Regulation of the Human Vascular Smooth Muscle Cell Transcriptome by Extracellular Matrix Stiffness

Total Page:16

File Type:pdf, Size:1020Kb

Regulation of the Human Vascular Smooth Muscle Cell Transcriptome by Extracellular Matrix Stiffness University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2017 Regulation Of The Human Vascular Smooth Muscle Cell Transcriptome By Extracellular Matrix Stiffness Christopher Yu University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Cell Biology Commons, and the Genetics Commons Recommended Citation Yu, Christopher, "Regulation Of The Human Vascular Smooth Muscle Cell Transcriptome By Extracellular Matrix Stiffness" (2017). Publicly Accessible Penn Dissertations. 2710. https://repository.upenn.edu/edissertations/2710 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/2710 For more information, please contact [email protected]. Regulation Of The Human Vascular Smooth Muscle Cell Transcriptome By Extracellular Matrix Stiffness Abstract Arterial stiffness is a risk factor for several cardiometabolic diseases and is caused by pathological remodeling of the vascular extracellular matrix (ECM). Vascular smooth muscle cells (VSMCs) respond to ECM stiffness by proliferating, migrating, and further remodeling the vascular ECM, thus contributing to vascular disease like atherosclerosis and hypertension. VSMCs along the vasculature are highly diverse as they arise from different embryologic origins, reside in ECMs of diverse compositions, and are exposed to various mechanical forces. This dissertation aims to understand how ECM stiffness regulates the transcriptional response of VSMCs from different origins, namely aortic (Ao) and coronary (Co) VSMCs. We conducted deep sequencing of RNA from Ao and Co VSMCs grown on engineered polyacrylamide hydrogel surfaces tuned to physiologic and pathologic stiffness. Using several bioinformatic approaches, we compared the transcriptional landscapes in Ao and Co VSMCs by looking at whole-gene level expression, splicing, and conservation properties, with a focus on long non-coding RNAs (lncRNAs), as they compose a significant portion of the unexplored VSMC transcriptome. We found evidence suggesting that the overall transcriptional response to stiffness may be conserved across species and cell types, and that stiffness significantly dictates VSMC transcriptional identity over contributions from embryologic origins. However, we also discovered instances of origin-specific stiffness esponsesr in stiffness-mediated lncRNA expression and stiffness-mediated splicing. We identified a highly correlated network of adjacent stiffness-sensitive lncRNAs-protein coding gene pairs, which led us to experimentally interrogate the lncRNA PACER as a regulator of stiffness-mediate expression. We surprisingly found that PACER’s previously established regulatory pathway is absent in VSMCs. Using enrichment methods, we identified TBX5 and show that it is a stiffness-sensitive transcription factor specific ot Co VSMCs. We also demonstrated a new role for MALAT1 as a lncRNA regulator of stiffness-dependent VSMC proliferation and migration. Thus, this dissertation reveals many novel characteristics of the VSMC stiffness-regulated transcriptome that may have clinical utility in understanding and managing the pathogenesis of arterial stiffening. Degree Type Dissertation Degree Name Doctor of Philosophy (PhD) Graduate Group Bioengineering First Advisor Daniel J. Rader Second Advisor Richard K. Assoian Keywords Extracellular matrix stiffness, Long non-coding RNAs, Transcriptome, Vascular smooth muscle cells, Vascular stiffness Subject Categories Cell Biology | Genetics This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/2710 REGULATION OF THE HUMAN VASCULAR SMOOTH MUSCLE CELL TRANSCRIPTOME BY EXTRACELLULAR MATRIX STIFFNESS Christopher Yu A DISSERTATION in Bioengineering Presented to the Faculties of the University of Pennsylvania in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy 2017 Supervisor of Dissertation Co-Supervisor of Dissertation ________________________ _________________________ Daniel J. Rader, M.D. Richard K. Assoian, Ph.D. Seymour Gray Professor of Molecular Medicine Professor of Pharmacology Graduate Group Chairperson _______________________ Ravi Radhakrishnan, Ph.D. Professor of Bioengineering; Professor of Chemical