Id3 Induces an Elk-1- and Caspase-8-Dependent Apoptotic Pathway In

Total Page:16

File Type:pdf, Size:1020Kb

Id3 Induces an Elk-1- and Caspase-8-Dependent Apoptotic Pathway In ID3 INDUCES AN ELK-1- AND CASPASE-8-DEPENDENT APOPTOTIC PATHWAY IN SQUAMOUS CARCINOMA CELLS A Dissertation submitted to the Faculty of the Graduate School of Arts and Sciences of Georgetown University in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biochemistry and Molecular & Cellular Biology By You-shin Chen, M.S. Washington, DC November 18, 2014 Copyright 2014 by You-shin Chen All Rights Reserved ii ID3 INDUCES AN ELK-1- AND CASPASE-8-DEPENDENT APOPTOTIC PATHWAY IN SQUAMOUS CARCINOMA CELLS You-shin Chen, M.S. Thesis Advisor: Dean S. Rosenthal, Ph.D. ABSTRACT Inhibitors of differentiation/DNA binding (Id) proteins are helix-loop-helix (HLH) transcription factors. The Id protein family (Id1-Id4) mediates tissue homeostasis by regulating cellular processes including differentiation, proliferation, and apoptosis. Previously, we found that Id3 induced apoptosis in immortalized human keratinocytes (Simbulan-Rosenthal et al., 2006), consistent with its role as a tumor suppressor (Richter et al., 2012; Schmitz et al., 2012). To investigate the role of Id3 in malignant SCC cells (A431), a tetracycline-regulated inducible system was used to induce Id3 in cell culture and mouse xenograft models. We found that upon Id3 induction, there was a decrease in cell number under low serum conditions, as well as in soft agar. Microarray, RT-PCR, immunoblot, siRNA, and inhibitor studies revealed that Id3 induced expression of Elk-1, an ETS-domain transcription factor, inducing procaspase-8 expression and activation. Id3 deletion mutants revealed that 80 C-terminal amino acids, including the HLH, are important for Id3-induced apoptosis. In a mouse xenograft model, Id3 induction decreased tumor size by 30%. Using immunofluorescence analysis, we determined that the tumor size decrease was also mediated through apoptosis. Further, we show that Id3 synergizes with 5-FU and cisplatin therapies for non-melanoma skin cancer cells. Our studies have shown that Id3 induces apoptosis in SCC cells via an Elk-1- and caspase-8-dependent pathway, and this information can provide some strategic insights for new SCC treatments. iii Acknowledgements First, I would like to sincerely thank my thesis advisor Dr. Dean Rosenthal for giving me the opportunity to work in his lab. Without his guidance, I couldn’t have completed the research and thesis writing. Besides, I feel fortunate to be given the opportunities to learn how to lead a research team comprised of people from different scientific and cultural background. The past years in the Rosenthal lab have been very productive, and I feel blessed to have this kind of training experience. I want to thank my committee members: Drs. Albanese, Chirikjian, Kasid, and Vasudevan and the Department Chair Dr. Crooke for their advice and feedback throughout the years. Special thanks to Dr. Cynthia Rosenthal, from whose life experience I find inspiring and for giving me the opportunities to develop teaching skills. I would also like to thank my colleagues along the way: Valerie, Kyle, Lucia, Virginia, Anirudh, and Maryam, for their constant encouragement and help in the lab. The good memories with them will always be remembered. Last but not least, I would like to thank my family and friends. My father Chin-tu, mother Tung-chin, sister You-jen, and brother Yu-fu form strongest support team to help me in any possible ways in the past years to complete my degree. My boyfriend Derrick has been my best friend, top cheerleader, and supporter. Derrick has offered me his warmest hands during my down times, without whom, the completion of my research would not be possible. I also want to thank my best friends, Lynn, Peiyu, and Tina who never abandon me even when I miss their important life events while pursuing this degree. Many thanks, You-shin iv Table of Contents Table of Abbreviations and Definitions ....................................................................................... viii List of Figures ............................................................................................................................... xv List of Tables ............................................................................................................................. xviii Chapter 1. Introduction ................................................................................................................... 1 1.1. Hypothesis, Rationale, and Research Aims .............................................................. 1 1.2. Introduction to Squamous Cell Carcinoma (SCC) .................................................... 2 1.2.1. Skin Cancer Statistics .................................................................................... 2 1.2.2. Risk Factors for Squamous Cell Carcinoma ................................................. 3 1.2.3. SCC Progression ........................................................................................... 4 1.2.4. Current Treatment Options for SCC ............................................................. 5 1.3. Introduction to Id Proteins ......................................................................................... 9 1.3.1. Id Genetic and Protein Information ............................................................... 9 1.3.2. Ids as HLH Transcription Factors ............................................................... 11 1.3.3. Ids as Regulators of Differentiation and Proliferation ................................ 17 1.3.4. Ids as Regulators of Apoptosis .................................................................... 