HMGA2 As a Critical Regulator in Cancer Development

Total Page:16

File Type:pdf, Size:1020Kb

HMGA2 As a Critical Regulator in Cancer Development G C A T T A C G G C A T genes Review HMGA2 as a Critical Regulator in Cancer Development Behzad Mansoori 1,2,3,4 , Ali Mohammadi 1,3, Henrik J. Ditzel 3,5,6 , Pascal H. G. Duijf 7,8 , Vahid Khaze 1,3, Morten F. Gjerstorff 3,5,6,* and Behzad Baradaran 1,* 1 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; [email protected] (B.M.); [email protected] (A.M.); [email protected] (V.K.) 2 Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran 3 Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark; [email protected] 4 Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran 5 Department of Oncology, Odense University Hospital, 5000 Odense, Denmark 6 Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark 7 Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4102, Australia; [email protected] 8 University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD 4102, Australia * Correspondence: [email protected] (M.F.G.); [email protected] (B.B.) Abstract: The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human ma- lignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 Citation: Mansoori, B.; Mohammadi, influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition A.; Ditzel, H.J.; Duijf, P.H.G.; Khaze, by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 V.; Gjerstorff, M.F.; Baradaran, B. pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells HMGA2 as a Critical Regulator in resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in Cancer Development. Genes 2021, 12, different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, 269. https://doi.org/10.3390/ and therapeutic purposes. genes12020269 Keywords: β Academic Editor: Félix Machín HMGA2; cancer; cell cycle; apoptosis; DNA repair; MAPK; TGF ; RKIP; miRNA Received: 7 January 2021 Accepted: 8 February 2021 Published: 13 February 2021 1. Introduction High mobility group (HMG) proteins are nonhistone, chromatin-associated molecules Publisher’s Note: MDPI stays neutral involved in maintenance and functional regulation of DNA, including the processes of repli- with regard to jurisdictional claims in cation, recombination, transcription, and DNA repair [1]. The proteins of this superfamily published maps and institutional affil- bend DNA upon binding to the minor groove and thereby contribute to transcriptional regu- iations. lation and maintenance of the chromatin structure [2]. HMG proteins have extremely broad DNA sequence specificity, therefore supplying an exceptional mechanism for protein–DNA interactions [3]. The biological activities of HMG proteins are regulated through epigenetic mechanisms, which affect their interactions with DNA and other transcription factors. Copyright: © 2021 by the authors. Consequently, this affects signaling pathways by recruitment of different transcription Licensee MDPI, Basel, Switzerland. factors to the target gene promotors, which cause a specific phenotypic outcome. This article is an open access article The HMG superfamily is grouped into three main families, including HMGA, HMGB distributed under the terms and and HMGN. The HMGA gene family consists of two functional members, high mobility conditions of the Creative Commons group A1 (HMGA1or HMGI/Y) and high mobility group A2 (HMGA2 or HMGI-C). Attribution (CC BY) license (https:// HMGA proteins bind to AT-rich DNA regions that exist in short stretches in the minor creativecommons.org/licenses/by/ groove of the DNA (Figure1)[ 4]. In addition to DNA, they also interact with different 4.0/). Genes 2021, 12, 269. https://doi.org/10.3390/genes12020269 https://www.mdpi.com/journal/genes Genes 2021, 12, x FOR PEER REVIEW 2 of 23 C). HMGA proteins bind to AT-rich DNA regions that exist in short stretches in the minor groove of the DNA (Figure 1) [4]. In addition to DNA, they also interact with different proteins, such as E2 promoter-binding factor 1 (E2F1) and retinoblastoma protein (Rb). Increasing evidence indicates that HMGA proteins do not directly regulate transcriptional activity, but rather control gene expression by