorgensen 2 , Jennifer E. Cassels 2 , Ashley Hamilton 2 , Andrew Crossan 1 , Amy Sinclair 2 , Tessa L. Holyoake 2 , and David Vetrie 1 ABSTRACT A major obstacle to curing chronic myeloid leukemia (CML) is residual disease main- tained by tyrosine kinase inhibitor (TKI)–persistent leukemic stem cells (LSC). These are BCR–ABL1 kinase independent, refractory to apoptosis, and serve as a reservoir to drive relapse or TKI resistance. We demonstrate that Polycomb Repressive Complex 2 is misregulated in chronic phase CML LSCs. This is associated with extensive reprogramming of H3K27me3 targets in LSCs, thus sensi- tizing them to apoptosis upon treatment with an EZH2-specifi c inhibitor (EZH2i). EZH2i does not impair normal hematopoietic stem cell survival. Strikingly, treatment of primary CML cells with either EZH2i or TKI alone caused signifi cant upregulation of H3K27me3 targets, and combined treatment further potentiated these effects and resulted in signifi cant loss of LSCs compared to TKI alone, in vitro , and in long-term bone marrow murine xenografts. Our fi ndings point to a promising epigenetic-based thera- peutic strategy to more effectively target LSCs in patients with CML receiving TKIs. SIGNIFICANCE: In CML, TKI-persistent LSCs remain an obstacle to cure, and approaches to eradicate them remain a signifi cant unmet clinical need.
    [Show full text]
  • Gene Essentiality Landscape and Druggable Oncogenic Dependencies in Herpesviral Primary Effusion Lymphoma
    ARTICLE DOI: 10.1038/s41467-018-05506-9 OPEN Gene essentiality landscape and druggable oncogenic dependencies in herpesviral primary effusion lymphoma Mark Manzano1, Ajinkya Patil1, Alexander Waldrop2, Sandeep S. Dave2, Amir Behdad3 & Eva Gottwein1 Primary effusion lymphoma (PEL) is caused by Kaposi’s sarcoma-associated herpesvirus. Our understanding of PEL is poor and therefore treatment strategies are lacking. To address this 1234567890():,; need, we conducted genome-wide CRISPR/Cas9 knockout screens in eight PEL cell lines. Integration with data from unrelated cancers identifies 210 genes as PEL-specific oncogenic dependencies. Genetic requirements of PEL cell lines are largely independent of Epstein-Barr virus co-infection. Genes of the NF-κB pathway are individually non-essential. Instead, we demonstrate requirements for IRF4 and MDM2. PEL cell lines depend on cellular cyclin D2 and c-FLIP despite expression of viral homologs. Moreover, PEL cell lines are addicted to high levels of MCL1 expression, which are also evident in PEL tumors. Strong dependencies on cyclin D2 and MCL1 render PEL cell lines highly sensitive to palbociclib and S63845. In summary, this work comprehensively identifies genetic dependencies in PEL cell lines and identifies novel strategies for therapeutic intervention. 1 Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA. 2 Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA. 3 Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA. Correspondence and requests for materials should be addressed to E.G. (email: [email protected]) NATURE COMMUNICATIONS | (2018) 9:3263 | DOI: 10.1038/s41467-018-05506-9 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-05506-9 he human oncogenic γ-herpesvirus Kaposi’s sarcoma- (IRF4), a critical oncogene in multiple myeloma33.
