In Silco Study of MIR-124-1 Transcription Factors in Glioblastoma

Total Page:16

File Type:pdf, Size:1020Kb

In Silco Study of MIR-124-1 Transcription Factors in Glioblastoma Journal of Pharmaceutical Research International 33(12): 71-83, 2021; Article no.JPRI.65792 ISSN: 2456-9119 (Past name: British Journal of Pharmaceutical Research, Past ISSN: 2231-2919, NLM ID: 101631759) In Silco Study of MIR-124-1 Transcription Factors in Glioblastoma Khloud M. Algothmi1* 1Department of Biology, King Abdulaziz University, Jedda, Saudi Arabia. Author’s contribution The sole author designed, analysed, interpreted and prepared the manuscript. Article Information DOI: 10.9734/JPRI/2021/v33i1231257 Editor(s): (1) Dr. Arun Singh, Rohilkhand Medical College and Hospital, India. Reviewers: (1) Dewa Putu Wisnu Wardhana, Udayana University, Indonesia. (2) Xiaodong Li, The Third Affiliated Hospital of Soochow University, P. R. China. Complete Peer review History: http://www.sdiarticle4.com/review-history/65792 Received 20 December 2020 Original Research Article Accepted 23 February 2021 Published 17 March 2021 ABSTRACT MIR-124-1 is a brain-abundant MIRNA, whose expression is important for neuronal tissue division, growth and actions. However, expression of miR-124-1 regulatory mechanisms controlling its actions in neuronal cells in health and diseases still poorly addressed. To understand mechanism for transcriptional and functional regulation of miR-124 in neuronal and glioblastoma cells, this study combined gene expression profiling data and computational transcription factor and microRNA target predictions. The present research focuses on transcription factors and DNA methylation, which are central to miR-124-1 expression regulation. A core promoter sequence of miR-124-1 was predicted to be 500 bp and 100 bp, upstream and downstream of its transcription start sites. Seventy three binding sites of fifty transcription factors in promoter region were found, using MatInspector software. Among these transcription factors, MEIS1, POU3F2, SALL2, ETV1, and MAZ, are known to be brain-enriched transcriptional activators. By using omics data analysis, expression of MAZ, PLAG1 KLF2 as well as a transcriptional repressor ZNF239 showed significant correlation with decline in miR-124-1 expression in glioblastoma cells. Furthermore, a potential CpG island was reported in the promoter, providing another mechanism for transcriptional inhibition of miR-124. As miR-124-1 regulates a number of neuronal physiological and pathological processes, we made an attempt to define its potential targets. A computational prediction of miR- 124-1 targets suggested 265 targets with two or more conserved seed sites. Pathway-based analysis of these target genes revealed a significant enrichment for axonal guidance and cancer signaling pathways. At least ten of these targets, SRGAP1, GNAI3, PLXNA3, SEMA5A, SEMA6A, CEBPA, CBL, RASSF5, MITF, and RPS6KB1, showed expected inverse correlation between their _____________________________________________________________________________________________________ *Corresponding author: E-mail: [email protected]; Algothmi; JPRI, 33(12): 71-83, 2021; Article no.JPRI.65792 expression values and miR-124-1 suppression in glioblastoma cells. Taken together, our data form foundation of subsequent future validation researches for miR-124-1 expression regulation including transcription factors and CpG Island within its promoter as well as functional regulation comprising biological pathways controlled by its target genes. Keywords: Bioinformatics; CpG islands; DNA methylation; glioblastoma; miR-124; neurons; promoter; transcriptional factor. 1. INTRODUCTION Understanding miRNAs' expression manners and identifying miRNA profiles can help as Transcription is regulated by protein factors and biomarkers for the diagnosis and prognosis of small RNA molecules, microRNAs (miRNAs). target treatments had begun to unfold. Notably, These micromolecules are a group of the deletion of only a single miR-124 gene, miR- evolutionarily conserved small non-coding RNAs 124-1, in the Mus musculus led to a significant (ncRNA) with a length of about ~19-23 defect in neuronal developmental processes nucleotides. A single miRNA can regulate such as smaller brain size and axonal distribution hundreds of gene expression at the post- neuronal apoptosis[13]. The molecular translational level leading to translational mechanism behind the exceptionally high level of inhibition of destruction of target mRNAs. The miR-124-1 expression in neuronal cells is still