changing chromatin structure and organ- Genes 2021, 12, 269 2 of 23 izing several transcription factors on a so-called “enhanceosome” [5,6]. When HMGA2 binds to AT-rich hooks of DNA, the conformation changes from disordered to ordered, and in turn this introduces bending, unwinding, straightening, and looping of DNA [7]. Theproteins, acidic such tail of as the E2 HMGA2 promoter-binding protein is resp factoronsible 1 (E2F1) for protein–protein and retinoblastoma interactions. protein Thus, (Rb). thisIncreasing domain evidence mediates indicates binding that of HMGA2 HMGA proteins to other do DNA not directlybinding regulateproteins, transcriptional such as tran- scriptionactivity, but factors rather [8]. control gene expression by changing chromatin structure and organizing severalRecent transcription studies suggest factors a on possible a so-called link “enhanceosome”between HMG proteins [5,6]. When and cancer HMGA2 develop- binds mentto AT-rich [9,10]. hooks HMGA2 of DNA, is important the conformation during ea changesrly embryogenesis from disordered and is to highly ordered, expressed and in duringturn this oncogenesis. introduces bending,Studies have unwinding, shown straightening,that HMGA2 andincreases looping cancer of DNA progression [7]. The throughacidic tail the of activation the HMGA2 of several protein pathways. is responsible In this for protein–proteinliterature review, interactions. we highlight Thus, the thison- cogenicdomain role mediates of HMGA2 binding in of tumor HMGA2 development to other DNA and bindingprogression proteins, and discuss such as transcriptionthe probable signalingfactors [8 ].pathways affected by this architectural protein. Figure 1. Schematic of the highFigure mobility 1. Schematic group of protein the high 2 (HMGA2)mobility group gene protein and protein. 2 (HMGA2) Located gene on and chromosome protein. Located 12, the on HMGA2 gene encodes a smallchromosome protein, which12, thecan HMGA2 bind togene AT-rich encodes sites a insmall the pr minorotein, groove which ofcan DNA bind andto AT-rich subsequently sites in the regulate transcription by inducingminor groove conformational of DNA and changes subsequently to the DNA. regulate transcription by inducing conformational changes to the DNA. Recent studies suggest a possible link between HMG proteins and cancer develop- 2.ment HMGA2 [9,10]. Structure HMGA2 isand important Function during early embryogenesis and is highly expressed dur- ing oncogenesis.The HMGA2 Studies gene contains have shown five exons that HMGA2 distributed increases over a cancergenomic progression area of more through than 140the activationkb located ofat severalhuman pathways.chromosome In this12q13-15 literature (Figure review, 1). The we first highlight three theexons oncogenic encode therole AT-binding of HMGA2 indomain tumor site, development while exon and 4 en progressioncodes a protein and discuss linker theand probable exon 5 the signaling acidic domainpathways which affected does by not this activate architectural transcription protein. (Figure 1). HMGA2 is a rather small protein of less than 12 kDa, comprising 108 amino acids. Akin to the other protein family mem- bers,2. HMGA2 HMGA2 Structure regulates and gene Function expression by binding to the DNA minor groove and chro- matinThe management HMGA2 gene [11]. contains five exons distributed over a genomic area of more than 140 kb located at human chromosome 12q13-15 (Figure1). The first three exons encode the AT-binding domain site, while exon 4 encodes a protein linker and exon 5 the acidic domain which does not activate transcription (Figure1). HMGA2 is a rather small protein of less than 12 kDa, comprising 108 amino acids. Akin to the other protein family members, HMGA2 regulates gene expression by binding to the DNA minor groove and chromatin management [11]. The HMGA2 AT-binding domains are each comprised of nine amino acids, including the invariable repeat Arg-Gly-Arg-Pro [12]. During transcription, these domains can bind Genes 2021, 12, x FOR PEER REVIEW 3 of 23 Genes 2021, 12, 269 3 of 23 The HMGA2 AT-binding domains are each comprised of nine amino acids, including the invariable repeat Arg-Gly-Arg-Pro [12]. During transcription, these domains can bind toto B-shaped B-shaped DNA DNA and and convert convert it itfrom from disordered disordered to toordered ordered conformations. conformations. Based Based on the on numberthe number and andlocation location of the of theAT-rich AT-rich binding binding sites,