and Biomolecular Engineering Dissertation Committee: (Chair): Peter F. Davies, Ph.D., Sc.D., Professor of Pathology & Laboratory Medicine; Professor of Bioengineering Rebecca G. Wells, M.D., Professor of Medicine Christopher S. Chen, M.D., Ph.D., Professor of Biomedical Engineering (Boston University) REGULATION OF THE HUMAN VASCULAR SMOOTH MUSCLE CELL TRANSCRIPTOME BY EXTRACELLULAR MATRIX STIFFNESS COPYRIGHT 2017 Christopher Ku Yu This work is licensed under the Creative Commons Attribution- NonCommercial-ShareAlike 3.0 License To view a copy of this license, visit https://creativecommons.org/licenses/by-nc-sa/3.0/us/ Dedication I dedicate this thesis to my parents (Robin H. Yu, M.D., and Estrelita Ku, M.D.), who prioritized and spared no expense on the education and life opportunities for me and my brothers. This thesis and my pursuit of a combined MD/PhD degree would never have been possible without their life-long support. I also dedicate this thesis to my incredible wife, Sue Lee, who put her own Ph.D studies on hold to take care of her sick mom. She has the biggest heart for family. Without her constant support, I could not have completed my dissertation. En Taro Adun iii ACKNOWLEDGMENTS Early in my Ph.D, I assumed that my prior research experiences in high school and college would have made my Ph.D experience smooth and straightforward. But it became quickly apparent that I had a lot to learn. There’s a lot of people, programs, and organizations to thank along the way for teaching me and providing me scientific, financial, and social support. First, I have to thank all the mentors and teachers I had before starting my Ph.D. who gave me a foundation for critical thinking and the scientific method. Thanks to everyone in the Rader Lab, Assoian Lab, and Chen Lab. I was lucky to be immersed in three different scientific environments utilizing microfabrication/tissue engineering, cell and molecular biology, computation/next-generation sequencing and human genetics. From the Rader Lab: Deepti Abbey, Guha Arunkumar, Rob Bauer, Xin Bi, Linda Carmichael, Stephanie DerOhannessian, Susannah Elwyn, Nicholas Hand, Sumeet Khetarpal, Hyein Kim, Wen Lin, Nicholas Lyssenko, Dawn Marchadier, Minal Mehta, Sylvia Nuremberg, Amirth Rodrigues, Kevin Trindade, Cathy Warford. From the Assoian Lab: Nate Bade, Yong Ho Bae, Paola Castagnino, Beth Hawthorne, Shulin Liu, Keeley Mui, Ziba Razinia, Ryan Von Kleeck, and Tina Xu. From the Chen Lab: Colin Choi, Peter Galie, and Michael Yang. I’d also like to thank others from other labs and cores. The Reily Lab (when they were at Penn), especially, Rachel Ballantyne and Chenyi Xue. The CDB microscopy core and its director Andrea Stout. The ITMAT biomechanics core, especially Jeff Byfield. Thanks my committee members Peter Davies and Rebecca Wells for their never-ending support through both the ups and downs of a PhD Study. Keeping me on track during thesis committee meetings and being very accommodating to me. The NIH funding sources: HL119346 and T32HL007954-17 I would also like to acknowledge all my friends for all their support and for the shared fun times through all the years. All the people I started med school, the group collectively known as zergling rush, especially Mike Sheng, Ray Zhang, James Hui, and Nabil Thalji. My long-time friends from college, Yang Li and Sophie Su. To the friends I met along the Ph.D path, especially Rosa Maria Alvarez, Keeley Mui, Kristy Ou, and Sean Yeldell. All the members of Penn Lions, for keeping me physically active, culturally active, and feel young. Thanks to my family. My mom, my dad, and my brothers, Robinson and Michael. My dad got sick and passed away toward the end of my Ph.D, but our family stuck together to support each other through that difficult time. As this thesis is dedicated to my mom and dad, I literally could not have completed this dissertation without them. Lastly, thank you so so much to my loving wife, Sue Lee. She has given me unlimited support in every aspect of my life, not only during my Ph.D studies, but almost every day in the last 10 years of knowing each other. She will discuss with me ad nauseam any scientific and non-scientific issue that I cannot believe or understand. She has helped iv me with all my grammar issues, even in writing of this dissertation, and practiced presentations with me. She has always