Dynamics of Mrna and Microrna Expression in the Estrogen Response of Breast Cancer Cells

Total Page:16

File Type:pdf, Size:1020Kb

Dynamics of Mrna and Microrna Expression in the Estrogen Response of Breast Cancer Cells DYNAMICS OF MRNA AND MICRORNA EXPRESSION IN THE ESTROGEN RESPONSE OF BREAST CANCER CELLS Jeanette Baran-Gale A dissertation submitted to the faculty at the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Curriculum in Bioinformatics and Computational Biology. Chapel Hill 2016 Approved by: Praveen Sethupathy Jeremy Purvis Terry Furey Jan Prins Alain Laederach Shawn Gomez © 2016 Jeanette Baran-Gale ALL RIGHTS RESERVED ii ABSTRACT Jeanette Baran-Gale: Dynamics of mRNA and microRNA expression in the estrogen response of breast cancer cells (Under the direction of Praveen Sethupathy and Jeremy Purvis) Cellular signaling leads to broad changes in gene expression that reprogram the cell and alter cell state. Signaling often begins with cellular receptors binding a ligand and initiating a transcriptional response. One example of this is the estrogen receptor, which binds the ligand estrogen and translocates to the nucleus where it binds to estrogen response elements and regulates the expression numerous target RNAs. The regulatory network of both messenger RNAs (mRNAs) and microRNAs (miRNAs) responding to estrogen stimulation is a complex, dynamic and multilayered program that is critical to the etiology of breast cancer. Estrogen receptor α (ERα) is an important biomarker of breast cancer severity and a common therapeutic target. Recent studies have demonstrated that in addition to its role in promoting proliferation, ERα also protects tumors against metastatic transformation. Current therapeutic strategies inhibit estrogen stimulated signaling and interfere with both beneficial and detrimental signaling pathways regulated by ERα. Additionally, ERα cyclically binds estrogen response elements and induces bursts of transcriptional activity. Together these observations suggest that ERα regulated genes and miRNAs may exhibit temporal variation in expression. Furthermore, it remains unclear if estrogen stimulated pathways exhibit the same temporal expression patterns, or if different pathways exhibit different temporal expression patterns. By combining both RNA-sequencing and small RNA-sequencing of cells responding to estrogen, we uncover the dynamics of both mRNA and miRNA expression in response to iii estrogen stimulation. Furthermore, we identify a regulatory circuit with potential therapeutic relevance to breast cancer that more specifically inhibits ERα-stimulated growth and survival pathways without interfering with its protective features. In response to estrogen stimulation, MCF7 cells (an estrogen receptor positive model cell line) exhibit induction of miR-503, and repression of the oncogene ZNF217. miR-503 inhibits proliferation in MCF7 cells, in part through its inhibition of the oncogene ZNF217 and the cell-cycle gene CCND1. While numerous regulatory interactions can be mined from this temporal profile of estrogen responsive mRNAs and miRNAs, the induction of the anti-proliferative microRNA, miR-503, both highlights the protective aspects of estrogen signaling and indicates that miR-503 holds promise as a therapeutic for breast cancer. iv ACKNOWLEDGEMENTS My project would not have been possible without the support and guidance of members of the UNC Chapel Hill community. I would like to thank the members of both the Sethupathy and Purvis labs. I am grateful for the daily help and guidance of my mentors Praveen Sethupathy and Jeremy Purvis, without which I would not be the scientist I am today. Their support and guidance has helped me to grow as a scientist, and thanks to that guidance I am a much better writer, presenter and experimentalist today than I was at the beginning of this journey. I would also like to thank Lisa Kurtz, for her extensive help in teaching me the laboratory techniques used throughout this work. Also, I would like to thank Sam Wolff, Bailey Peck, Denise Davis and Phil Coryell for additional guidance on experimental techniques. I also want to extend my thanks to the rest of my thesis committee: Terry Furey, Jan Prins, Alain Laederach and Shawn Gomez. Each of my committee members has provided invaluable feedback, constructive criticism, and guidance both during our official committee meetings and in one-on-one conversations. I am especially grateful to Terry Furey for (1) agreeing to be my committee chair, (2) keeping us all on track during committee meetings and (3) always having time to joke around with me. I am truly lucky to have the support of these amazing faculty members. I am truly grateful for all the support I have received from the entire community at UNC Chapel Hill. Interactions with my fellow Bioinformatics and Computational Biology graduate students, weekly Genetics department seminars, and numerous other scientific and professional development opportunities have all contributed to my scientific development. In particular, I v would like to thank Jessica Harrell and Bailey Peck for giving me the opportunity to co-develop and co-teach a Bioinformatics module to scholars from the UNC Postbaccalaureate Research Education Program. Lastly, I would like to thank my husband for his infinite patience, unending support and unswerving confidence in my eventual success. vi TABLE OF CONTENTS LIST OF TABLES ......................................................................................................................... x LIST OF FIGURES ...................................................................................................................... xi LIST OF ABBREVIATIONS ...................................................................................................... xiii CHAPTER 1 ................................................................................................................................. 