21 1.3.5. Ids as Potential Tumor Suppressors ............................................................ 22 1.3.6. Ids and SCC ................................................................................................. 25 1.3.7. Ids and TCF Transcription Factors .............................................................. 27 1.4. Introduction to Apoptosis ........................................................................................ 34 1.4.1. Caspases ...................................................................................................... 34 1.4.2. Bcl-2 Family Proteins .................................................................................. 37 1.4.3. Apoptotic Signaling Pathways .................................................................... 39 1.4.4. Ids and Apoptosis ........................................................................................ 41 Chapter 2. Materials and Methods ................................................................................................ 43 2.1. Cell Culture ............................................................................................................. 43 2.2. Tet-inducible System ............................................................................................... 44 2.3. Plasmids .................................................................................................................. 47 2.4. Inducible Cell Lines ................................................................................................ 48 2.5. Fluorescently-labeled Cell Lines ............................................................................. 49 v 2.6. Growth Curve .......................................................................................................... 49 2.7. Cell Cycle Analysis ................................................................................................. 50 2.8. Cell Invasion Assay ................................................................................................. 51 2.9. Microarray Analysis ................................................................................................ 51 2.10. siRNA Transfection ................................................................................................. 51 2.11. Tumor Xenografts ................................................................................................... 52 2.12. Immunoblot Analysis .............................................................................................. 52 2.13. Immunostaining ....................................................................................................... 53 2.14. RT-PCR/qRT-PCR .................................................................................................. 53 2.15. Soft Agar Growth Assay ......................................................................................... 54 2.16. 5-FU and Cisplatin Treatment ................................................................................. 54 2.17. Statistical Analysis .................................................................................................. 55 Chapter 3. Results ......................................................................................................................... 56 3.1. Establishment of A431/Id3 and A431/Vc Cell Lines .............................................. 56 3.1.1. Endogenous Levels of Id3 are Down-regulated in A431 Cells ................... 56 3.1.2. Verification of Inducible A431/Id3 Clones ................................................. 57 3.2. Study of the Effects of Id3 in vitro .......................................................................... 59 3.2.1. Id3 Does Not Affect Cell Growth in Normal Serum, in Matrigel, or Invasion ....................................................................................................... 59 3.2.2. Id3 Reduces Viable Cell
Recommended publications
  • Peripheral T Cells Ets-1 Maintains IL-7 Receptor Expression In
    The Journal of Immunology Ets-1 Maintains IL-7 Receptor Expression in Peripheral T Cells Roland Grenningloh,*,† Tzong-Shyuan Tai,* Nicole Frahm,†,‡,1 Tomoyuki C. Hongo,‡ Adam T. Chicoine,‡ Christian Brander,†,‡,x,{ Daniel E. Kaufmann,†,‡,‖ and I-Cheng Ho*,† The expression of CD127, the IL-7–binding subunit of the IL-7 R, is tightly regulated during the development and activation of T cells and is reduced during chronic viral infection. However, the molecular mechanism regulating the dynamic expression of CD127 is still poorly understood. In this study, we report that the transcription factor Ets-1 is required for maintaining the expression of CD127 in murine peripheral T cells. Ets-1 binds to and activates the CD127 promoter, and its absence leads to reduced CD127 expression, attenuated IL-7 signaling, and impaired IL-7–dependent homeostatic proliferation of T cells. The expression of CD127 and Ets-1 is strongly correlated in human T cells. Both CD127 and Ets-1 expression are decreased in CD8+ T cells during HIV infection. In addition, HIV-associated loss of CD127 is only observed in Ets-1low effector memory and central memory but not in Ets-1high naive CD8+ T cells. Taken together, our data identify Ets-1 as a critical regulator of CD127 expression in T cells. The Journal of Immunology, 2011, 186: 969–976. nterleukin-7 signals are required for T cell development, GABPa or another Ets protein is responsible for maintaining maintaining the naive T cell pool, mounting proper primary CD127 expression in peripheral T cells is unknown. I responses, and inducing and maintaining CD4+ and CD8+ Ets-1 (E26 transformation-specific sequence) is the founding T cell memory (1–3).