    [Show full text]
  • Identifying and Mapping Cell-Type-Specific Chromatin PNAS PLUS Programming of Gene Expression
    Identifying and mapping cell-type-specific chromatin PNAS PLUS programming of gene expression Troels T. Marstranda and John D. Storeya,b,1 aLewis-Sigler Institute for Integrative Genomics, and bDepartment of Molecular Biology, Princeton University, Princeton, NJ 08544 Edited by Wing Hung Wong, Stanford University, Stanford, CA, and approved January 2, 2014 (received for review July 2, 2013) A problem of substantial interest is to systematically map variation Relating DHS to gene-expression levels across multiple cell in chromatin structure to gene-expression regulation across con- types is challenging because the DHS represents a continuous ditions, environments, or differentiated cell types. We developed variable along the genome not bound to any specific region, and and applied a quantitative framework for determining the exis- the relationship between DHS and gene expression is largely tence, strength, and type of relationship between high-resolution uncharacterized. To exploit variation across cell types and test chromatin structure in terms of DNaseI hypersensitivity and genome- for cell-type-specific relationships between DHS and gene expres- wide gene-expression levels in 20 diverse human cell types. We sion, the measurement units must be placed on a common scale, show that ∼25% of genes show cell-type-specific expression ex- the continuous DHS measure associated to each gene in a well- plained by alterations in chromatin structure. We find that distal defined manner, and all measurements considered simultaneously. regions of chromatin structure (e.g., ±200 kb) capture more genes Moreover, the chromatin and gene-expression relationship may with this relationship than local regions (e.g., ±2.5 kb), yet the local only manifest in a single cell type, making standard measures of regions show a more pronounced effect.
    [Show full text]
  • Viewed Under 23 (B) Or 203 (C) fi M M Male Cko Mice, and Largely Unaffected Magni Cation; Scale Bars, 500 M (B) and 50 M (C)
    BRIEF COMMUNICATION www.jasn.org Renal Fanconi Syndrome and Hypophosphatemic Rickets in the Absence of Xenotropic and Polytropic Retroviral Receptor in the Nephron Camille Ansermet,* Matthias B. Moor,* Gabriel Centeno,* Muriel Auberson,* † † ‡ Dorothy Zhang Hu, Roland Baron, Svetlana Nikolaeva,* Barbara Haenzi,* | Natalya Katanaeva,* Ivan Gautschi,* Vladimir Katanaev,*§ Samuel Rotman, Robert Koesters,¶ †† Laurent Schild,* Sylvain Pradervand,** Olivier Bonny,* and Dmitri Firsov* BRIEF COMMUNICATION *Department of Pharmacology and Toxicology and **Genomic Technologies Facility, University of Lausanne, Lausanne, Switzerland; †Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts; ‡Institute of Evolutionary Physiology and Biochemistry, St. Petersburg, Russia; §School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; |Services of Pathology and ††Nephrology, Department of Medicine, University Hospital of Lausanne, Lausanne, Switzerland; and ¶Université Pierre et Marie Curie, Paris, France ABSTRACT Tight control of extracellular and intracellular inorganic phosphate (Pi) levels is crit- leaves.4 Most recently, Legati et al. have ical to most biochemical and physiologic processes. Urinary Pi is freely filtered at the shown an association between genetic kidney glomerulus and is reabsorbed in the renal tubule by the action of the apical polymorphisms in Xpr1 and primary fa- sodium-dependent phosphate transporters, NaPi-IIa/NaPi-IIc/Pit2. However, the milial brain calcification disorder.5 How- molecular identity of the protein(s) participating in the basolateral Pi efflux remains ever, the role of XPR1 in the maintenance unknown. Evidence has suggested that xenotropic and polytropic retroviral recep- of Pi homeostasis remains unknown. Here, tor 1 (XPR1) might be involved in this process. Here, we show that conditional in- we addressed this issue in mice deficient for activation of Xpr1 in the renal tubule in mice resulted in impaired renal Pi Xpr1 in the nephron.