2 Introduction ........................................................................................................................... 2 1.1 Dynamic RNA expression ........................................................................................... 2 1.2 Post-transcriptional repression by microRNAs ........................................................ 3 1.2.1 microRNA introduction ............................................................................................. 3 1.2.2 Considerations for small RNA-sequencing ............................................................ 4 1.3 Breast cancer and the estrogen response ................................................................ 6 1.3.1 miRNAs in breast cancer ....................................................................................... 7 1.3.2 Estrogen responsive coding RNAs ....................................................................... 8 1.4 Dynamic expression of coding and non-coding RNAs in breast cancer ......................................................................................................................... 8 CHAPTER 2: QUANTIFICATION OF MIRNAS AND THEIR ISOMIRS FROM HIGH THROUGHPUT SEQUENCING ........................................................................... 10 2.1 Overview ..................................................................................................................... 10 2.2 Introduction ................................................................................................................ 11 2.3 Results ........................................................................................................................ 15 2.3.1 A pipeline for detailed quantification of miRNAs and their isomiRs .................... 15 2.3.2 Comparing the miRNA and 5’-isomiR profiles of MIN6, human beta cell, and human islet ................................................................................... 17 2.3.3 Characterization of beta cell 5’-shifted isomiRs .................................................. 18 2.4 Discussion .................................................................................................................. 22 vii 2.5 Materials and methods .............................................................................................. 22 CHAPTER 3: ADDRESSING BIAS IN SMALL RNA LIBRARY PREPARATION FOR SEQUENCING: A NEW PROTOCOL RECOVERS MICRORNAS THAT EVADE CAPTURE BY CURRENT METHODS ................................................... 24 3.1 Overview ..................................................................................................................... 24 3.2 Introduction ................................................................................................................ 25 3.3 Results ........................................................................................................................ 29 3.4 Discussion .................................................................................................................. 36 3.5 Materials and Methods .............................................................................................. 39 CHAPTER 4: IDENTIFYING MIRNA “MASTER REGULATORS” THROUGH TARGET SITE ENRICHMENT ....................................................................................... 43 4.1 Overview ..................................................................................................................... 43 4.2 Introduction ................................................................................................................ 44 4.3 Results .......................................................................................................................
Recommended publications
  • Disruption of the Neuronal PAS3 Gene in a Family Affected with Schizophrenia D Kamnasaran, W J Muir, M a Ferguson-Smith,Dwcox
    325 ORIGINAL ARTICLE J Med Genet: first published as 10.1136/jmg.40.5.325 on 1 May 2003. Downloaded from Disruption of the neuronal PAS3 gene in a family affected with schizophrenia D Kamnasaran, W J Muir, M A Ferguson-Smith,DWCox ............................................................................................................................. J Med Genet 2003;40:325–332 Schizophrenia and its subtypes are part of a complex brain disorder with multiple postulated aetiolo- gies. There is evidence that this common disease is genetically heterogeneous, with many loci involved. See end of article for In this report, we describe a mother and daughter affected with schizophrenia, who are carriers of a authors’ affiliations t(9;14)(q34;q13) chromosome. By mapping on flow sorted aberrant chromosomes isolated from lym- ....................... phoblast cell lines, both subjects were found to have a translocation breakpoint junction between the Correspondence to: markers D14S730 and D14S70, a 683 kb interval on chromosome 14q13. This interval was found to Dr D W Cox, 8-39 Medical contain the neuronal PAS3 gene (NPAS3), by annotating the genomic sequence for ESTs and perform- Sciences Building, ing RACE and cDNA library screenings. The NPAS3 gene was characterised with respect to the University of Alberta, genomic structure, human expression profile, and protein cellular localisation to gain insight into gene Edmonton, Alberta T6G function. The translocation breakpoint junction lies within the third intron of NPAS3, resulting in the dis- 2H7, Canada; [email protected] ruption of the coding potential. The fact that the bHLH and PAS domains are disrupted from the remain- ing parts of the encoded protein suggests that the DNA binding and dimerisation functions of this Revised version received protein are destroyed.
