2016 Mian Syed Ethesis

Total Page:16

File Type:pdf, Size:1020Kb

2016 Mian Syed Ethesis This electronic thesis or dissertation has been downloaded from the King’s Research Portal at https://kclpure.kcl.ac.uk/portal/ Unravelling the molecular pathogenesis of Myelodysplastic Syndrome Patients with Ringed Sideroblasts Mian, Syed Asif Awarding institution: King's College London The copyright of this thesis rests with the author and no quotation from it or information derived from it may be published without proper acknowledgement. END USER LICENCE AGREEMENT Unless another licence is stated on the immediately following page this work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International licence. https://creativecommons.org/licenses/by-nc-nd/4.0/ You are free to copy, distribute and transmit the work Under the following conditions: Attribution: You must attribute the work in the manner specified by the author (but not in any way that suggests that they endorse you or your use of the work). Non Commercial: You may not use this work for commercial purposes. No Derivative Works - You may not alter, transform, or build upon this work. Any of these conditions can be waived if you receive permission from the author. Your fair dealings and other rights are in no way affected by the above. Take down policy If you believe that this document breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 06. Oct. 2021 Unravelling the molecular pathogenesis of Myelodysplastic Syndrome Patients with Ringed Sideroblasts Syed Asif Mian Thesis submitted for the Degree of Doctor of Philosophy King’s College London October 2015 1 Declaration of Authorship I hereby declare that I alone composed this thesis and that work presented here is my own, except where stated otherwise. October 2015 2 Abstract Myelodysplastic syndromes (MDS) are a collection of clonal hematopoietic stem cell disorders with diverse phenotypes, characterized mainly by ineffective haematopoiesis. Refractory anaemia with ringed sideroblasts (RARS) is an acquired form of MDS with accumulation of mitochondrial iron that gives rise to ringed sideroblasts. In this study, whole-exome sequencing performed in 12 patients all of whom had >25% ringed sideroblasts identified mutations in one particular gene, splicing factor 3b subunit 1 (SF3B1), a component of the major and minor spliceosomes, in 11/12 cases. No SF3B1 mutations were observed in a single case of congenital sideroblastic anaemia, however, a constitutional mutation in ALAS1 gene was confirmed in this patient. Subsequently, amplicon sequencing using a 22 myeloid-gene panel in 154 MDS patients revealed 76% (n=117) of the patients had mutations in at least one of the genes, with 38% (n=59) having splicing gene mutations and 49% (n=75) patients harbouring more than one gene mutation. Interestingly, single and specific epigenetic modifier mutations tended to coexist with SF3B1 and SRSF2 mutations (p<0.03). Moreover, patients with splicing factor mutations alone had a better overall survival than those with coexisting additional mutations. Next, combination of techniques including xenotransplantation and single cell clonogenic assays were used to study the characteristics of SF3B1 mutant cells. Mutational analysis of hematopoietic stem/progenitor cells showed that SF3B1 mutations arise in phenotypically defined CD34+CD38-CD45RA-CD90+CD49f+ stem cells, therefore revealing the existence of MDS-initiating stem cells that propagate mutations to myeloid progenitors only. A persistent engraftment restricted to myeloid lineage was observed which was initiated only via the xenotransplantation of HSCs. Genetically diverse evolving subclones of mutant SF3B1 were detected in mice, indicating a branching multi-clonal as well as ancestral evolutionary paradigm. Subclonal evolution in mice was also mirrored in the clinical evolution in patients. Sequential sample analysis uncovered a clonal evolution in hematopoietic progeny and selection of the malignant driving clone leading to AML transformation. 3 Acknowledgements Apart from one’s own efforts, the success of any project depends largely on the encouragement and guidelines of many others. I would like to take this opportunity to express my gratitude to the people who have been instrumental in the successful completion of my PhD. First of all I would like to show my greatest appreciation and extend my heartfelt gratitude to my supervisors Prof. Ghulam J Mufti and Dr. Alexander Smith for giving me the opportunity to carry out this work which was funded by Leukaemia and Lymphoma Research. I can’t say thank you enough for their tremendous support and help which I received whilst working on this project. Without their encouragement and guidance this project would not have