    [Show full text]
  • PLEKHM2 Mutation Leads to Abnormal Localization of Lysosomes, Impaired
    Human Molecular Genetics, 2015, Vol. 24, No. 25 7227–7240 doi: 10.1093/hmg/ddv423 Advance Access Publication Date: 12 October 2015 Original Article ORIGINAL ARTICLE PLEKHM2 mutation leads to abnormal localization Downloaded from of lysosomes, impaired autophagy flux and associates with recessive dilated cardiomyopathy and left ventricular noncompaction http://hmg.oxfordjournals.org/ Emad Muhammad1,†, Aviva Levitas2,†, Sonia R. Singh3,4, Alex Braiman1, Rivka Ofir5, Sharon Etzion5, Val C. Sheffield6, Yoram Etzion5,7, Lucie Carrier3,4 and Ruti Parvari1,8,* 1Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel, 2Department of Pediatric Cardiology, Soroka University Medical Center and at Bibliothekssystem Universitaet Hamburg on December 2, 2015 Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel, 3Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany, 4DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany, 5Regenerative Medicine and Stem Cell Research Center, Beer-Sheva 84105, Israel, 6Department of Pediatrics, Division of Medical Genetics and Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA, 7Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel and 8National Institute for Biotechnology in the Negev, Ben- Gurion University of the Negev, Beer-Sheva, Israel *To whom correspondence should be addressed. Tel: +972 86479967; Fax: +972 86472983; Email: [email protected] Abstract Gene mutations, mostly segregating with a dominant mode of inheritance, are important causes of dilated cardiomyopathy (DCM), a disease characterized by enlarged ventricular dimensions, impaired cardiac function, heart failure and high risk of death.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Vascular Homeostasis and Inflammation in Health and Disease
    International Journal of Molecular Sciences Review Vascular Homeostasis and Inflammation in Health and Disease—Lessons from Single Cell Technologies Olga Bondareva * and Bilal N. Sheikh * Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany * Correspondence: [email protected] (O.B.); [email protected] (B.N.S.); Tel.: +49-341-9722912 (B.N.S.) Received: 5 June 2020; Accepted: 30 June 2020; Published: 30 June 2020 Abstract: The vascular system is critical infrastructure that transports oxygen and nutrients around the body, and dynamically adapts its function to an array of environmental changes. To fulfil the demands of diverse organs, each with unique functions and requirements, the vascular system displays vast regional heterogeneity as well as specialized cell types. Our understanding of the heterogeneity of vascular cells and the molecular mechanisms that regulate their function is beginning to benefit greatly from the rapid development of single cell technologies. Recent studies have started to analyze and map vascular beds in a range of organs in healthy and diseased states at single cell resolution. The current review focuses on recent biological insights on the vascular system garnered from single cell analyses. We cover the themes of vascular heterogeneity, phenotypic plasticity of vascular cells in pathologies such as atherosclerosis and cardiovascular disease, as well as the contribution of defective microvasculature to the development of neurodegenerative disorders such as Alzheimer’s disease. Further adaptation of single cell technologies to study the vascular system will be pivotal in uncovering the mechanisms that drive the array of diseases underpinned by vascular dysfunction.
    [Show full text]
  • Contributions of the Renin Angiotensin System to Fear Memory and Fear Conditioned Cardiovascular Responses
    Contributions of the Renin Angiotensin System to Fear Memory and Fear Conditioned Cardiovascular Responses by Adam Swiercz B.S. in Biology, May 2006, The George Washington University M.P.S. in Molecular Biotechnology, May 2009, The George Washington University M.S. in Physiology, May 2011, Georgetown University A Dissertation submitted to The Faculty of The Columbian College of Arts & Sciences of The George Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy January 10, 2020 Dissertation co-directed by Paul J. Marvar Associate Professor of Pharmacology and Physiology and David Mendelowitz Professor of Pharmacology & Physiology The Columbian College of Arts and Sciences of The George Washington University certifies that Adam Swiercz has passed the Final Examination for the degree of Doctor of Philosophy as of