    [Show full text]
  • Transcription Factor P73 Regulates Th1 Differentiation
    ARTICLE https://doi.org/10.1038/s41467-020-15172-5 OPEN Transcription factor p73 regulates Th1 differentiation Min Ren1, Majid Kazemian 1,4, Ming Zheng2, JianPing He3, Peng Li1, Jangsuk Oh1, Wei Liao1, Jessica Li1, ✉ Jonathan Rajaseelan1, Brian L. Kelsall 3, Gary Peltz 2 & Warren J. Leonard1 Inter-individual differences in T helper (Th) cell responses affect susceptibility to infectious, allergic and autoimmune diseases. To identify factors contributing to these response differ- 1234567890():,; ences, here we analyze in vitro differentiated Th1 cells from 16 inbred mouse strains. Haplotype-based computational genetic analysis indicates that the p53 family protein, p73, affects Th1 differentiation. In cells differentiated under Th1 conditions in vitro, p73 negatively regulates IFNγ production. p73 binds within, or upstream of, and modulates the expression of Th1 differentiation-related genes such as Ifng and Il12rb2. Furthermore, in mouse experimental autoimmune encephalitis, p73-deficient mice have increased IFNγ production and less dis- ease severity, whereas in an adoptive transfer model of inflammatory bowel disease, transfer of p73-deficient naïve CD4+ T cells increases Th1 responses and augments disease severity. Our results thus identify p73 as a negative regulator of the Th1 immune response, suggesting that p73 dysregulation may contribute to susceptibility to autoimmune disease. 1 Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, Bethesda, MD 20892-1674, USA. 2 Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305, USA. 3 Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA. 4Present address: Department of Biochemistry and Computer Science, Purdue University, West ✉ Lafayette, IN 37906, USA.
    [Show full text]
  • Regulation of Expression and Activity of the Bhlh-PAS Transcription
    Regulation of Expression and Activity of the bHLH-PAS Transcription Factor NPAS4 David Christopher Bersten B.Sc. (Biomedical Science), Honours (Biochemistry) A thesis submitted in fulfilment of the requirements for the degree of Doctor of Philosophy Discipline of Biochemistry School of Molecular and Biomedical Science University of Adelaide, Australia June 2014 1 Contents Abstract ................................................................................................................................................... 3 PhD Thesis Declaration ........................................................................................................................... 5 Acknowledgements ................................................................................................................................. 6 Publications ............................................................................................................................................. 8 Conference oral presentations ........................................................................................................... 9 Additional publications ....................................................................................................................... 9 Chapter 1: .............................................................................................................................................. 10 Introduction .....................................................................................................................................
    [Show full text]
  • Steroid-Dependent Regulation of the Oviduct: a Cross-Species Transcriptomal Analysis
    University of Kentucky UKnowledge Theses and Dissertations--Animal and Food Sciences Animal and Food Sciences 2015 Steroid-dependent regulation of the oviduct: A cross-species transcriptomal analysis Katheryn L. Cerny University of Kentucky, [email protected] Right click to open a feedback form in a new tab to let us know how this document benefits ou.y Recommended Citation Cerny, Katheryn L., "Steroid-dependent regulation of the oviduct: A cross-species transcriptomal analysis" (2015). Theses and Dissertations--Animal and Food Sciences. 49. https://uknowledge.uky.edu/animalsci_etds/49 This Doctoral Dissertation is brought to you for free and open access by the Animal and Food Sciences at UKnowledge. It has been accepted for inclusion in Theses and Dissertations--Animal and Food Sciences by an authorized administrator of UKnowledge. For more information, please contact [email protected]. STUDENT AGREEMENT: I represent that my thesis or dissertation and abstract are my original work. Proper attribution has been given to all outside sources. I understand that I am solely responsible for obtaining any needed copyright permissions. I have obtained needed written permission statement(s) from the owner(s) of each third-party copyrighted matter to be included in my work, allowing electronic distribution (if such use is not permitted by the fair use doctrine) which will be submitted to UKnowledge as Additional File. I hereby grant to The University of Kentucky and its agents the irrevocable, non-exclusive, and royalty-free license to archive and make accessible my work in whole or in part in all forms of media, now or hereafter known.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Supplementary Table 1