simultaneous regulation of several genes by a unclear. Few studies showed that single miRNA changes biological processes and hypermethylation is associated with cells' physiological activities [1]. microRNA-124 transcriptional silencing of miR-124-1 in human (miR-124) is encoded by three different genes cancers, including colon and hematopoietic (miR-124-1, miR-124-2, and miR-124-3) in all malignancies [14,15,16]. Besides the vertebrates. miR-124-3 resides on chromosome transcriptional regulation, the functional nature of 20, and the other two are on chromosome 8 in miR-124-1 as a negative regulator of its target the human genome. Though primary transcripts genes has been reported and linked to of miR-124 genes (pri-miRNAs) are non- neurological functions and disorders (Review in homologous, mature miR-124 sequences are [8]). However, it is still challenging to understand identical and conserved from worm to human. its functional regulation, given the existence of miR-124 is uniquely expressed in nervous tissue many potential target genes for any miRNA. This and its levels about 100 fold higher than other is the first work to investigate the regulation non-nervous tissues[2,3]. So, the primary role of mechanism of miR-124-1 transcription in miR-124 is to participate in neuronal neuronal cells to the best of our knowledge. The differentiation, development, and biological study addressed two central questions: Whether functions of the brain. Also, miR-124 stimulates brain-enriched transcription factors have binding neuron outgrowth during neurogenesis and sites in the proximal promoter of miR-124-1, and contributes to converting human fibroblasts into whether there are more potential locations of neurons [4,5,6,7]. Furthermore, the association methylation may affect miR-124-1 gene between the aberrant expression of miR-124 and transcription. neurological disorders, as neurodegenerative and neuroimmune diseases, oxidative stress, 2. METHODS stroke, and cancer, was reported previously (Reviewed in [8]). Several studies revealed 2.1 The Sequence of MIR-124 significant down-regulation of miR-124 in various brain tumors like glioblastoma and MIR-124-1 and miR-124-2 are intergenic medulloblastoma [9,10]. miRNAs located on human chromosome 8 ranging from 9903388 to 9903472 bp and from Moreover, its reduction level in 64379149 to 64379257 bp, respectively. MIR- neurodegenerative diseases such as Alzheimer's 124-3 is located on chromosome 20 ranging from disease (AD) and Parkinson's disease (PD) was 63178500 to 63178586 bp. The precursor also reported [11,12]. Therefore, miR-124 had sequences were retrieved from miRBase been proposed as a diagnostic and prognostic (http://www.mirbase.org) and aligned using Basic marker and a therapeutical target for neurological Local Alignment Search Tool (BLAST) disorders as brain tumors, neurodegenerative (https://blast.ncbi.nlm.nih.gov/Blast.cgi). disorders, and stroke (Reviewed in [8]). 72 Algothmi; JPRI, 33(12): 71-83, 2021; Article no.JPRI.65792 2.2 The Sequence of MIR-124 Promoters visualization of the metabolic pathways collected KEGG database. Fisher’s exact test was applied The promoter sequences of miR-124-1 and miR- to discriminate if those genes are enriched in the 124-2 were obtained using the GenBank, KEGG pathway. P-value < 0.05 cut off was Nucleotide database, National Center for chosen to determine the significant level of Biotechnology Information (NCBI). DNA pathways. methylation analysis and potential transcription factor binding sites were predicted using the 3. RESULTS MethPrimer and MatInspector programs. 3.1 Low Conservation between Human 2.3 Meth Primer MIR-124-1 Promoter and MIR-124-2 Promoter Regions CpG islands were predicted across miR-124-1 promoter sequence using the default settings of The precursor sequences of human miR-124-1, the MethPrimer program [17] miR-124-2, and miR-124-3 were extracted from (http://www.urogene.org/methprimer/). The the NCBI database (June 2016), alignments default settings are: reveal high conservation among them (~89%) (Fig.1). However, mature sequences of miR-124- 1. The promoter comprises a DNA sequence 5p and miR-124-3p are 100% conserved of at least 200bp in length between all three precursors. The pri-miRNA 2. CG content in the promoter is over 50% containing the pre-miR-124-2 is known as miR- 3. The ratio of observed to expect the number 124-2 host gene (miR-124-2HG), whereas the of CG dinucleotides is over 0.6 pre-miR-124-1 appears to be derived from annotated long intergenic non-protein coding 2.4 Genomatix MatInspector RNA 599 (LINC00599). As the promoter sequence of miR-124-1 and miR-124-2 are not Genomatix MatInspector prediction search characterized yet, the genomic sequences were (http://www.genomatix.de/) for transcription factor obtained from GenBank in June 2016 (Supp. 1A, binding sites (TFBSs), several parameters B). The host transcript of pre-miR-124-3 has not include the core and matrix similarities, which are been annotated yet; therefore, it is excluded from calculated within MatInspector [18]. The core our work. The sequence comparison of human similarity was set to 1 and >0.95 for