    [Show full text]
  • Deciphering the Functions of Ets2, Pten and P53 in Stromal Fibroblasts in Multiple
    Deciphering the Functions of Ets2, Pten and p53 in Stromal Fibroblasts in Multiple Breast Cancer Models DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Julie Wallace Graduate Program in Molecular, Cellular and Developmental Biology The Ohio State University 2013 Dissertation Committee: Michael C. Ostrowski, PhD, Advisor Gustavo Leone, PhD Denis Guttridge, PhD Dawn Chandler, PhD Copyright by Julie Wallace 2013 Abstract Breast cancer is the second most common cancer in American women, and is also the second leading cause of cancer death in women. It is estimated that nearly a quarter of a million new cases of invasive breast cancer will be diagnosed in women in the United States this year, and approximately 40,000 of these women will die from breast cancer. Although death rates have been on the decline for the past decade, there is still much we need to learn about this disease to improve prevention, detection and treatment strategies. The majority of early studies have focused on the malignant tumor cells themselves, and much has been learned concerning mutations, amplifications and other genetic and epigenetic alterations of these cells. However more recent work has acknowledged the strong influence of tumor stroma on the initiation, progression and recurrence of cancer. Under normal conditions this stroma has been shown to have protective effects against tumorigenesis, however the transformation of tumor cells manipulates this surrounding environment to actually promote malignancy. Fibroblasts in particular make up a significant portion of this stroma, and have been shown to impact various aspects of tumor cell biology.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 9,109,232 B2 Schwartz Et Al
    US009 109232B2 (12) United States Patent (10) Patent No.: US 9,109,232 B2 Schwartz et al. (45) Date of Patent: Aug. 18, 2015 (54) ETS2 AND MESP1 GENERATE CARDIAC Islas et al., Transcription factors ETS2 and MESP1 transdifferentiate PROGENITORS FROM FIBROBLASTS human dermal fibroblasts into cardiac progenitors; PNAS, Published online before print Jul. 23, 2012, doi: 10.1073/pnas. 11202991.09, 2012.* (71) Applicants: University of Houston, Houston, TX Melotti et al., Ets-2 and c-Myb Act Independently in Regulating (US); Texas Heart Institute, Houston, Expression of the Hematopoietic StemCell Antigen CD34:JBC, vol. TX (US); The Texas A&M University 269, No. 41, pp. 25303-25309, 1994.* System, College Station, TX (US) Takahashi et al., Induction of pluripotent stem cells from adulthuman fibroblasts by defined factors; Cell, vol. 131, pp. 861-872, 2007.* Naldini et al. In vivo gene delivery and stable transduction of (72) Inventors: Robert J. Schwartz, Houston, TX (US); nondividing cells by a lentiviral vector; Science, vol. 272, pp. 263 Vladimir N. Potaman, Houston, TX 267, 1996.* (US); Jose Francisco Islas, Houston, TX European Patent Office; Office Action; European Application No. (US) 117114082; Jul 22, 2013. European Patent Office; Response to Office Action; European Appli (73) Assignees: University of Houston, Houston, TX cation No. 11711408.2; Aug. 30, 2013. Chinese Patent Office; Office Action; Chinese Patent Application No. (US); Texas Heart Institute, Houston, 2011800 19561.0; Sep. 18, 2013. TX (US); The Texas A&M University Chinese Patent Office; Office Action (English translation); Chinese System, College Station, TX (US) Patent Application No. 2011800 19561.0; Sep.