been successful. I would also like to thank all the patients who have donated their bone marrow tissues to King’s College London tissue bank. Also, I would like to acknowledge and extend my heartfelt gratitude to the people who made the completion of my project successful including Prof. Dominique Bonnet and Dr. Kevin Rouault. I owe my most sincere gratitude to Dr. Aytug Kizilors not only for her scientific advice but also for her deep understanding and help during my difficult moments. I would like to thank my colleagues in the department of Haematological medicine for their help and support when it was needed, but most of all for making me feel at home amongst them. It was a pleasure to work with them all. I would also like to thank all my friends especially Selen Bozkurt for her support and encouragement over the years. Finally, I am forever indebted to my family especially my parents for their understanding, encouragement, and endless love ‘come rain or shine’. I owe them so much. 4 Table of Contents Title…………………………………………………………………….…………………….. 1 Declaration………………………………………………………………………………..… 2 Abstract………………………………………………………………………………..……. 3 Acknowledgements………………………………………………………………………… 4 List of Figures…………………………………………………………………………...….. 9 List of Tables…………………………………………………………………………..….. 11 List of Abbreviations……………………………………………………………………… 13 1 General Introduction ....................................................................................... 15 1.1 Myelodysplastic syndromes ....................................................................................... 16 1.2 Spectrum of cytogenetic abnormalities in MDS ...................................................... 20 1.2.1 Deletion 5q ................................................................................................................... 21 1.2.2 Chromosome 7 and 7q deletion ................................................................................ 22 1.2.3 Trisomy 8 ...................................................................................................................... 23 1.2.4 Other Cytogenic Lesions ............................................................................................ 23 1.3 SNP Karyotyping ......................................................................................................... 24 1.4 Gene Mutations in MDS ............................................................................................. 25 1.4.1 Epigenetic modifier gene mutations ......................................................................... 26 1.4.2 Transcription regulation/Cell signalling pathway gene mutations ........................ 37 1.5 Spliceosome ................................................................................................................ 40 1.5.1 Splicing factor-3 b1 (SF3B1) ..................................................................................... 46 1.6 Refractory anaemia with ring sideroblasts and Iron metabolism ......................... 49 1.7 Therapeutic strategies for RARS .............................................................................. 59 1.8 High throughput molecular technologies ................................................................. 60 5 1.8.1 Second Generation Sequencing ............................................................................... 60 1.8.2 Microarray analysis ..................................................................................................... 62 1.9 Aims .............................................................................................................................. 64 2 Materials and Methods ................................................................................... 65 2.1 Materials ....................................................................................................................... 66 2.1.1 Patient Samples .......................................................................................................... 66 2.1.2 Commercial Kits .......................................................................................................... 69 2.1.3 Buffers ........................................................................................................................... 70 2.2 Methods ........................................................................................................................ 72 2.2.1 Extraction of Genomic DNA ....................................................................................... 72 2.2.2 DNA Quantitation using Quant-iT picoGreen dsDNA Assay kit. .......................... 72 2.2.3 RNA Extraction ...........................................................................................................