Recommended publications
  • Activated Peripheral-Blood-Derived Mononuclear Cells
    Transcription factor expression in lipopolysaccharide- activated peripheral-blood-derived mononuclear cells Jared C. Roach*†, Kelly D. Smith*‡, Katie L. Strobe*, Stephanie M. Nissen*, Christian D. Haudenschild§, Daixing Zhou§, Thomas J. Vasicek¶, G. A. Heldʈ, Gustavo A. Stolovitzkyʈ, Leroy E. Hood*†, and Alan Aderem* *Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103; ‡Department of Pathology, University of Washington, Seattle, WA 98195; §Illumina, 25861 Industrial Boulevard, Hayward, CA 94545; ¶Medtronic, 710 Medtronic Parkway, Minneapolis, MN 55432; and ʈIBM Computational Biology Center, P.O. Box 218, Yorktown Heights, NY 10598 Contributed by Leroy E. Hood, August 21, 2007 (sent for review January 7, 2007) Transcription factors play a key role in integrating and modulating system. In this model system, we activated peripheral-blood-derived biological information. In this study, we comprehensively measured mononuclear cells, which can be loosely termed ‘‘macrophages,’’ the changing abundances of mRNAs over a time course of activation with lipopolysaccharide (LPS). We focused on the precise mea- of human peripheral-blood-derived mononuclear cells (‘‘macro- surement of mRNA concentrations. There is currently no high- phages’’) with lipopolysaccharide. Global and dynamic analysis of throughput technology that can precisely and sensitively measure all transcription factors in response to a physiological stimulus has yet to mRNAs in a system, although such technologies are likely to be be achieved in a human system, and our efforts significantly available in the near future. To demonstrate the potential utility of advanced this goal. We used multiple global high-throughput tech- such technologies, and to motivate their development and encour- nologies for measuring mRNA levels, including massively parallel age their use, we produced data from a combination of two distinct signature sequencing and GeneChip microarrays.
    [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]
  • Evaluation of Chromatin Accessibility in Prefrontal Cortex of Individuals with Schizophrenia
    ARTICLE DOI: 10.1038/s41467-018-05379-y OPEN Evaluation of chromatin accessibility in prefrontal cortex of individuals with schizophrenia Julien Bryois 1, Melanie E. Garrett2, Lingyun Song3, Alexias Safi3, Paola Giusti-Rodriguez 4, Graham D. Johnson 3, Annie W. Shieh13, Alfonso Buil5, John F. Fullard6, Panos Roussos 6,7,8, Pamela Sklar6, Schahram Akbarian 6, Vahram Haroutunian 6,9, Craig A. Stockmeier 10, Gregory A. Wray3,11, Kevin P. White12, Chunyu Liu13, Timothy E. Reddy 3,14, Allison Ashley-Koch2,15, Patrick F. Sullivan 1,4,16 & Gregory E. Crawford 3,17 1234567890():,; Schizophrenia genome-wide association studies have identified >150 regions of the genome associated with disease risk, yet there is little evidence that coding mutations contribute to this disorder. To explore the mechanism of non-coding regulatory elements in schizophrenia, we performed ATAC-seq on adult prefrontal cortex brain samples from 135 individuals with schizophrenia and 137 controls, and identified 118,152 ATAC-seq peaks. These accessible chromatin regions in the brain are highly enriched for schizophrenia SNP heritability. Accessible chromatin regions that overlap evolutionarily conserved regions exhibit an even higher heritability enrichment, indicating that sequence conservation can further refine functional risk variants. We identify few differences in chromatin accessibility between cases and controls, in contrast to thousands of age-related differential accessible chromatin regions. Altogether, we characterize chromatin accessibility in the human prefrontal cortex, the effect of schizophrenia and age on chromatin accessibility, and provide evidence that our dataset will allow for fine mapping of risk variants. 1 Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-17177 Stockholm, Sweden.