    [Show full text]
  • PAX5 Expression in Acute Leukemias: Higher B-Lineage Specificity Than Cd79a and Selective Association with T(8;21)-Acute Myelogenous Leukemia
    [CANCER RESEARCH 64, 7399–7404, October 15, 2004] PAX5 Expression in Acute Leukemias: Higher B-Lineage Specificity Than CD79a and Selective Association with t(8;21)-Acute Myelogenous Leukemia Enrico Tiacci,1 Stefano Pileri,2 Annette Orleth,1 Roberta Pacini,1 Alessia Tabarrini,1 Federica Frenguelli,1 Arcangelo Liso,3 Daniela Diverio,4 Francesco Lo-Coco,5 and Brunangelo Falini1 1Institutes of Hematology and Internal Medicine, University of Perugia, Perugia, Italy; 2Unit of Hematopathology, University of Bologne, Bologne, Italy; 3Section of Hematology, University of Foggia, Foggia, Italy; 4Department of Cellular Biotechnologies and Hematology, University La Sapienza of Rome, Rome, Italy; and 5Department of Biopathology, University Tor Vergata of Rome, Rome, Italy ABSTRACT (13, 16). PAX5 expression also occurs in the adult testis and in the mesencephalon and spinal cord during embryogenesis (17), suggesting an The transcription factor PAX5 plays a key role in the commitment of important role in the development of these tissues. hematopoietic precursors to the B-cell lineage, but its expression in acute Rearrangement of the PAX5 gene through reciprocal chromosomal leukemias has not been thoroughly investigated. Hereby, we analyzed routine biopsies from 360 acute leukemias of lymphoid (ALLs) and mye- translocations has been described in different types of B-cell malig- loid (AMLs) origin with a specific anti-PAX5 monoclonal antibody. Blasts nancies (18–23), and, more recently, PAX5 has also been shown to be from 150 B-cell ALLs showed strong PAX5 nuclear expression, paralleling targeted by aberrant hypermutation in Ͼ50% of diffuse large B-cell that of CD79a in the cytoplasm. Conversely, PAX5 was not detected in 50 lymphomas (24).
    [Show full text]
  • Loss of the NKX3.1 Tumorsuppressor Promotes the TMPRSS2-ERG
    Thangapazham et al. BMC Cancer 2014, 14:16 http://www.biomedcentral.com/1471-2407/14/16 RESEARCH ARTICLE Open Access Loss of the NKX3.1 tumorsuppressor promotes the TMPRSS2-ERG fusion gene expression in prostate cancer Rajesh Thangapazham, Francisco Saenz, Shilpa Katta, Ahmed A Mohamed, Shyh-Han Tan, Gyorgy Petrovics, Shiv Srivastava and Albert Dobi* Abstract Background: In normal prostate epithelium the TMPRSS2 gene encoding a type II serine protease is directly regulated by male hormones through the androgen receptor. In prostate cancer ERG protooncogene frequently gains hormonal control by seizing gene regulatory elements of TMPRSS2 through genomic fusion events. Although, the androgenic activation of TMPRSS2 gene has been established, little is known about other elements that may interact with TMPRSS2 promoter sequences to modulate ERG expression in TMPRSS2-ERG gene fusion context. Methods: Comparative genomic analyses of the TMPRSS2 promoter upstream sequences and pathway analyses were performed by the Genomatix Software. NKX3.1 and ERG genes expressions were evaluated by immunoblot or by quantitative Real-Time PCR (qRT-PCR) assays in response to siRNA knockdown or heterologous expression. QRT-PCR assay was used for monitoring the gene expression levels of NKX3.1-regulated genes. Transcriptional regulatory function of NKX3.1 was assessed by luciferase assay. Recruitment of NKX3.1 to its cognate elements was monitored by Chromatin Immunoprecipitation assay. Results: Comparative analysis of the TMPRSS2 promoter upstream sequences among different species revealed the conservation of binding sites for the androgen inducible NKX3.1 tumor suppressor. Defects of NKX3.1, such as, allelic loss, haploinsufficiency, attenuated expression or decreased protein stability represent established pathways in prostate tumorigenesis.