    Supplementary Table 1. 492 genes are unique to 0 h post-heat timepoint. The name, p-value, fold change, location and family of each gene are indicated. Genes were filtered for an absolute value log2 ration 1.5 and a significance value of p ≤ 0.05. Symbol p-value Log Gene Name Location Family Ratio ABCA13 1.87E-02 3.292 ATP-binding cassette, sub-family unknown transporter A (ABC1), member 13 ABCB1 1.93E-02 −1.819 ATP-binding cassette, sub-family Plasma transporter B (MDR/TAP), member 1 Membrane ABCC3 2.83E-02 2.016 ATP-binding cassette, sub-family Plasma transporter C (CFTR/MRP), member 3 Membrane ABHD6 7.79E-03 −2.717 abhydrolase domain containing 6 Cytoplasm enzyme ACAT1 4.10E-02 3.009 acetyl-CoA acetyltransferase 1 Cytoplasm enzyme ACBD4 2.66E-03 1.722 acyl-CoA binding domain unknown other containing 4 ACSL5 1.86E-02 −2.876 acyl-CoA synthetase long-chain Cytoplasm enzyme family member 5 ADAM23 3.33E-02 −3.008 ADAM metallopeptidase domain Plasma peptidase 23 Membrane ADAM29 5.58E-03 3.463 ADAM metallopeptidase domain Plasma peptidase 29 Membrane ADAMTS17 2.67E-04 3.051 ADAM metallopeptidase with Extracellular other thrombospondin type 1 motif, 17 Space ADCYAP1R1 1.20E-02 1.848 adenylate cyclase activating Plasma G-protein polypeptide 1 (pituitary) receptor Membrane coupled type I receptor ADH6 (includes 4.02E-02 −1.845 alcohol dehydrogenase 6 (class Cytoplasm enzyme EG:130) V) AHSA2 1.54E-04 −1.6 AHA1, activator of heat shock unknown other 90kDa protein ATPase homolog 2 (yeast) AK5 3.32E-02 1.658 adenylate kinase 5 Cytoplasm kinase AK7
    [Show full text]
  • Reconstruction and Analysis of Transcriptional Regulatory Networks with Trena
    Reconstruction and Analysis of Transcriptional Regulatory Networks with TReNA Seth A. Ament Institute for Systems Biology Seattle, Washington Genes influence phenotypes through a network of networks Social Network Brain Connectivity Network Individual Neuronal Network Molecular Network DNA Transcriptional Regulatory Network Analysis (TReNA) Sequence Motifs DNase footprints Epigenomic States Evolutionary Conservation JASPAR ENCODE ROADMAP/FANTOM phastCons FootprintFinder Tissue-Specific TF Binding Sites Software Availability: https://github.com/PriceLab/TReNA Transcriptional Regulatory Network Analysis (TReNA) Sequence Motifs DNase footprints Epigenomic States Evolutionary Conservation JASPAR ENCODE ROADMAP/FANTOM phastCons FootprintFinder Tissue-Specific Tissue-Specific Transcriptome Profiles TF Binding Sites GTEx/GEO fitTRN Tissue-Specific Transcriptional Regulatory Network (TF-Target Gene Interactions) Software Availability: https://github.com/PriceLab/TReNA Combining diverse annotations improves prediction of TF binding sites 1.0 TRUE/FALSE classes: USF1 DNase footprints with/without USF1 ChIP-seq 0.8 peaks 0.6 All USF1 footprints: 79% sensitivity Sensitivity 31% specificity 0.4 USF1 footprints with 0.2 modeled probability > 50%: FIMO + Wellington + ChromHMM + phastCons 55% sensitivity FIMO p−value 70% specificity Wellington p−value 0.0 1.0 0.8 0.6 0.4 0.2 0.0 Specificity Combining TF binding sites and gene co-expression improves prediction of TFs’ functional target genes Co-Expression TF Binding Sites Ensemble 1.0 0.8 *** *** 0.6 OC
    [Show full text]
  • Single Cell Transcriptomics in Schizophrenia Postmortem Brain: Moving Beyond Bulk Lysate
    SINGLE CELL TRANSCRIPTOMICS IN SCHIZOPHRENIA POSTMORTEM BRAIN: MOVING BEYOND BULK LYSATE Richard Crist June 8th, 2020 Schizophrenia Transcriptomics ■ Microarray and RNA sequencing ■ Differentially expressed genes across many cortical and sub-cortical regions – Dorsolateral prefrontal cortex (dlPFC) (Fillman et al, 2013) – Anterior Cingulate Cortex (Zhao et al, 2015; Hong et al, 2013) – Superior temporal gyrus (Wu et al, 2012) – Hippocampus (Hwang et al, 2013; Kohen et al, 2014) – Amygdala (Chang et al, 2017) ■ Enrichment of pathways and gene networks – Neural development – Axon guidance – Inflammation and immune-related proteins CommonMind Consortium ■ Largest transcriptomic analysis of schizophrenia – 258 cases/279 controls – RNAseq in dlPFC ■ 693 differentially expressed genes