    [Show full text]
  • The E–Id Protein Axis Modulates the Activities of the PI3K–AKT–Mtorc1
    Downloaded from genesdev.cshlp.org on October 6, 2021 - Published by Cold Spring Harbor Laboratory Press The E–Id protein axis modulates the activities of the PI3K–AKT–mTORC1– Hif1a and c-myc/p19Arf pathways to suppress innate variant TFH cell development, thymocyte expansion, and lymphomagenesis Masaki Miyazaki,1,8 Kazuko Miyazaki,1,8 Shuwen Chen,1 Vivek Chandra,1 Keisuke Wagatsuma,2 Yasutoshi Agata,2 Hans-Reimer Rodewald,3 Rintaro Saito,4 Aaron N. Chang,5 Nissi Varki,6 Hiroshi Kawamoto,7 and Cornelis Murre1 1Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA; 2Department of Biochemistry and Molecular Biology, Shiga University of Medical School, Shiga 520-2192, Japan; 3Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany; 4Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA; 5Center for Computational Biology, Institute for Genomic Medicine, University of California at San Diego, La Jolla, California 92093, USA; 6Department of Pathology, University of California at San Diego, La Jolla, California 92093, USA; 7Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan It is now well established that the E and Id protein axis regulates multiple steps in lymphocyte development. However, it remains unknown how E and Id proteins mechanistically enforce and maintain the naı¨ve T-cell fate. Here we show that Id2 and Id3 suppressed the development and expansion of innate variant follicular helper T (TFH) cells. Innate variant TFH cells required major histocompatibility complex (MHC) class I-like signaling and were associated with germinal center B cells.
    [Show full text]
  • Abstract Book
    ISSN 0390-6078 Volume 105 OCTOBER 2020 - S2 XVI Congress of the Italian Society of Experimental Hematology Napoli, Italy, October 15-17, 2020 ABSTRACT BOOK www.haematologica.org XVI Congress of the Italian Society of Experimental Hematology Napoli, Italy, October 15-17, 2020 COMITATO SCIENTIFICO Pellegrino Musto, Presidente Antonio Curti, Vice Presidente Mario Luppi, Past President Francesco Albano Niccolò Bolli Antonella Caivano Roberta La Starza Luca Malcovati Luca Maurillo Stefano Sacchi SEGRETERIA SIES Via De' Poeti, 1/7 - 40124 Bologna Tel. 051 6390906 - Fax 051 4210174 e-mail: [email protected] www.siesonline.it SEGRETERIA ORGANIZZATIVA Studio ER Congressi Via De' Poeti, 1/7 - 40124 Bologna Tel. 051 4210559 - Fax 051 4210174 e-mail: [email protected] www.ercongressi.it ABSTRACT BOOK supplement 2 - October 2020 Table of Contents XVI Congress of the Italian Society of Experimental Hematology Napoli, Italy, October 15-17, 2020 Main Program . 1 Best Abstracts . 20 Oral Communications Session 1. C001-C008 Acute Leukemia 1 . 23 Session 2. C009-C016 Chronic Lymphocytic Leukemia 1 . 28 Session 3. C017-C024 Multiple Myeloma 1 . 32 Session 4. C025-C032 Benign Hematology . 36 Session 5. C033-C040 Multiple Myeloma 2 . 42 Session 6. C041-C048 Acute Leukemia 2 . 45 Session 7. C049-C056 Molecular Hematology . 50 Session 8. C057-C064 Lymphomas. 54 Session 9. C065-C072 Chronic Lymphocytic Leukemia 2 . 57 Session 10. C073-C080 Myelodisplastic Syndromes and Acute Leukemia . 62 Session 11. C081-C088 Myeloproliferative Disorders and Chronic Myeloid Leukemia . 66 Session 12. C089-C096 Stem Cell Transplantation. 71 Posters Session 1. P001 Stem cells and growth factors .