    [Show full text]
  • Further Delineation of Chromosomal Consensus Regions in Primary
    Leukemia (2007) 21, 2463–2469 & 2007 Nature Publishing Group All rights reserved 0887-6924/07 $30.00 www.nature.com/leu ORIGINAL ARTICLE Further delineation of chromosomal consensus regions in primary mediastinal B-cell lymphomas: an analysis of 37 tumor samples using high-resolution genomic profiling (array-CGH) S Wessendorf1,6, TFE Barth2,6, A Viardot1, A Mueller3, HA Kestler3, H Kohlhammer1, P Lichter4, M Bentz5,HDo¨hner1,PMo¨ller2 and C Schwaenen1 1Klinik fu¨r Innere Medizin III, Zentrum fu¨r Innere Medizin der Universita¨t Ulm, Ulm, Germany; 2Institut fu¨r Pathologie, Universita¨t Ulm, Ulm, Germany; 3Forschungsdozentur Bioinformatik, Universita¨t Ulm, Ulm, Germany; 4Abt. Molekulare Genetik, Deutsches Krebsforschungszentrum, Heidelberg, Germany and 5Sta¨dtisches Klinikum Karlsruhe, Karlsruhe, Germany Primary mediastinal B-cell lymphoma (PMBL) is an aggressive the expression of BSAP, BOB1, OCT2, PAX5 and PU1 was extranodal B-cell non-Hodgkin’s lymphoma with specific clin- added to the spectrum typical of PMBL features.9 ical, histopathological and genomic features. To characterize Genetically, a pattern of highly recurrent karyotype alterations further the genotype of PMBL, we analyzed 37 tumor samples and PMBL cell lines Med-B1 and Karpas1106P using array- with the hallmark of chromosomal gains of the subtelomeric based comparative genomic hybridization (matrix- or array- region of chromosome 9 supported the concept of a unique CGH) to a 2.8k genomic microarray. Due to a higher genomic disease entity that distinguishes PMBL from other B-cell non- resolution, we identified altered chromosomal regions in much Hodgkin’s lymphomas.10,11 Together with less specific gains on higher frequencies compared with standard CGH: for example, 2p15 and frequent mutations of the SOCS1 gene, a notable þ 9p24 (68%), þ 2p15 (51%), þ 7q22 (32%), þ 9q34 (32%), genomic similarity to classical Hodgkin’s lymphoma was þ 11q23 (18%), þ 12q (30%) and þ 18q21 (24%).
    [Show full text]
  • Additive Effects of Micrornas and Transcription Factors on CCL2 Production in Human White Adipose Tissue
    1248 Diabetes Volume 63, April 2014 Agné Kulyté,1 Yasmina Belarbi,1 Silvia Lorente-Cebrián,1 Clara Bambace,1 Erik Arner,1,2 Carsten O. Daub,3 Per Hedén,4 Mikael Rydén,1 Niklas Mejhert,1 and Peter Arner1 Additive Effects of MicroRNAs and Transcription Factors on CCL2 Production in Human White Adipose Tissue Adipose tissue inflammation is present in insulin- converged on the nuclear factor-kB pathway. In resistant conditions. We recently proposed conclusion, TF and miRNA-mediated regulation of a network of microRNAs (miRNAs) and transcription CCL2 production is additive and partly relayed by factors (TFs) regulating the production of the cell-specific networks in human adipose tissue that proinflammatory chemokine (C-C motif) ligand-2 may be important for the development of insulin (CCL2) in adipose tissue. We presently extended and resistance/type 2 diabetes. further validated this network and investigated if the Diabetes 2014;63:1248–1258 | DOI: 10.2337/db13-0702 METABOLISM circuits controlling CCL2 can interact in human adipocytes and macrophages. The updated subnetwork predicted that miR-126/-193b/-92a White adipose tissue (WAT) function plays an important control CCL2 production by several TFs, including role in the development of insulin resistance/type 2 di- v-ets erythroblastosis virus E26 oncogene homolog 1 abetes. Fat cells present in WAT secrete a number of (avian) (ETS1), MYC-associated factor X (MAX), molecules, collectively termed adipokines, which affect and specificity protein 12 (SP1). This was confirmed insulin sensitivity by autocrine and/or paracrine mecha- in human adipocytes by the observation that gene nisms (1,2). In insulin-resistant obese subjects, WAT silencing of ETS1, MAX, or SP1 attenuated CCL2 displays a chronic low-grade inflammation, which is production.