October 2nd, 2019. This is the final and approved form of the dissertation. Contributions of the Renin Angiotensin System to Fear Memory and Fear Conditioned Cardiovascular Responses Adam Swiercz Dissertation Research Committee: Paul J. Marvar, Associate Professor of Pharmacology & Physiology, Dissertation Co-Director David Mendelowitz, Professor of Pharmacology & Physiology, Dissertation Co-Director Abigail Polter, Assistant Professor of Pharmacology & Physiology, Committee Member Colin Young, Assistant Professor of Pharmacology & Physiology, Committee Member ii © Copyright 2020 by Adam Swiercz All rights reserved iii Acknowledgements I would like to thank and acknowledge Dr. Paul Marvar, whose mentorship has made this dissertation possible. It has been a pleasure working in your lab, and I am truly grateful for your support and encouragement throughout the years. Thanks to the current and former members of the Marvar lab who have made my time at GW a rewarding and enjoyable experience.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • C-Terminal Region (ARP58987 P050) Data Sheet
    CASP3 antibody - C-terminal region (ARP58987_P050) Data Sheet Product Number ARP58987_P050 Product Name CASP3 antibody - C-terminal region (ARP58987_P050) Size 50ug Gene Symbol CASP3 Alias Symbols CPP32; CPP32B; SCA-1 Nucleotide Accession# NM_032991 Protein Size (# AA) 277 amino acids Molecular Weight 12kDa Product Format Lyophilized powder NCBI Gene Id 836 Host Rabbit Clonality Polyclonal Official Gene Full Name Caspase 3, apoptosis-related cysteine peptidase Gene Family CASP This is a rabbit polyclonal antibody against CASP3. It was validated on Western Blot by Aviva Systems Biology. At Aviva Systems Biology we manufacture rabbit polyclonal antibodies on a large scale (200-1000 Description products/month) of high throughput manner. Our antibodies are peptide based and protein family oriented. We usually provide antibodies covering each member of a whole protein family of your interest. We also use our best efforts to provide you antibodies recognize various epitopes of a target protein. For availability of antibody needed for your experiment, please inquire (). Peptide Sequence Synthetic peptide located within the following region: NLKYEVRNKNDLTREEIVELMRDVSKEDHSKRSSFVCVLLSHGEEGIIFG CASP3 is a protein which is a member of the cysteine-aspartic acid protease (caspase) family. Sequential activation of caspases plays a central role in the execution-phase of cell apoptosis. Caspases exist as inactive proenzymes which undergo proteolytic processing at conserved aspartic residues to produce two subunits, Description of Target large and small, that dimerize to form the active enzyme. This protein cleaves and activates caspases 6, 7 and 9, and the protein itself is processed by caspases 8, 9 and 10. It is the predominant caspase involved in the cleavage of amyloid-beta 4A precursor protein, which is associated with neuronal death in Alzheimer's disease.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • PLEKHM2 (NM 015164) Human Recombinant Protein – TP320299
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for TP320299 PLEKHM2 (NM_015164) Human Recombinant Protein Product data: Product Type: Recombinant Proteins Description: Recombinant protein of human pleckstrin homology domain containing, family M (with RUN domain) member 2 (PLEKHM2) Species: Human Expression Host: HEK293T Tag: C-Myc/DDK Predicted MW: 112.6 kDa Concentration: >50 ug/mL as determined by microplate BCA method Purity: > 80% as determined by SDS-PAGE and Coomassie blue staining Buffer: 25 mM Tris.HCl, pH 7.3, 100 mM glycine, 10% glycerol Preparation: Recombinant protein was captured through anti-DDK affinity column followed by conventional chromatography steps. Storage: Store at -80°C. Stability: Stable for 12 months from the date of receipt of the product under proper storage and handling conditions. Avoid repeated freeze-thaw cycles. RefSeq: NP_055979 Locus ID: 23207 UniProt ID: Q8IWE5 RefSeq Size: 4231 Cytogenetics: 1p36.21 RefSeq ORF: 3057 Synonyms: SKIP This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 2 PLEKHM2 (NM_015164) Human Recombinant Protein – TP320299 Summary: This gene encodes a protein that binds the plus-end directed microtubule motor protein kinesin, together with the lysosomal GTPase Arl8, and is required for lysosomes to distribute away from the microtubule-organizing center. The encoded protein belongs to the multisubunit BLOC-one-related complex that regulates lysosome positioning.
    [Show full text]