Recommended publications
  • RNA-Binding Protein Hnrnpll Regulates Mrna Splicing and Stability During B-Cell to Plasma-Cell Differentiation
    RNA-binding protein hnRNPLL regulates mRNA splicing and stability during B-cell to plasma-cell differentiation Xing Changa,b, Bin Lic, and Anjana Raoa,b,d,e,1 Divisions of aSignaling and Gene Expression and cVaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; bSanford Consortium for Regenerative Medicine, La Jolla, CA 92037; and dDepartment of Pharmacology and eMoores Cancer Center, University of California at San Diego, La Jolla, CA 92093 Contributed by Anjana Rao, December 2, 2014 (sent for review July 20, 2014) Posttranscriptional regulation is a major mechanism to rewire the RBP-binding sites, thus validating the specificity of RBP binding transcriptomes during differentiation. Heterogeneous nuclear to coprecipitating RNAs and mapping RBP-binding sites on the RNA-binding protein LL (hnRNPLL) is specifically induced in terminally validated RNAs at close to single-nucleotide resolution (8). differentiated lymphocytes, including effector T cells and plasma Heterogeneous nuclear RNA-binding proteins (hnRNPs) is cells. To study the molecular functions of hnRNPLL at a genome- the term applied to a collection of unrelated nuclear RBPs. wide level, we identified hnRNPLL RNA targets and binding sites in hnRNPLL was identified through a targeted lentiviral shRNA plasma cells through integrated Photoactivatable-Ribonucleoside- screen for regulators of CD45RA to CD45RO switching during Enhanced Cross-Linking and Immunoprecipitation (PAR-CLIP) and memory T-cell development (9) and independently through RNA sequencing. hnRNPLL preferentially recognizes CA dinucleo- two separate screens performed by different groups for exclusion tide-containing sequences in introns and 3′ untranslated regions of CD45 exon 4 in a minigene context (10) and for altered CD44 (UTRs), promotes exon inclusion or exclusion in a context-dependent and CD45R expression on T cells in N-ethyl-N-nitrosourea manner, and stabilizes mRNA when associated with 3′ UTRs.
    [Show full text]
  • TGF-Β1 Signaling Targets Metastasis-Associated Protein 1, a New Effector in Epithelial Cells
    Oncogene (2011) 30, 2230–2241 & 2011 Macmillan Publishers Limited All rights reserved 0950-9232/11 www.nature.com/onc ORIGINAL ARTICLE TGF-b1 signaling targets metastasis-associated protein 1, a new effector in epithelial cells SB Pakala1, K Singh1,3, SDN Reddy1, K Ohshiro1, D-Q Li1, L Mishra2 and R Kumar1 1Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, DC, USA and 2Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA In spite of a large number of transforming growth factor b1 gene chromatin in response to upstream signals. The (TGF-b1)-regulated genes, the nature of its targets with TGF-b1-signaling is largely mediated by Smad proteins roles in transformation continues to be poorly understood. (Massague et al., 2005) where Smad2 and Smad3 are Here, we discovered that TGF-b1 stimulates transcription phosphorylated by TGF-b1-receptors and associate with of metastasis-associated protein 1 (MTA1), a dual master the common mediator Smad4, which translocates to the coregulator, in epithelial cells, and that MTA1 status is a nucleus to participate in the expression of TGF-b1-target determinant of TGF-b1-induced epithelial-to-mesenchymal genes (Deckers et al., 2006). Previous studies have shown transition (EMT) phenotypes. In addition, we found that that CUTL1, also known as CDP (CCAAT displacement MTA1/polymerase II/activator protein-1 (AP-1) co-activator protein), a target of TGF-b1, is needed for its short-term complex interacts with the FosB-gene chromatin and stimu- effects of TGF-b1 on cell motility involving Smad4- lates its transcription, and FosB in turn, utilizes FosB/histone dependent pathway (Michl et al.,2005).