    [Show full text]
  • The YAP/HIF-1Α/Mir-182/EGR2 Axis Is Implicated in Asthma Severity
    Zhou et al. Cell Biosci (2021) 11:84 https://doi.org/10.1186/s13578-021-00560-1 Cell & Bioscience RESEARCH Open Access The YAP/HIF-1α/miR-182/EGR2 axis is implicated in asthma severity through the control of Th17 cell diferentiation Jing Zhou1, Ning Zhang2, Wei Zhang1, Caiju Lu1 and Fei Xu1* Abstract Background: Asthma is a heterogeneous chronic infammatory disease of the airway, involving reversible airfow limitation and airway remodeling. T helper 17 (Th17) cells play an important role in the pathogenesis of allergic asthma. However, there is limited understanding of the signaling pathways controlling Th17 cell diferentiation in asthma. The aim of this study was to investigate if the Yes-associated protein (YAP)/hypoxia inducible factor-1α (HIF-1α)/microRNA-182 (miR-182)/early growth response 2 (EGR2) axis is involved in mediating Th17 cell diferentia- tion and disease severity in asthma. Methods: The study included 29 pediatric patients with asthma, 22 healthy volunteers, ovalbumin-induced murine asthma models, and mouse naive CD4+ T cells. The subpopulation of Th17 cells was examined by fow cytometry. The levels of interleukin-17A were determined by enzyme linked immunosorbent assay. Chromatin immunoprecipitation- quantitative polymerase chain reaction assays and dual-luciferase reporter gene assays were performed to examine interactions between HIF-1α and miR-182, and between miR-182 and EGR2. Results: YAP, HIF-1α, and miR-182 were upregulated but EGR2 was downregulated in human and mouse peripheral blood mononuclear cells from the asthma group. Abundant expression of YAP and HIF-1α promoted miR-182 expres- sion and then inhibited EGR2, a target of miR-182, thus enhancing Th17 diferentiation and deteriorating asthma and lipid metabolism dysfunction.
    [Show full text]
  • Whole Exome Sequencing in Families at High Risk for Hodgkin Lymphoma: Identification of a Predisposing Mutation in the KDR Gene
    Hodgkin Lymphoma SUPPLEMENTARY APPENDIX Whole exome sequencing in families at high risk for Hodgkin lymphoma: identification of a predisposing mutation in the KDR gene Melissa Rotunno, 1 Mary L. McMaster, 1 Joseph Boland, 2 Sara Bass, 2 Xijun Zhang, 2 Laurie Burdett, 2 Belynda Hicks, 2 Sarangan Ravichandran, 3 Brian T. Luke, 3 Meredith Yeager, 2 Laura Fontaine, 4 Paula L. Hyland, 1 Alisa M. Goldstein, 1 NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, Stephen J. Chanock, 5 Neil E. Caporaso, 1 Margaret A. Tucker, 6 and Lynn R. Goldin 1 1Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 2Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 3Ad - vanced Biomedical Computing Center, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Frederick, MD; 4Westat, Inc., Rockville MD; 5Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; and 6Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA ©2016 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2015.135475 Received: August 19, 2015. Accepted: January 7, 2016. Pre-published: June 13, 2016. Correspondence: [email protected] Supplemental Author Information: NCI DCEG Cancer Sequencing Working Group: Mark H. Greene, Allan Hildesheim, Nan Hu, Maria Theresa Landi, Jennifer Loud, Phuong Mai, Lisa Mirabello, Lindsay Morton, Dilys Parry, Anand Pathak, Douglas R. Stewart, Philip R. Taylor, Geoffrey S. Tobias, Xiaohong R. Yang, Guoqin Yu NCI DCEG Cancer Genomics Research Laboratory: Salma Chowdhury, Michael Cullen, Casey Dagnall, Herbert Higson, Amy A.