Fromer et al, 2016 Cell Diversity in Postmortem Brain ■ Brain, like all tissues, consists of many cell types – Major cell populations (e.g. astrocytes) – Distinct sub-populations (e.g. PVALB+ interneurons) ■ Problems in assessing differential expression in bulk lysate – Inability to identify which cells are affected – Missed expression changes in less common cell types Penney et al, 2019 Schizophrenia Single Cell Transcriptomics ■ Immunofluorescence and laser capture microdissection to collect individual populations of cells ■ Layer III/V pyramidal neurons (Arion et al, 2017) – 72 PFC samples – 36 cases/36 controls – 100 cells per layer for each sample – Expression assessed by microarray – 1,783 differentially expressed probe sets corresponding to 1,420 genes
    [Show full text]
  • Downregulation of Carnitine Acyl-Carnitine Translocase by Mirnas
    Page 1 of 288 Diabetes 1 Downregulation of Carnitine acyl-carnitine translocase by miRNAs 132 and 212 amplifies glucose-stimulated insulin secretion Mufaddal S. Soni1, Mary E. Rabaglia1, Sushant Bhatnagar1, Jin Shang2, Olga Ilkayeva3, Randall Mynatt4, Yun-Ping Zhou2, Eric E. Schadt6, Nancy A.Thornberry2, Deborah M. Muoio5, Mark P. Keller1 and Alan D. Attie1 From the 1Department of Biochemistry, University of Wisconsin, Madison, Wisconsin; 2Department of Metabolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey; 3Sarah W. Stedman Nutrition and Metabolism Center, Duke Institute of Molecular Physiology, 5Departments of Medicine and Pharmacology and Cancer Biology, Durham, North Carolina. 4Pennington Biomedical Research Center, Louisiana State University system, Baton Rouge, Louisiana; 6Institute for Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York. Corresponding author Alan D. Attie, 543A Biochemistry Addition, 433 Babcock Drive, Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, (608) 262-1372 (Ph), (608) 263-9608 (fax), [email protected]. Running Title: Fatty acyl-carnitines enhance insulin secretion Abstract word count: 163 Main text Word count: 3960 Number of tables: 0 Number of figures: 5 Diabetes Publish Ahead of Print, published online June 26, 2014 Diabetes Page 2 of 288 2 ABSTRACT We previously demonstrated that micro-RNAs 132 and 212 are differentially upregulated in response to obesity in two mouse strains that differ in their susceptibility to obesity-induced diabetes. Here we show the overexpression of micro-RNAs 132 and 212 enhances insulin secretion (IS) in response to glucose and other secretagogues including non-fuel stimuli. We determined that carnitine acyl-carnitine translocase (CACT, Slc25a20) is a direct target of these miRNAs.
    [Show full text]
  • Transcriptional Regulation of Neurodevelopmental and Metabolic Pathways by NPAS3
    Molecular Psychiatry (2012) 17, 267–279 & 2012 Macmillan Publishers Limited All rights reserved 1359-4184/12 www.nature.com/mp ORIGINAL ARTICLE Transcriptional regulation of neurodevelopmental and metabolic pathways by NPAS3 L Sha1,7, L MacIntyre2,7, JA Machell1, MP Kelly3, DJ Porteous1, NJ Brandon3, WJ Muir4,{, DH Blackwood4, DG Watson2, SJ Clapcote5 and BS Pickard2,6 1Department of Medical Genetics, Institute for Genetics and Molecular Medicine, University of Edinburgh, Molecular Medicine Centre, Western General Hospital, Edinburgh, UK; 2Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK; 3Pfizer, Pfizer Global Research and Development, Neuroscience Research Unit, Groton, CT, USA; 4Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK; 5Institute of Membrane and Systems Biology, University of Leeds, Leeds, UK and 6CeNsUS—Centre for Neuroscience, University of Strathclyde, Glasgow, UK The basic helix-loop-helix PAS (Per, Arnt, Sim) domain transcription factor gene NPAS3 is a replicated genetic risk factor for psychiatric disorders. A knockout (KO) mouse model exhibits behavioral and adult neurogenesis deficits consistent with human illness. To define the location and mechanism of NPAS3 etiopathology, we combined immunofluorescent, transcriptomic and metabonomic approaches. Intense Npas3 immunoreactivity was observed in the hippocampal subgranular zone—the site of adult neurogenesis—but was restricted to maturing, rather than proliferating, neuronal precursor cells. Microarray analysis of a HEK293 cell line over-expressing NPAS3 showed that transcriptional targets varied according to circadian rhythm context and C-terminal deletion. The most highly up-regulated NPAS3 target gene, VGF, encodes secretory peptides with established roles in neurogenesis, depression and schizophrenia. VGF was just one of many NPAS3 target genes also regulated by the SOX family of transcription factors, suggesting an overlap in neurodevelopmental function.