    [Show full text]
  • Functional Compensation Among HMGN Variants Modulates the Dnase I Hypersensitive Sites at Enhancers
    Downloaded from genome.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press Research Functional compensation among HMGN variants modulates the DNase I hypersensitive sites at enhancers Tao Deng,1,12 Z. Iris Zhu,2,12 Shaofei Zhang,1 Yuri Postnikov,1 Di Huang,2 Marion Horsch,3 Takashi Furusawa,1 Johannes Beckers,3,4,5 Jan Rozman,3,5 Martin Klingenspor,6,7 Oana Amarie,3,8 Jochen Graw,3,8 Birgit Rathkolb,3,5,9 Eckhard Wolf,9 Thure Adler,3 Dirk H. Busch,10 Valérie Gailus-Durner,3 Helmut Fuchs,3 Martin Hrabeˇ de Angelis,3,4,5 Arjan van der Velde,2,13 Lino Tessarollo,11 Ivan Ovcherenko,2 David Landsman,2 and Michael Bustin1 1Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA; 2Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, Bethesda, Maryland 20892, USA; 3German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; 4Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany; 5German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; 6Molecular Nutritional Medicine, Technische Universität München, 85350 Freising, Germany; 7Center for Nutrition and Food Sciences, Technische Universität München, 85350 Freising, Germany; 8Institute of Developmental Genetics (IDG), 85764
    [Show full text]
  • Multiple Facets of Jund Gene Expression Are Atypical Among AP-1 Family Members
    Oncogene (2008) 27, 4757–4767 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $30.00 www.nature.com/onc REVIEW Multiple facets of junD gene expression are atypical among AP-1 family members JM Hernandez1, DH Floyd2, KN Weilbaecher2, PL Green1,3 and K Boris-Lawrie1,3 1Department of Veterinary Biosciences and Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA and 2Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St Louis, MO, USA and 3Department of Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA JunD is a versatile AP-1 transcription factor that can 2003; Milde-Langosch, 2005). The AP-1 component activate or repress a diverse collection of target genes. proteins are characterized structurally by their leucine- Precise control of junD expression and JunD protein– zipper dimerization motif and basic DNA-binding protein interactions modulate tumor angiogenesis, cellular domain. They can either activate or repress transcription differentiation, proliferation and apoptosis. Molecular and this versatile functional activity is dependent on the and clinical knowledge of two decades has revealed specific components of the dimeric complex and the that precise JunD activity is elaborated by interrelated cellular environment (Eferl and Wagner, 2003; Hess layers of constitutive transcriptional control, complex et al., 2004). AP-1 figures prominently in transcriptional post-transcriptional regulation and a collection of regulation of early response genes (reviewed by Jochum post-translational modifications and protein–protein et al., 2001; Mechta-Grigoriou et al., 2001; Eferl and interactions. The stakes are high, as inappropriate JunD Wagner, 2003).
    [Show full text]
  • Mrna-Lncrna Co-Expression Network Analysis Reveals the Role of Lncrnas in Immune Dysfunction During Severe SARS-Cov-2 Infection
    viruses Article mRNA-lncRNA Co-Expression Network Analysis Reveals the Role of lncRNAs in Immune Dysfunction during Severe SARS-CoV-2 Infection Sumit Mukherjee 1 , Bodhisattwa Banerjee 2 , David Karasik 2 and Milana Frenkel-Morgenstern 1,* 1 Cancer Genomics and BioComputing of Complex Diseases Lab, Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; [email protected] 2 Musculoskeletal Genetics Laboratory, Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; [email protected] (B.B.); [email protected] (D.K.) * Correspondence: [email protected]; Tel.: +972-72-264-4901 Abstract: The recently emerged SARS-CoV-2 virus is responsible for the ongoing COVID-19 pan- demic that has rapidly developed into a global public health threat. Patients severely affected with COVID-19 present distinct clinical features, including acute respiratory disorder, neutrophilia, cy- tokine storm, and sepsis. In addition, multiple pro-inflammatory cytokines are found in the plasma of such patients. Transcriptome sequencing of different specimens obtained from patients suffering from severe episodes of COVID-19 shows dynamics in terms of their immune responses. However, those host factors required for SARS-CoV-2 propagation and the underlying molecular mechanisms responsible for dysfunctional immune responses during COVID-19 infection remain elusive. In the present study, we analyzed the mRNA-long non-coding RNA (lncRNA) co-expression network derived from publicly available SARS-CoV-2-infected transcriptome data of human lung epithelial Citation: Mukherjee, S.; Banerjee, B.; cell lines and bronchoalveolar lavage fluid (BALF) from COVID-19 patients. Through co-expression Karasik, D.; Frenkel-Morgenstern, M.