Recommended publications
  • 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]
  • Download Author Version (PDF)
    Molecular BioSystems Accepted Manuscript This is an Accepted Manuscript, which has been through the Royal Society of Chemistry peer review process and has been accepted for publication. Accepted Manuscripts are published online shortly after acceptance, before technical editing, formatting and proof reading. Using this free service, authors can make their results available to the community, in citable form, before we publish the edited article. We will replace this Accepted Manuscript with the edited and formatted Advance Article as soon as it is available. You can find more information about Accepted Manuscripts in the Information for Authors. Please note that technical editing may introduce minor changes to the text and/or graphics, which may alter content. The journal’s standard Terms & Conditions and the Ethical guidelines still apply. In no event shall the Royal Society of Chemistry be held responsible for any errors or omissions in this Accepted Manuscript or any consequences arising from the use of any information it contains. www.rsc.org/molecularbiosystems Page 1 of 29 Molecular BioSystems Mutated Genes and Driver Pathways Involved in Myelodysplastic Syndromes—A Transcriptome Sequencing Based Approach Liang Liu1*, Hongyan Wang1*, Jianguo Wen2*, Chih-En Tseng2,3*, Youli Zu2, Chung-che Chang4§, Xiaobo Zhou1§ 1 Center for Bioinformatics and Systems Biology, Division of Radiologic Sciences, Wake Forest University Baptist Medical Center, Winston-Salem, NC 27157, USA. 2 Department of Pathology, the Methodist Hospital Research Institute,
    [Show full text]
  • Phosphoproteomics of Retinoblastoma: a Pilot Study Identifies Aberrant Kinases
    molecules Article Phosphoproteomics of Retinoblastoma: A Pilot Study Identifies Aberrant Kinases Lakshmi Dhevi Nagarajha Selvan 1,†, Ravikanth Danda 1,2,†, Anil K. Madugundu 3 ID , Vinuth N. Puttamallesh 3, Gajanan J. Sathe 3,4, Uma Maheswari Krishnan 2, Vikas Khetan 5, Pukhraj Rishi 5, Thottethodi Subrahmanya Keshava Prasad 3,6 ID , Akhilesh Pandey 3,7,8, Subramanian Krishnakumar 1, Harsha Gowda 3,* and Sailaja V. Elchuri 9,* 1 L&T Opthalmic Pathology, Vision Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu 600 006, India; [email protected] (L.D.N.S.); [email protected] (R.D.); [email protected] (S.K.) 2 Centre for Nanotechnology and Advanced Biomaterials, Shanmugha Arts, Science, Technology and Research Academy University, Tanjore, Tamil Nadu 613 401, India; [email protected] 3 Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka 560 066, India; [email protected] (A.K.M.); [email protected] (V.N.P.); [email protected] (G.J.S.); [email protected] (T.S.K.P.); [email protected] (A.P.) 4 Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India 5 Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu 600 006, India; [email protected] (V.K.); [email protected] (P.R.) 6 Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575 108, India 7 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 8 Departments of Biological Chemistry, Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 9 Department of Nanotechnology, Vision Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu 600 006, India * Correspondence: [email protected] (H.G.); [email protected] (S.V.E.); Tel.: +91-80-28416140 (H.G.); +91-44-28271616 (S.V.E.) † These authors contributed equally to this work.