    [Show full text]
  • Lncegfl7os Regulates Human Angiogenesis by Interacting
    RESEARCH ARTICLE LncEGFL7OS regulates human angiogenesis by interacting with MAX at the EGFL7/miR-126 locus Qinbo Zhou1†, Bo Yu1†*, Chastain Anderson1, Zhan-Peng Huang2, Jakub Hanus1, Wensheng Zhang3, Yu Han4, Partha S Bhattacharjee5, Sathish Srinivasan6, Kun Zhang3, Da-zhi Wang2, Shusheng Wang1,7* 1Department of Cell and Molecular Biology, Tulane University, New Orleans, United States; 2Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, United States; 3Department of Computer Science, Xavier University, New Orleans, United States; 4Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, United States; 5Department of Biology, Xavier University, New Orleans, United States; 6Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma, United States; 7Department of Ophthalmology, Tulane University, New Orleans, United States Abstract In an effort to identify human endothelial cell (EC)-enriched lncRNAs,~500 lncRNAs were shown to be highly restricted in primary human ECs. Among them, lncEGFL7OS, located in the opposite strand of the EGFL7/miR-126 gene, is regulated by ETS factors through a bidirectional promoter in ECs. It is enriched in highly vascularized human tissues, and upregulated in the hearts of dilated cardiomyopathy patients. LncEGFL7OS silencing impairs angiogenesis as shown by EC/fibroblast co-culture, in vitro/in vivo and ex vivo human choroid sprouting angiogenesis assays, while lncEGFL7OS overexpression has the opposite function. Mechanistically, *For correspondence: lncEGFL7OS is required for MAPK and AKT pathway activation by regulating EGFL7/miR-126 [email protected] (BY); expression. MAX protein was identified as a lncEGFL7OS-interacting protein that functions to [email protected] (SW) regulate histone acetylation in the EGFL7/miR-126 promoter/enhancer.
    [Show full text]
  • Genomic Profiling of Adult Acute Lymphoblastic Leukemia by Single
    SUPPLEMENTARY APPENDIX Genomic profiling of adult acute lymphoblastic leukemia by single nucleotide polymorphism oligonucleotide microarray and comparison to pediatric acute lymphoblastic leukemia Ryoko Okamoto,1 Seishi Ogawa,2 Daniel Nowak,1 Norihiko Kawamata,1 Tadayuki Akagi,1,3 Motohiro Kato,2 Masashi Sanada,2 Tamara Weiss,4 Claudia Haferlach,4 Martin Dugas,5 Christian Ruckert,5 Torsten Haferlach,4 and H. Phillip Koeffler1,6 1Division of Hematology and Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, USA; 2Cancer Genomics Project, Graduate School of Medicine, University of Tokyo, Tokyo, Japan; 3Department of Stem Cell Biology, Graduate School of Medical Science, Kanazawa University 4MLL Munich Leukemia Laboratory, Munich, Germany; 5Department of Medical Informatics and Biomathematics, University of Münster, Münster, Germany; 6Cancer Science Institute of Singapore, National University of Singapore, Singapore Citation: Okamoto R, Ogawa S, Nowak D, Kawamata N, Akagi T, Kato M, Sanada M, Weiss T, Haferlach C, Dugas M, Ruckert C, Haferlach T, and Koeffler HP. Genomic profiling of adult acute lymphoblastic leukemia by single nucleotide polymorphism oligonu- cleotide microarray and comparison to pediatric acute lymphoblastic leukemia. Haematologica 2010;95(9):1481-1488. doi:10.3324/haematol.2009.011114 Online Supplementary Data ed by PCR of genomic DNA and subsequent direct sequencing of SNP in a region of CNN-LOH in an ALL sample versus the corresponding Design and Methods matched normal sample (Online Supplementary
    [Show full text]
  • A PAX5-OCT4-PRDM1 Developmental Switch Specifies Human Primordial Germ Cells