    [Show full text]
  • Human Stem Cells from Single Blastomeres Reveal Pathways of Embryonic Or Trophoblast Fate Specification Tamara Zdravkovic1,2,3,4,5,‡, Kristopher L
    © 2015. Published by The Company of Biologists Ltd | Development (2015) 142, 4010-4025 doi:10.1242/dev.122846 STEM CELLS AND REGENERATION RESEARCH ARTICLE Human stem cells from single blastomeres reveal pathways of embryonic or trophoblast fate specification Tamara Zdravkovic1,2,3,4,5,‡, Kristopher L. Nazor6,‡, Nicholas Larocque1,2,3,4,5, Matthew Gormley1,2,3,4,5, Matthew Donne1,2,3,7, Nathan Hunkapillar1,2,3,4,5, Gnanaratnam Giritharan8, Harold S. Bernstein4,9, Grace Wei4,10, Matthias Hebrok10, Xianmin Zeng11, Olga Genbacev1,2,3,4,5, Aras Mattis4,12, Michael T. McMaster4,5,13, Ana Krtolica8,*, Diana Valbuena14, Carlos Simón14, Louise C. Laurent6,15, Jeanne F. Loring6 and Susan J. Fisher1,2,3,4,5,7,§ ABSTRACT INTRODUCTION For many reasons, relatively little is known about human Mechanisms of initial cell fate decisions differ among species. To gain preimplantation development. The small number of cells makes insights into lineage allocation in humans, we derived ten human embryos of any species difficult to study. In humans, the technical embryonic stem cell lines (designated UCSFB1-10) from single difficulties are compounded by other challenges. Genetic variation blastomeres of four 8-cell embryos and one 12-cell embryo from a among individuals could contribute to developmental differences, a single couple. Compared with numerous conventional lines from well-appreciated phenomenon in the mouse (Dackor et al., 2009), blastocysts, they had unique gene expression and DNA methylation which is difficult to assess in humans owing to the limited patterns that were, in part, indicative of trophoblast competence. At a availability of embryos that are donated for research.
    [Show full text]
  • The Activator Protein-1 Transcription Factor in Respiratory Epithelium Carcinogenesis
    Subject Review The Activator Protein-1 Transcription Factor in Respiratory Epithelium Carcinogenesis Michalis V. Karamouzis,1 Panagiotis A. Konstantinopoulos,1,2 and Athanasios G. Papavassiliou1 1Department of Biological Chemistry, Medical School, University of Athens, Athens, Greece and 2Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts Abstract Much of the current anticancer research effort is focused on Respiratory epithelium cancers are the leading cause cell-surface receptors and their cognate upstream molecules of cancer-related death worldwide. The multistep natural because they provide the easiest route for drugs to affect history of carcinogenesis can be considered as a cellular behavior, whereas agents acting at the level of gradual accumulation of genetic and epigenetic transcription need to invade the nucleus. However, the aberrations, resulting in the deregulation of cellular therapeutic effect of surface receptor manipulation might be homeostasis. Growing evidence suggests that cross- considered less than specific because their actions are talk between membrane and nuclear receptor signaling modulated by complex interacting downstream signal trans- pathways along with the activator protein-1 (AP-1) duction pathways. A pivotal transcription factor during cascade and its cofactor network represent a pivotal respiratory epithelium carcinogenesis is activator protein-1 molecular circuitry participating directly or indirectly in (AP-1). AP-1–regulated genes include important modulators of respiratory epithelium carcinogenesis. The crucial role invasion and metastasis, proliferation, differentiation, and of AP-1 transcription factor renders it an appealing survival as well as genes associated with hypoxia and target of future nuclear-directed anticancer therapeutic angiogenesis (7). Nuclear-directed therapeutic strategies might and chemoprevention approaches.
    [Show full text]
  • High-Mobility Group A1 Proteins May Be Involved in Estrogen Receptor Status of Breast Cancer
    3786 Editorial Commentary High-mobility group A1 proteins may be involved in estrogen receptor status of breast cancer Yoshihiro Harada, Kenji Ohe Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Jonan-ku, Fukuoka, Japan Correspondence to: Kenji Ohe. Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Building 17, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-180, Japan. Email: [email protected]. Provenance and Peer Review: This article was commissioned by the editorial office, Translational Cancer Research. The article did not undergo external peer review. Comment on: Gorbounov M, Carleton NM, Asch-Kendrick RJ, et al. High mobility group A1 (HMGA1) protein and gene expression correlate with ER-negativity and poor outcomes in breast cancer. Breast Cancer Res Treat 2020;179:25-35. Submitted Apr 27, 2020. Accepted for publication May 13, 2020. doi: 10.21037/tcr-20-1921 View this article at: http://dx.doi.org/10.21037/tcr-20-1921 The three types of breast cancer HMGA1 proteins: old and new proteins in breast cancer Breast cancer is one of the most common of cancers in woman. About 316,700 new cases were diagnosed as breast HMGA1 (previously called HMGI/Y) is a member of the cancer in US woman in 2019 and 41,760 were predicted to high-mobility group (HMG) proteins that were found from die from it (1). Breast cancer has a characteristic of therapy- their high mobility characteristics during polyacrylamide targeting receptors: hormone receptors (HR) that are electrophoresis of non-histone chromatin-associated estrogen receptor [ER: human ERα protein (NCBI protein proteins (3).