been there to keep me sane through all the strings of failed experiments and turbulent times in life. I love her very much and owe her my deepest gratitude. v ABSTRACT REGULATION OF THE HUMAN VASCULAR SMOOTH MUSCLE CELL TRANSCRIPTOME BY EXTRACELLULAR MATRIX STIFFNESS Christopher Yu Daniel J. Rader, M.D. Richard K. Assoian, Ph.D. Arterial stiffness is a risk factor for several cardiometabolic diseases and is caused by pathological remodeling of the vascular extracellular matrix (ECM). Vascular smooth muscle cells (VSMCs) respond to ECM stiffness by proliferating, migrating, and further remodeling the vascular ECM, thus contributing to vascular disease like atherosclerosis and hypertension. VSMCs along the vasculature are highly diverse as they arise from different embryologic origins, reside in ECMs of diverse compositions, and are exposed to various mechanical forces.
Recommended publications
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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 Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [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]
  • Transcriptional Control of Tissue-Resident Memory T Cell Generation
    Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019 © 2019 Filip Cvetkovski All rights reserved ABSTRACT Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Tissue-resident memory T cells (TRM) are a non-circulating subset of memory that are maintained at sites of pathogen entry and mediate optimal protection against reinfection. Lung TRM can be generated in response to respiratory infection or vaccination, however, the molecular pathways involved in CD4+TRM establishment have not been defined. Here, we performed transcriptional profiling of influenza-specific lung CD4+TRM following influenza infection to identify pathways implicated in CD4+TRM generation and homeostasis. Lung CD4+TRM displayed a unique transcriptional profile distinct from spleen memory, including up-regulation of a gene network induced by the transcription factor IRF4, a known regulator of effector T cell differentiation. In addition, the gene expression profile of lung CD4+TRM was enriched in gene sets previously described in tissue-resident regulatory T cells. Up-regulation of immunomodulatory molecules such as CTLA-4, PD-1, and ICOS, suggested a potential regulatory role for CD4+TRM in tissues. Using loss-of-function genetic experiments in mice, we demonstrate that IRF4 is required for the generation of lung-localized pathogen-specific effector CD4+T cells during acute influenza infection. Influenza-specific IRF4−/− T cells failed to fully express CD44, and maintained high levels of CD62L compared to wild type, suggesting a defect in complete differentiation into lung-tropic effector T cells.
    [Show full text]
  • Contribution of Enhancer-Driven and Master-Regulator Genes to Autoimmune Disease Revealed Using Functionally Informed SNP-To-Gene Linking Strategies Kushal K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.02.279059; this version posted March 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Contribution of enhancer-driven and master-regulator genes to autoimmune disease revealed using functionally informed SNP-to-gene linking strategies Kushal K. Dey1, Steven Gazal1, Bryce van de Geijn 1,3, Samuel Sungil Kim 1,4, Joseph Nasser5, Jesse M. Engreitz5, Alkes L. Price 1,2,5 1 Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA 2 Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 3 Genentech, South San Francisco, CA 4 Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 5 Broad Institute of MIT and Harvard, Cambridge, MA, USA Abstract Gene regulation is known to play a fundamental role in human disease, but mechanisms of regulation vary greatly across genes. Here, we explore the con- tributions to disease of two types of genes: genes whose regulation is driven by enhancer regions as opposed to promoter regions (Enhancer-driven) and genes that regulate many other genes in trans (Master-regulator). We link these genes to SNPs using a comprehensive set of SNP-to-gene (S2G) strategies and apply stratified LD score regression to the resulting SNP annotations to draw three main conclusions about 11 autoimmune diseases and blood cell traits (average Ncase=13K across 6 autoimmune diseases, average N=443K across 5 blood cell traits).