    [Show full text]
  • Application of Microrna Database Mining in Biomarker Discovery and Identification of Therapeutic Targets for Complex Disease
    Article Application of microRNA Database Mining in Biomarker Discovery and Identification of Therapeutic Targets for Complex Disease Jennifer L. Major, Rushita A. Bagchi * and Julie Pires da Silva * Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; [email protected] * Correspondence: [email protected] (R.A.B.); [email protected] (J.P.d.S.) Supplementary Tables Methods Protoc. 2021, 4, 5. https://doi.org/10.3390/mps4010005 www.mdpi.com/journal/mps Methods Protoc. 2021, 4, 5. https://doi.org/10.3390/mps4010005 2 of 25 Table 1. List of all hsa-miRs identified by Human microRNA Disease Database (HMDD; v3.2) analysis. hsa-miRs were identified using the term “genetics” and “circulating” as input in HMDD. Targets CAD hsa-miR-1 Targets IR injury hsa-miR-423 Targets Obesity hsa-miR-499 hsa-miR-146a Circulating Obesity Genetics CAD hsa-miR-423 hsa-miR-146a Circulating CAD hsa-miR-149 hsa-miR-499 Circulating IR Injury hsa-miR-146a Circulating Obesity hsa-miR-122 Genetics Stroke Circulating CAD hsa-miR-122 Circulating Stroke hsa-miR-122 Genetics Obesity Circulating Stroke hsa-miR-26b hsa-miR-17 hsa-miR-223 Targets CAD hsa-miR-340 hsa-miR-34a hsa-miR-92a hsa-miR-126 Circulating Obesity Targets IR injury hsa-miR-21 hsa-miR-423 hsa-miR-126 hsa-miR-143 Targets Obesity hsa-miR-21 hsa-miR-223 hsa-miR-34a hsa-miR-17 Targets CAD hsa-miR-223 hsa-miR-92a hsa-miR-126 Targets IR injury hsa-miR-155 hsa-miR-21 Circulating CAD hsa-miR-126 hsa-miR-145 hsa-miR-21 Targets Obesity hsa-mir-223 hsa-mir-499 hsa-mir-574 Targets IR injury hsa-mir-21 Circulating IR injury Targets Obesity hsa-mir-21 Targets CAD hsa-mir-22 hsa-mir-133a Targets IR injury hsa-mir-155 hsa-mir-21 Circulating Stroke hsa-mir-145 hsa-mir-146b Targets Obesity hsa-mir-21 hsa-mir-29b Methods Protoc.
    [Show full text]
  • Human Prefoldin Modulates Co-Transcriptional Pre-Mrna Splicing
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.14.150466; this version posted July 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. BIOLOGICAL SCIENCES: Biochemistry Human prefoldin modulates co-transcriptional pre-mRNA splicing Payán-Bravo L 1,2, Peñate X 1,2 *, Cases I 3, Pareja-Sánchez Y 1, Fontalva S 1,2, Odriozola Y 1,2, Lara E 1, Jimeno-González S 2,5, Suñé C 4, Reyes JC 5, Chávez S 1,2. 1 Instituto de Biomedicina de Sevilla, Universidad de Sevilla-CSIC-Hospital Universitario V. del Rocío, Seville, Spain. 2 Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain. 3 Centro Andaluz de Biología del Desarrollo, CSIC-Universidad Pablo de Olavide, Seville, Spain. 4 Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" IPBLN-CSIC, PTS, Granada, Spain. 5 Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain. Correspondence: Sebastián Chávez, IBiS, campus HUVR, Avda. Manuel Siurot s/n, Sevilla, 41013, Spain. Phone: +34-955923127: e-mail: [email protected]. * Co- corresponding; [email protected]. bioRxiv preprint doi: https://doi.org/10.1101/2020.06.14.150466; this version posted July 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract Prefoldin is a heterohexameric complex conserved from archaea to humans that plays a cochaperone role during the cotranslational folding of actin and tubulin monomers.
    [Show full text]
  • Mutations in Splicing Factor Genes in Myeloid Malignancies: Significance and Impact on Clinical Features
    cancers Review Mutations in Splicing Factor Genes in Myeloid Malignancies: Significance and Impact on Clinical Features Valeria Visconte 1, Megan O. Nakashima 2 and Heesun J. Rogers 2,* 1 Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; [email protected] 2 Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-216-445-2719 Received: 15 October 2019; Accepted: 19 November 2019; Published: 22 November 2019 Abstract: Components of the pre-messenger RNA splicing machinery are frequently mutated in myeloid malignancies. Mutations in LUC7L2, PRPF8, SF3B1, SRSF2, U2AF1, and ZRSR2 genes occur at various frequencies ranging between 40% and 85% in different subtypes of myelodysplastic syndrome (MDS) and 5% and 10% of acute myeloid leukemia (AML) and myeloproliferative neoplasms (MPNs). In some instances, splicing factor (SF) mutations have provided diagnostic utility and information on clinical outcomes as exemplified by SF3B1 mutations associated with increased ring sideroblasts (RS) in MDS-RS or MDS/MPN-RS with thrombocytosis. SF3B1 mutations are associated with better survival outcomes, while SRSF2 mutations are associated with a shorter survival time and increased AML progression, and U2AF1 mutations with a lower remission rate and shorter survival time. Beside the presence of mutations, transcriptomics technologies have shown that one third of genes in AML patients are differentially expressed, leading to altered transcript stability, interruption of protein function, and improper translation compared to those of healthy individuals. The detection of SF mutations demonstrates the importance of splicing abnormalities in the hematopoiesis of MDS and AML patients given the fact that abnormal splicing regulates the function of several transcriptional factors (PU.1, RUNX1, etc.) crucial in hematopoietic function.