    [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]
  • Long Non-Coding RNA EGOT Promotes the Malignant Phenotypes of Hepatocellular Carcinoma Cells and Increases the Expression of HMGA2 Via Down-Regulating Mir-33A-5P
    OncoTargets and Therapy Dovepress open access to scientific and medical research Open Access Full Text Article ORIGINAL RESEARCH Long Non-Coding RNA EGOT Promotes the Malignant Phenotypes of Hepatocellular Carcinoma Cells and Increases the Expression of HMGA2 via Down-Regulating miR-33a-5p This article was published in the following Dove Press journal: OncoTargets and Therapy Shimin Wu 1 Background: Chronic hepatitis C virus (HCV) infection is an important risk factor for hepato- Hongwu Ai2 cellular carcinoma (HCC). EGOT is a long non-coding RNA (lncRNA) induced after HCV Kehui Zhang3,4 infection that increases viral replication by antagonizing the antiviral response. Interestingly, Hao Yun3,4 EGOTalso acts as a crucial regulator in multiple cancers. However, its role in HCC remains unclear. Fei Xie3,4 Methods: Real-time PCR (RT-PCR) was used to detect the expression of EGOT in HCC samples and cell lines. CCK-8 assay and colony formation assay were performed to evaluate 1 Center for Clinical Laboratory, General the effect of EGOT on proliferation. Scratch healing assay and transwell assay were used to Hospital of the Yangtze River Shipping, Wuhan Brain Hospital, Wuhan 430030, detect the changes of migration and invasion. Flow cytometry was used to detect the effect of People’s Republic of China; 2Center for EGOT on apoptosis. Interaction between EGOT and miR-33a-5p was determined by bioin- Clinical Laboratory, Wuhan Kangjian formatics analysis, RT-PCR, and dual-luciferase reporter assay. Western blot was used to Maternal and Infant Hospital, Wuhan 430050, People’s Republic of China; confirm that high mobility group protein A2 (HMGA2) could be modulated by EGOT.
    [Show full text]
  • An Ontogenetic Switch Drives the Positive and Negative Selection of B Cells
    An ontogenetic switch drives the positive and negative selection of B cells Xijin Xua, Mukta Deobagkar-Lelea, Katherine R. Bulla, Tanya L. Crockforda, Adam J. Meadb, Adam P. Cribbsc, David Simsc, Consuelo Anzilottia, and Richard J. Cornalla,1 aMedical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom; bMedical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom; and cMedical Research Council, Weatherall Institute of Molecular Medicine, Centre for Computational Biology, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, United Kingdom Edited by Michael Reth, University of Freiburg, Freiburg, Germany, and approved January 6, 2020 (received for review September 3, 2019) + Developing B cells can be positively or negatively selected by self- BM HSCs increased CD5 B-1a B cell development (15), while antigens, but the mechanisms that determine these outcomes are expression of let-7b in FL pro-B cells blocked the development of incompletely understood. Here, we show that a B cell intrinsic B-1 B cells (17). These findings support the notion of hard-wired switch between positive and negative selection during ontogeny differences during ontogeny, but possibly downstream of the HSC is determined by a change from Lin28b to let-7 gene expression. commitment stage. Ectopic expression of a Lin28b transgene in murine B cells restored Several lines of evidence also suggest that B-1 B cells can un- the positive selection of autoreactive B-1 B cells by self-antigen in dergo positive selection, which is linked to their B cell receptor adult bone marrow.
    [Show full text]