    [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]
  • Accompanies CD8 T Cell Effector Function Global DNA Methylation
    Global DNA Methylation Remodeling Accompanies CD8 T Cell Effector Function Christopher D. Scharer, Benjamin G. Barwick, Benjamin A. Youngblood, Rafi Ahmed and Jeremy M. Boss This information is current as of October 1, 2021. J Immunol 2013; 191:3419-3429; Prepublished online 16 August 2013; doi: 10.4049/jimmunol.1301395 http://www.jimmunol.org/content/191/6/3419 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2013/08/20/jimmunol.130139 Material 5.DC1 References This article cites 81 articles, 25 of which you can access for free at: http://www.jimmunol.org/content/191/6/3419.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on October 1, 2021 • 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 © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Global DNA Methylation Remodeling Accompanies CD8 T Cell Effector Function Christopher D. Scharer,* Benjamin G. Barwick,* Benjamin A. Youngblood,*,† Rafi Ahmed,*,† and Jeremy M.
    [Show full text]
  • Negative Regulators of Colonic Peripheral Regulatory T Cell Development Teresa Lei Ai Washington University in St
    Washington University in St. Louis Washington University Open Scholarship Arts & Sciences Electronic Theses and Dissertations Arts & Sciences Spring 5-15-2018 Negative Regulators of Colonic Peripheral Regulatory T Cell Development Teresa Lei Ai Washington University in St. Louis Follow this and additional works at: https://openscholarship.wustl.edu/art_sci_etds Part of the Allergy and Immunology Commons, Immunology and Infectious Disease Commons, and the Medical Immunology Commons Recommended Citation Ai, Teresa Lei, "Negative Regulators of Colonic Peripheral Regulatory T Cell Development" (2018). Arts & Sciences Electronic Theses and Dissertations. 1509. https://openscholarship.wustl.edu/art_sci_etds/1509 This Dissertation is brought to you for free and open access by the Arts & Sciences at Washington University Open Scholarship. It has been accepted for inclusion in Arts & Sciences Electronic Theses and Dissertations by an authorized administrator of Washington University Open Scholarship. For more information, please contact [email protected]. WASHINGTON UNIVERSITY IN ST. LOUIS Division of Biology and Biomedical Sciences Immunology Dissertation Examination Committee: Chyi-Song Hsieh, Chair Paul M. Allen Kenneth M. Murphy Gwendalyn J. Randolph Wayne M. Yokoyama Negative Regulators of Colonic Peripheral Regulatory T Cell Development By Teresa Lei Ai A dissertation presented to The Graduate School of Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy May 2018 St. Louis, Missouri
    [Show full text]
  • Evolutionarily Conserved Tbx5–Wnt2/2B Pathway Orchestrates Cardiopulmonary Development
    Evolutionarily conserved Tbx5–Wnt2/2b pathway orchestrates cardiopulmonary development Jeffrey D. Steimlea,b,c, Scott A. Rankind,e,f,g, Christopher E. Slagleh,i,j,k, Jenna Bekenya,b,c, Ariel B. Rydeena,b,c, Sunny Sun-Kin Chanl,m, Junghun Kweona,b,c, Xinan H. Yanga,b,c, Kohta Ikegamia,b,c, Rangarajan D. Nadadura,b,c, Megan Rowtona,b,c, Andrew D. Hoffmanna,b,c, Sonja Lazarevica,b,c, William Thomasn,o, Erin A. T. Boyle Andersonp, Marko E. Horbn,o, Luis Luna-Zuritaq,r, Robert K. Hom, Michael Kybal,m, Bjarke Jensens, Aaron M. Zornd,e,f,g, Frank L. Conlonh,i,j,k, and Ivan P. Moskowitza,b,c,1 aDepartment of Pediatrics, University of Chicago, Chicago, IL 60637; bDepartment of Pathology, University of Chicago, Chicago, IL 60637; cDepartment of Human Genetics, University of Chicago, Chicago, IL 60637; dCenter for Stem Cell and Organoid Medicine, Cincinnati Children’s Research Foundation, Cincinnati, OH 45229; eDepartment of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229; fDivision of Developmental Biology, Perinatal Institute, Cincinnati Children’s Research Foundation, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229; gDepartment of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229; hDepartment of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; iDepartment of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; jIntegrative Program for Biological and Genome Sciences, University of North