    [Show full text]
  • A High Throughput, Functional Screen of Human Body Mass Index GWAS Loci Using Tissue-Specific Rnai Drosophila Melanogaster Crosses Thomas J
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2018 A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J. Baranski Washington University School of Medicine in St. Louis Aldi T. Kraja Washington University School of Medicine in St. Louis Jill L. Fink Washington University School of Medicine in St. Louis Mary Feitosa Washington University School of Medicine in St. Louis Petra A. Lenzini Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Baranski, Thomas J.; Kraja, Aldi T.; Fink, Jill L.; Feitosa, Mary; Lenzini, Petra A.; Borecki, Ingrid B.; Liu, Ching-Ti; Cupples, L. Adrienne; North, Kari E.; and Province, Michael A., ,"A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses." PLoS Genetics.14,4. e1007222. (2018). https://digitalcommons.wustl.edu/open_access_pubs/6820 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Thomas J. Baranski, Aldi T. Kraja, Jill L. Fink, Mary Feitosa, Petra A. Lenzini, Ingrid B. Borecki, Ching-Ti Liu, L. Adrienne Cupples, Kari E. North, and Michael A. Province This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/6820 RESEARCH ARTICLE A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J.
    [Show full text]
  • Human PRPF40B Regulates Hundreds of Alternative Splicing Targets and Represses a Hypoxia Expression Signature
    Downloaded from rnajournal.cshlp.org on October 10, 2021 - Published by Cold Spring Harbor Laboratory Press Human PRPF40B regulates hundreds of alternative splicing targets and represses a hypoxia expression signature PAOLO ALBERTO LORENZINI,1,2 RESILIND SU ERN CHEW,1 CHERYL WEIQI TAN,1 JING YEN YONG,1 FAN ZHANG,3 JIE ZHENG,3,4 and XAVIER ROCA1 1School of Biological Sciences, Nanyang Technological University, 637551 Singapore, Singapore 2Nanyang Institute of Technology in Health and Medicine, Interdisciplinary Graduate School (IGS), Nanyang Technological University, 637551 Singapore, Singapore 3School of Computer Science and Engineering, Nanyang Technological University, 637551 Singapore, Singapore 4School of Information Science and Technology, ShanghaiTech University, Pudong District, Shanghai 201210, China ABSTRACT Altered splicing contributes to the pathogenesis of human blood disorders including myelodysplastic syndromes (MDS) and leukemias. Here we characterize the transcriptomic regulation of PRPF40B, which is a splicing factor mutated in a small fraction of MDS patients. We generated a full PRPF40B knockout (KO) in the K562 cell line by CRISPR/Cas9 technology and rescued its levels by transient overexpression of wild-type (WT), P383L or P540S MDS alleles. Using RNA sequencing, we identified hundreds of differentially expressed genes and alternative splicing (AS) events in the KO that are rescued by WT PRPF40B, with a majority also rescued by MDS alleles, pointing to mild effects of these mutations. Among the PRPF40B- regulated AS events, we found a net increase in exon inclusion in the KO, suggesting that this splicing factor primarily acts as a repressor. PRPF40B-regulated splicing events are likely cotranscriptional, affecting exons with A-rich downstream intronic motifs and weak splice sites especially for 5′′′′′ splice sites, consistent with its PRP40 yeast ortholog being part of the U1 small nuclear ribonucleoprotein.