    A PAX5-OCT4-PRDM1 Developmental Switch Specifies Human Primordial Germ Cells Fang Fang1,2, Benjamin Angulo1,2, Ninuo Xia1,2, Meena Sukhwani3, Zhengyuan Wang4, Charles C Carey5, Aurélien Mazurie5, Jun Cui1,2, Royce Wilkinson5, Blake Wiedenheft5, Naoko Irie6, M. Azim Surani6, Kyle E Orwig3, Renee A Reijo Pera1,2 1Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT 59717, USA 2Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA 3Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, School of Medicine; Magee Women’s Research Institute, Pittsburgh, PA, 15213, USA 4Genomic Medicine Division, Hematology Branch, NHLBI/NIH, MD 20850, USA 5Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA. 6Wellcome Trust Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, UK. Correspondence should be addressed to F.F. (e-mail: [email protected]) 1 Abstract Dysregulation of genetic pathways during human germ cell development leads to infertility. Here, we analyzed bona fide human primordial germ cells (hPGCs) to probe the developmental genetics of human germ cell specification and differentiation. We examined distribution of OCT4 occupancy in hPGCs relative to human embryonic stem cells (hESCs). We demonstrate that development, from pluripotent stem cells to germ cells, is driven by switching partners with OCT4 from SOX2 to PAX5 and PRDM1. Gain- and loss-of-function studies revealed that PAX5 encodes a critical regulator of hPGC development. Moreover, analysis of epistasis indicates that PAX5 acts upstream of OCT4 and PRDM1. The PAX5-OCT4-PRDM1 proteins form a core transcriptional network that activates germline and represses somatic programs during human germ cell differentiation.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Agonists and Knockdown of Estrogen Receptor Β Differentially Affect
    Schüler-Toprak et al. BMC Cancer (2016) 16:951 DOI 10.1186/s12885-016-2973-y RESEARCH ARTICLE Open Access Agonists and knockdown of estrogen receptor β differentially affect invasion of triple-negative breast cancer cells in vitro Susanne Schüler-Toprak1*, Julia Häring1, Elisabeth C. Inwald1, Christoph Moehle2, Olaf Ortmann1 and Oliver Treeck1 Abstract Background: Estrogen receptor β (ERβ) is expressed in the majority of invasive breast cancer cases, irrespective of their subtype, including triple-negative breast cancer (TNBC). Thus, ERβ might be a potential target for therapy of this challenging cancer type. In this in vitro study, we examined the role of ERβ in invasion of two triple-negative breast cancer cell lines. Methods: MDA-MB-231 and HS578T breast cancer cells were treated with the specific ERβ agonists ERB-041, WAY200070, Liquiritigenin and 3β-Adiol. Knockdown of ERβ expression was performed by means of siRNA transfection. Effects on cellular invasion were assessed in vitro by means of a modified Boyden chamber assay. Transcriptome analyses were performed using Affymetrix Human Gene 1.0 ST microarrays. Pathway and gene network analyses were performed by means of Genomatix and Ingenuity Pathway Analysis software. Results: Invasiveness of MBA-MB-231 and HS578T breast cancer cells decreased after treatment with ERβ agonists ERB-041 and WAY200070. Agonists Liquiritigenin and 3β-Adiol only reduced invasion of MDA-MB-231 cells. Knockdown of ERβ expression increased invasiveness of MDA-MB-231 cells about 3-fold. Transcriptome and pathway analyses revealed that ERβ knockdown led to activation of TGFβ signalling and induced expression of a network of genes with functions in extracellular matrix, tumor cell invasion and vitamin D3 metabolism.
    [Show full text]