    [Show full text]
  • The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes Yunkai Dai Clemson University, [email protected]
    Clemson University TigerPrints All Dissertations Dissertations May 2019 The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes Yunkai Dai Clemson University, [email protected] Follow this and additional works at: https://tigerprints.clemson.edu/all_dissertations Recommended Citation Dai, Yunkai, "The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes" (2019). All Dissertations. 2377. https://tigerprints.clemson.edu/all_dissertations/2377 This Dissertation is brought to you for free and open access by the Dissertations at TigerPrints. It has been accepted for inclusion in All Dissertations by an authorized administrator of TigerPrints. For more information, please contact [email protected]. THE ROLE OF TBX5 IN SINOATRIAL NODE DIFFERENTIATION IN MOUSE EMBRYONIC STEM CELL DERIVED CARDIOMYOCYTES A Dissertation Presented to the Graduate School of Clemson University In Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Bioengineering by Yunkai Dai May 2019 Accepted by: Dr Ann Foley, Committee Chair Dr Agneta Simionescu Dr Robin C. Muise-Helmericks Dr Ying Mei ABSTRACT The sinoatrial node of the mouse embryo arises from the wall of the right atrium near the border of the sinus venosus. Early in development this region expresses the transcription factor Tbx5. Because of this, Tbx5 is thought to sit at the apex of a transcriptional cascade leading to sinoatrial node (SAN) differentiation. To test this we produced a mouse embryonic stem cell line B1 (pTripZ-mTbx5; αMHC::GFP) that conditionally overexpresses Tbx5, to determine if this would lead to enhanced SAN differentiation. We found that ES cells overexpressing Tbx5 showed enhanced overall cardiac differentiation and that cardiac cells showed increased beat rates as compared control embryos.
    [Show full text]
  • SUPPLEMENTAL FIGURE LEGENDS Supplemental Figure S1. RBPJ
    Xie et al. SUPPLEMENTAL FIGURE LEGENDS Supplemental Figure S1. RBPJ correlates with BTIC marker expression. A-D. The TCGA GBM dataset was downloaded and correlations analyzed by R. RBPJ mRNA levels were highly correlated with (A) Olig2, (B) Sox2, (C) CD133, and (D) Sox4 levels. E. RBPJ is preferentially expressed in proneural glioblastomas. The glioblastoma TCGA dataset was interrogated for RBPJ mRNA expression segregated by transcriptional profile. The proneural tumors were further divided into G-CIMP (glioma CpG-island methylator phenotype) or non-G-CIMP. **, p < 0.01. ****, p < 0.0001. *****, p < 0.00001. Supplemental Figure S2. Targeting RBPJ induces BTIC apoptosis. A. 3691 BTICs were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Lysates were prepared and immunoblotted with the indicated antibodies. shRNA-mediated knockdown of RBPJ was associated with increased cleaved (activated) PARP. B. 3691 BTICs were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Apoptosis measured by Annexin V staining. Data are presented as mean ± SEM (two- way ANOVA; **, p < 0.01; n = 3). Supplemental Figure S3. Targeting RBPJ does not affect non-BTIC proliferation. Non-BTICs (Top, 3691; Bottom, 4121) were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Cell proliferation was measured by CellTiter-Glo. 42 Xie et al. Supplemental Figure S4. RBPJ induces transcriptional profiles in BTICs distinct from Notch activation. A. In parallel experiments, 3691 BTICs were either treated with DAPT (at either 5 μM or 10 μM) vs. vehicle control (DMSO) or transduced with shRBPJ vs. shCONT. RNA-Seq was performed and the results displayed as a heat map with normalization to the relevant control.