    [Show full text]
  • The Pioneer Transcription Factors Foxa1 and Foxa2 Regulate Alternative RNA Splicing During Thymocyte Positive Selection Ching-In Lau1, Jasmine Rowell1, Diana C
    © 2021. Published by The Company of Biologists Ltd | Development (2021) 148, dev199754. doi:10.1242/dev.199754 RESEARCH ARTICLE The pioneer transcription factors Foxa1 and Foxa2 regulate alternative RNA splicing during thymocyte positive selection Ching-In Lau1, Jasmine Rowell1, Diana C. Yanez1, Anisha Solanki1, Susan Ross1, Masahiro Ono2 and Tessa Crompton1,* ABSTRACT and is followed by negative selection of potentially self-reactive During positive selection at the transition from CD4+CD8+ double- clones and selection of regulatory T cells (Tregs) (Huynh et al., positive (DP) to single-positive (SP) thymocyte, TCR signalling 2014; Littman, 2016; Starr et al., 2003). The strength and duration results in appropriate MHC restriction and signals for survival and of the TCR signal that a developing cell receives broadly determine progression. We show that the pioneer transcription factors Foxa1 its fate, with the strongest signals leading to negative selection and Foxa2 are required to regulate RNA splicing during positive or CD4 Treg differentiation, usually at the SP stage in the medulla, selection of mouse T cells and that Foxa1 and Foxa2 have intermediate signals leading to positive selection usually in the overlapping/compensatory roles. Conditional deletion of both Foxa1 cortex, and weaker signals or lack of TCR signalling leading to and Foxa2 from DP thymocytes reduced positive selection and death by neglect (Singer et al., 2008). For DP thymocytes development of CD4SP, CD8SP and peripheral naïve CD4+ T cells. undergoing positive selection, TCR signal strength and duration Foxa1 and Foxa2 regulated the expression of many genes encoding also influence CD4 and CD8 lineage choice. Those cells receiving splicing factors and regulators, including Mbnl1, H1f0, Sf3b1, stronger and longer TCR signals tend towards the CD4SP fate, Hnrnpa1, Rnpc3, Prpf4b, Prpf40b and Snrpd3.
    [Show full text]
  • Detection of H3k4me3 Identifies Neurohiv Signatures, Genomic
    viruses Article Detection of H3K4me3 Identifies NeuroHIV Signatures, Genomic Effects of Methamphetamine and Addiction Pathways in Postmortem HIV+ Brain Specimens that Are Not Amenable to Transcriptome Analysis Liana Basova 1, Alexander Lindsey 1, Anne Marie McGovern 1, Ronald J. Ellis 2 and Maria Cecilia Garibaldi Marcondes 1,* 1 San Diego Biomedical Research Institute, San Diego, CA 92121, USA; [email protected] (L.B.); [email protected] (A.L.); [email protected] (A.M.M.) 2 Departments of Neurosciences and Psychiatry, University of California San Diego, San Diego, CA 92103, USA; [email protected] * Correspondence: [email protected] Abstract: Human postmortem specimens are extremely valuable resources for investigating trans- lational hypotheses. Tissue repositories collect clinically assessed specimens from people with and without HIV, including age, viral load, treatments, substance use patterns and cognitive functions. One challenge is the limited number of specimens suitable for transcriptional studies, mainly due to poor RNA quality resulting from long postmortem intervals. We hypothesized that epigenomic Citation: Basova, L.; Lindsey, A.; signatures would be more stable than RNA for assessing global changes associated with outcomes McGovern, A.M.; Ellis, R.J.; of interest. We found that H3K27Ac or RNA Polymerase (Pol) were not consistently detected by Marcondes, M.C.G. Detection of H3K4me3 Identifies NeuroHIV Chromatin Immunoprecipitation (ChIP), while the enhancer H3K4me3 histone modification was Signatures, Genomic Effects of abundant and stable up to the 72 h postmortem. We tested our ability to use H3K4me3 in human Methamphetamine and Addiction prefrontal cortex from HIV+ individuals meeting criteria for methamphetamine use disorder or not Pathways in Postmortem HIV+ Brain (Meth +/−) which exhibited poor RNA quality and were not suitable for transcriptional profiling.