    [Show full text]
  • The Transcription Factor EGR2 Is the Molecular Linchpin Connecting STAT6 Activation to the Late, Stable Epigenomic Program of Alternative Macrophage Polarization
    Downloaded from genesdev.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press The transcription factor EGR2 is the molecular linchpin connecting STAT6 activation to the late, stable epigenomic program of alternative macrophage polarization Bence Daniel,1,2,3,11,12 Zsolt Czimmerer,4,12 Laszlo Halasz,1,2,3,12 Pal Boto,4 Zsuzsanna Kolostyak,4 Szilard Poliska,4 Wilhelm K. Berger,1,2,3 Petros Tzerpos,4 Gergely Nagy,4 Attila Horvath,4 György Hajas,5 Timea Cseh,4 Aniko Nagy,4 Sascha Sauer,6,7,8,9 Jean Francois-Deleuze,10 Istvan Szatmari,4 Attila Bacsi,5 and Laszlo Nagy1,2,3,4 1Department of Medicine, 2Department of Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida 33701, USA; 3Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida 33701, USA; 4Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; 5Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; 6Otto Warburg Laboratory, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany; 7CU Systems Medicine, University of Würzburg, Würzburg 97070, Germany; 8Berlin Institute for Medical Systems Biology, 9Berlin Institute of Health, Max-Delbrück- Center for Molecular Medicine, Berlin 13125, Germany; 10Centre National de Génotypage, Institut de Génomique, Commissariat à l’Énergie Atomique, Evry 91000, France Macrophages polarize into functionally distinct subtypes while responding to microenvironmental cues. The identity of proximal transcription factors (TFs) downstream from the polarization signals are known, but their ac- tivity is typically transient, failing to explain the long-term, stable epigenomic programs developed.
    [Show full text]
  • Egr2 and 3 Control Adaptive Immune Responses by Temporally Uncoupling Expansion from T Cell Differentiation
    Published Online: 9 May, 2017 | Supp Info: http://doi.org/10.1084/jem.20160553 Downloaded from jem.rupress.org on October 9, 2018 Article Egr2 and 3 control adaptive immune responses by temporally uncoupling expansion from T cell differentiation Tizong Miao,1* Alistair L.J. Symonds,1* Randeep Singh,1,2 Janine D. Symonds,3 Ane Ogbe,2 Becky Omodho,2 Bo Zhu,1,4 Suling Li,2 and Ping Wang1 1The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, England, UK 2Bioscience, Brunel University, Uxbridge UB8 3PH, England, UK 3Centre for Mathematics and Physics in the Life Sciences and Experimental Biology (CoMPLEX), University College London, London WC1E 6BT, England, UK 4Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People’s Republic of China Egr2 and 3 are important for maintaining immune homeostasis. Here we define a fundamental function of Egr2 and 3 oper- ating as a checkpoint that controls the transition between clonal expansion and differentiation of effector T cells. Egr2 and 3 deficiency resulted in defective clonal expansion but hyperactivation and excessive differentiation of T cells in response to viral infection. Conversely, sustained Egr2 expression enhanced expansion but severely impaired effector differentiation. Egr2 bound to and controlled the expression of genes regulating proliferation (Myc and Myb) and differentiation repressors (Bcl6, Id3), while repressing transcription factors required for effector function (Zeb2, RORa, RORc, and Bhlhe40). Egr2 and 3 ex- pression in T cells was regulated reciprocally by antigen and IFNγ, providing a mechanism for adjusting proliferation and dif- ferentiation of individual T cells.
    [Show full text]
  • A Dissertation Entitled the Androgen Receptor
    A Dissertation entitled The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit Submitted to the Graduate Faculty as partial fulfillment of the requirements for the PhD Degree in Biomedical science Dr. Manohar Ratnam, Committee Chair Dr. Lirim Shemshedini, Committee Member Dr. Robert Trumbly, Committee Member Dr. Edwin Sanchez, Committee Member Dr. Beata Lecka -Czernik, Committee Member Dr. Patricia R. Komuniecki, Dean College of Graduate Studies The University of Toledo August 2011 Copyright 2011, Mesfin Gonit This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit As partial fulfillment of the requirements for the PhD Degree in Biomedical science The University of Toledo August 2011 Prostate cancer depends on the androgen receptor (AR) for growth and survival even in the absence of androgen. In the classical models of gene activation by AR, ligand activated AR signals through binding to the androgen response elements (AREs) in the target gene promoter/enhancer. In the present study the role of AREs in the androgen- independent transcriptional signaling was investigated using LP50 cells, derived from parental LNCaP cells through extended passage in vitro. LP50 cells reflected the signature gene overexpression profile of advanced clinical prostate tumors. The growth of LP50 cells was profoundly dependent on nuclear localized AR but was independent of androgen. Nevertheless, in these cells AR was unable to bind to AREs in the absence of androgen.