    [Show full text]
  • NPAS1-ARNT and NPAS3-ARNT Crystal Structures Implicate The
    RESEARCH ARTICLE NPAS1-ARNT and NPAS3-ARNT crystal structures implicate the bHLH-PAS family as multi-ligand binding transcription factors Dalei Wu1†, Xiaoyu Su1, Nalini Potluri1, Youngchang Kim2, Fraydoon Rastinejad1* 1Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, United States; 2Structural Biology Center, Biosciences Division, Argonne National Laboratory, Argonne, United States Abstract The neuronal PAS domain proteins NPAS1 and NPAS3 are members of the basic helix- loop-helix-PER-ARNT-SIM (bHLH-PAS) family, and their genetic deficiencies are linked to a variety of human psychiatric disorders including schizophrenia, autism spectrum disorders and bipolar disease. NPAS1 and NPAS3 must each heterodimerize with the aryl hydrocarbon receptor nuclear translocator (ARNT), to form functional transcription complexes capable of DNA binding and gene regulation. Here we examined the crystal structures of multi-domain NPAS1-ARNT and NPAS3- ARNT-DNA complexes, discovering each to contain four putative ligand-binding pockets. Through expanded architectural comparisons between these complexes and HIF-1a-ARNT, HIF-2a-ARNT *For correspondence: and CLOCK-BMAL1, we show the wider mammalian bHLH-PAS family is capable of multi-ligand- [email protected] binding and presents as an ideal class of transcription factors for direct targeting by small-molecule Present address: †State Key drugs. Laboratory of Microbial DOI: 10.7554/eLife.18790.001 Technology, School of Life Sciences, Shandong University, Qingdao, China Competing interests: The Introduction authors declare that no The mammalian bHLH-PAS transcription factors share a common protein architecture consisting of a competing interests exist. conserved bHLH DNA-binding domain, tandem PAS domains (PAS-A and PAS-B), and a variable activation or repression domain.
    [Show full text]
  • Perkinelmer Genomics to Request the Saliva Swab Collection Kit for Patients That Cannot Provide a Blood Sample As Whole Blood Is the Preferred Sample
    Autism and Intellectual Disability TRIO Panel Test Code TR002 Test Summary This test analyzes 2429 genes that have been associated with Autism and Intellectual Disability and/or disorders associated with Autism and Intellectual Disability with the analysis being performed as a TRIO Turn-Around-Time (TAT)* 3 - 5 weeks Acceptable Sample Types Whole Blood (EDTA) (Preferred sample type) DNA, Isolated Dried Blood Spots Saliva Acceptable Billing Types Self (patient) Payment Institutional Billing Commercial Insurance Indications for Testing Comprehensive test for patients with intellectual disability or global developmental delays (Moeschler et al 2014 PMID: 25157020). Comprehensive test for individuals with multiple congenital anomalies (Miller et al. 2010 PMID 20466091). Patients with autism/autism spectrum disorders (ASDs). Suspected autosomal recessive condition due to close familial relations Previously negative karyotyping and/or chromosomal microarray results. Test Description This panel analyzes 2429 genes that have been associated with Autism and ID and/or disorders associated with Autism and ID. Both sequencing and deletion/duplication (CNV) analysis will be performed on the coding regions of all genes included (unless otherwise marked). All analysis is performed utilizing Next Generation Sequencing (NGS) technology. CNV analysis is designed to detect the majority of deletions and duplications of three exons or greater in size. Smaller CNV events may also be detected and reported, but additional follow-up testing is recommended if a smaller CNV is suspected. All variants are classified according to ACMG guidelines. Condition Description Autism Spectrum Disorder (ASD) refers to a group of developmental disabilities that are typically associated with challenges of varying severity in the areas of social interaction, communication, and repetitive/restricted behaviors.
    [Show full text]