    [Show full text]
  • SUPPLEMENTARY MATERIALS and METHODS PBMC Transcriptomics
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) Gut SUPPLEMENTARY MATERIALS AND METHODS PBMC transcriptomics identifies immune-metabolism disorder during the development of HBV-ACLF Contents l Supplementary methods l Supplementary Figure 1 l Supplementary Figure 2 l Supplementary Figure 3 l Supplementary Figure 4 l Supplementary Figure 5 l Supplementary Table 1 l Supplementary Table 2 l Supplementary Table 3 l Supplementary Table 4 l Supplementary Tables 5-14 l Supplementary Table 15 l Supplementary Table 16 l Supplementary Table 17 Li J, et al. Gut 2021;0:1–13. doi: 10.1136/gutjnl-2020-323395 BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) Gut SUPPLEMENTARY METHODS Test for HBV DNA The levels of HBV DNA were detected using real-time PCR with a COBAS® AmpliPrep/COBAS® TaqMan 48 System (Roche, Basel, Switzerland) and HBV Test v2.0. Criteria for diagnosing cirrhosis Pathology The gold standard for the diagnosis of cirrhosis is a liver biopsy obtained through a percutaneous or transjugular approach.1 Ultrasonography was performed 2-4 hours before biopsy. Liver biopsy specimens were obtained by experienced physicians. Percutaneous transthoracic puncture of the liver was performed according to the standard criteria. After biopsy, patients were monitored in the hospital with periodic analyses of haematocrit and other vital signs for 24 hours. Cirrhosis was diagnosed according to the globally agreed upon criteria.2 Cirrhosis is defined based on its pathological features under a microscope: (a) the presence of parenchymal nodules, (b) differences in liver cell size and appearance, (c) fragmentation of the biopsy specimen, (d) fibrous septa, and (d) an altered architecture and vascular relationships.
    [Show full text]
  • Alternative Splicing and Cancer: a Systematic Review
    Signal Transduction and Targeted Therapy www.nature.com/sigtrans REVIEW ARTICLE OPEN Alternative splicing and cancer: a systematic review Yuanjiao Zhang1,2, Jinjun Qian 2, Chunyan Gu 1,2 and Ye Yang1,2 The abnormal regulation of alternative splicing is usually accompanied by the occurrence and development of tumors, which would produce multiple different isoforms and diversify protein expression. The aim of the present study was to conduct a systematic review in order to describe the regulatory mechanisms of alternative splicing, as well as its functions in tumor cells, from proliferation and apoptosis to invasion and metastasis, and from angiogenesis to metabolism. The abnormal splicing events contributed to tumor progression as oncogenic drivers and/or bystander factors. The alterations in splicing factors detected in tumors and other mis-splicing events (i.e., long non-coding and circular RNAs) in tumorigenesis were also included. The findings of recent therapeutic approaches targeting splicing catalysis and splicing regulatory proteins to modulate pathogenically spliced events (including tumor-specific neo-antigens for cancer immunotherapy) were introduced. The emerging RNA-based strategies for the treatment of cancer with abnormally alternative splicing isoforms were also discussed. However, further studies are still required to address the association between alternative splicing and cancer in more detail. Signal Transduction and Targeted Therapy (2021) ;6:78 https://doi.org/10.1038/s41392-021-00486-7 1234567890();,: INTRODUCTION
    [Show full text]