    [Show full text]
  • Epigenetic Services Citations
    Active Motif Epigenetic Services Publications The papers below contain data generated by Active Motif’s Epigenetic Services team. To learn more about our services, please give us a call or visit us at www.activemotif.com/services. Technique Target Journal Year Reference Justin C. Boucher et al. CD28 Costimulatory Domain- ATAC-Seq, Cancer Immunol. Targeted Mutations Enhance Chimeric Antigen Receptor — 2021 RNA-Seq Res. T-cell Function. Cancer Immunol. Res. doi: 10.1158/2326- 6066.CIR-20-0253. Satvik Mareedu et al. Sarcolipin haploinsufficiency Am. J. Physiol. prevents dystrophic cardiomyopathy in mdx mice. RNA-Seq — Heart Circ. 2021 Am J Physiol Heart Circ Physiol. doi: 10.1152/ Physiol. ajpheart.00601.2020. Gabi Schutzius et al. BET bromodomain inhibitors regulate Nature Chemical ChIP-Seq BRD4 2021 keratinocyte plasticity. Nat. Chem. Biol. doi: 10.1038/ Biology s41589-020-00716-z. Siyun Wang et al. cMET promotes metastasis and ChIP-qPCR FOXO3 J. Cell Physiol. 2021 epithelial-mesenchymal transition in colorectal carcinoma by repressing RKIP. J. Cell Physiol. doi: 10.1002/jcp.30142. Sonia Iyer et al. Genetically Defined Syngeneic Mouse Models of Ovarian Cancer as Tools for the Discovery of ATAC-Seq — Cancer Discovery 2021 Combination Immunotherapy. Cancer Discov. doi: doi: 10.1158/2159-8290 Vinod Krishna et al. Integration of the Transcriptome and Genome-Wide Landscape of BRD2 and BRD4 Binding BRD2, BRD4, RNA Motifs Identifies Key Superenhancer Genes and Reveals ChIP-Seq J. Immunol. 2021 Pol II the Mechanism of Bet Inhibitor Action in Rheumatoid Arthritis Synovial Fibroblasts. J. Immunol. doi: doi: 10.4049/ jimmunol.2000286. Daniel Haag et al.
    [Show full text]
  • Lhx1 Maintains Synchrony Among Circadian Oscillator Neurons
    RESEARCH ARTICLE elifesciences.org Lhx1 maintains synchrony among circadian oscillator neurons of the SCN Megumi Hatori1*†‡, Shubhroz Gill1†, Ludovic S Mure1, Martyn Goulding2, Dennis D M O'Leary2, Satchidananda Panda1* 1Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, United States; 2Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, United States Abstract The robustness and limited plasticity of the master circadian clock in the suprachiasmatic nucleus (SCN) is attributed to strong intercellular communication among its constituent neurons. However, factors that specify this characteristic feature of the SCN are unknown. Here, we identified Lhx1 as a regulator of SCN coupling. A phase-shifting light pulse causes acute reduction in Lhx1 expression and of its target genes that participate in SCN coupling. Mice lacking Lhx1 in the SCN have intact circadian oscillators, but reduced levels of coupling factors. Consequently, the mice rapidly phase shift under a jet lag paradigm and their behavior rhythms gradually deteriorate under constant condition. Ex vivo recordings of the SCN from these mice showed rapid desynchronization of unit oscillators. Therefore, by regulating expression of genes mediating intercellular communication, Lhx1 imparts synchrony among SCN neurons and ensures consolidated rhythms of activity and rest that is resistant to photic noise. *For correspondence: mhatori@ DOI: 10.7554/eLife.03357.001 a6.keio.jp (MH); [email protected] (SP) †These authors contributed equally to this work Introduction Present address: ‡School of Circadian clocks generate ∼24 hr rhythms in behavior and physiology which allow an organism to Medicine, Keio University, Tokyo, anticipate and adjust to environmental changes accompanying the earth's day/night cycle.
    [Show full text]