    [Show full text]
  • SOX4 Is Activated by C-MYC in Prostate Cancer
    Medical Oncology (2019) 36:92 https://doi.org/10.1007/s12032-019-1317-6 ORIGINAL PAPER SOX4 is activated by C‑MYC in prostate cancer Hongyan Dong1,2 · Jing Hu1 · Lin Wang3 · Mei Qi1 · Ning Lu1 · Xiao Tan2 · Muyi Yang1 · Xinnuo Bai4 · Xuemei Zhan2 · Bo Han1,5 Received: 26 July 2019 / Accepted: 12 September 2019 / Published online: 27 September 2019 © Springer Science+Business Media, LLC, part of Springer Nature 2019 Abstract Although MYC proto-oncogene (C-MYC) amplifcation has been consistently reported to be a potential marker for prostate cancer (PCa) progression and prognosis, the clinicopathological and prognostic signifcance of C-MYC protein expression remains controversial. Overexpression of SOX4 has been shown to play important roles in multiple cancers including PCa. However, the link between these two critical genetic aberrations is unclear. In the current study, we showed that C-MYC was overexpressed in 16.2% (17/105) of Chinese patients with localized PCa. Overexpression of C-MYC was signifcantly associated with high Gleason scores (P = 0.012) and high Ki67 labeling index (P = 0.005). C-MYC overexpression was cor- related with cancer-related mortality and suggested to be an unfavorable prognostic factor in Chinese PCa patients (P = 0.018). Overexpression of C-MYC is associated with SOX4 overexpression in PCa tissues. Notably, SOX4 is a direct target gene of C-MYC; C-MYC activates SOX4 expression via binding to its promoter. In addition, Co-IP analysis demonstrated a physi- cal interaction between C-MYC and SOX4 protein in PCa cells. Clinically, C-MYC+/SOX4+ characterized poor prognosis in a subset of PCa patients.
    [Show full text]
  • Transcription Factor Gene Expression Profiling and Analysis of SOX Gene Family Transcription Factors in Human Limbal Epithelial
    Transcription factor gene expression profiling and analysis of SOX gene family transcription factors in human limbal epithelial progenitor cells Der Naturwissenschaftlichen Fakultät der Friedrich-Alexander-Universität Erlangen-Nürnberg zur Erlangung des Doktorgrades Dr. rer. nat. vorgelegt von Dr. med. Johannes Menzel-Severing aus Bonn Als Dissertation genehmigt von der Naturwissenschaftlichen Fakultät der Friedrich-Alexander-Universität Erlangen-Nürnberg Tag der mündlichen Prüfung: 7. Februar 2018 Vorsitzender des Promotionsorgans: Prof. Dr. Georg Kreimer Gutachter: Prof. Dr. Andreas Feigenspan Prof. Dr. Ursula Schlötzer-Schrehardt 1 INDEX 1. ABSTRACTS Page 1.1. Abstract in English 4 1.2. Zusammenfassung auf Deutsch 7 2. INTRODUCTION 2.1. Anatomy and histology of the cornea and the corneal surface 11 2.2. Homeostasis of corneal epithelium and the limbal stem cell paradigm 13 2.3. The limbal stem cell niche 15 2.4. Cell therapeutic strategies in ocular surface disease 17 2.5. Alternative cell sources for transplantation to the corneal surface 18 2.6. Transcription factors in cell differentiation and reprogramming 21 2.7. Transcription factors in limbal epithelial cells 22 2.8. Research question 25 3. MATERIALS AND METHODS 3.1. Human donor corneas 27 3.2. Laser Capture Microdissection (LCM) 28 3.3. RNA amplification and RT2 profiler PCR arrays 29 3.4. Real-time PCR analysis 33 3.5. Immunohistochemistry 34 3.6. Limbal epithelial cell culture 38 3.7. Transcription-factor knockdown/overexpression in vitro 39 3.8. Proliferation assay 40 3.9. Western blot 40 3.10. Statistical analysis 41 2 4. RESULTS 4.1. Quality control of LCM-isolated and amplified RNA 42 4.2.
    [Show full text]