151
|
Domínguez-Soto A, de las Casas-Engel M, Bragado R, Medina-Echeverz J, Aragoneses-Fenoll L, Martín-Gayo E, van Rooijen N, Berraondo P, Toribio ML, Moro MA, Cuartero I, Castrillo A, Sancho D, Sánchez-Torres C, Bruhns P, Sánchez-Ramón S, Corbí AL. Intravenous immunoglobulin promotes antitumor responses by modulating macrophage polarization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:5181-9. [PMID: 25326025 DOI: 10.4049/jimmunol.1303375] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intravenous Igs (IVIg) therapy is widely used as an immunomodulatory strategy in inflammatory pathologies and is suggested to promote cancer regression. Because progression of tumors depends on their ability to redirect the polarization state of tumor-associated macrophages (from M1/immunogenic/proinflammatory to M2/anti-inflammatory), we have evaluated whether IVIg limits tumor progression and dissemination through modulation of macrophage polarization. In vitro, IVIg inhibited proinflammatory cytokine production from M1 macrophages and induced a M2-to-M1 polarization switch on human and murine M2 macrophages. In vivo, IVIg modified the polarization of tumor-associated myeloid cells in a Fcεr1γ chain-dependent manner, modulated cytokine blood levels in tumor-bearing animals, and impaired tumor progression via FcγRIII (CD16), FcγRIV, and FcRγ engagement, the latter two effects being macrophage mediated. Therefore, IVIg immunomodulatory activity is dependent on the polarization state of the responding macrophages, and its ability to trigger a M2-to-M1 macrophage polarization switch might be therapeutically useful in cancer, in which proinflammatory or immunogenic functions should be promoted.
Collapse
Affiliation(s)
- Angeles Domínguez-Soto
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain;
| | - Mateo de las Casas-Engel
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain
| | - Rafael Bragado
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid 28040, Spain
| | | | - Laura Aragoneses-Fenoll
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain
| | - Enrique Martín-Gayo
- Centro de Biología Molecular, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Free University Medical Center, Amsterdam 1081, the Netherlands
| | | | - María L Toribio
- Centro de Biología Molecular, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - María A Moro
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Isabel Cuartero
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid 28029, Spain; Unidad Asociada de Biomedicina, Consejo Superior de Investigaciones Cientificas-Universidad de Las Palmas de Gran Canarias 35001, Spain
| | - David Sancho
- Fundación Centro Nacional de Investigaciones Cardiovasculares, Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain
| | - Carmen Sánchez-Torres
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico
| | - Pierre Bruhns
- Institut Pasteur, Département d'Immunologie, Unité des Anticorps en Thérapie et Pathologie, Paris 75015, France; INSERM U760, Paris 75015, France; and
| | | | - Angel L Corbí
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain;
| |
Collapse
|
152
|
Uncoupling Malt1 Threshold Function from Paracaspase Activity Results in Destructive Autoimmune Inflammation. Cell Rep 2014; 9:1292-305. [DOI: 10.1016/j.celrep.2014.10.044] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 09/15/2014] [Accepted: 10/15/2014] [Indexed: 11/21/2022] Open
|
153
|
Abstract
Many ribonucleases (RNases) are able to inhibit the reproduction of viruses in infected cell cultures and laboratory animals, but the molecular mechanisms of their antiviral activity remain unclear. The review discusses the well-known RNases that possess established antiviral effects, including both intracellular RNases (RNase L, MCPIP1 protein, and eosinophil-associated RNases) and exogenous RNases (RNase A, BS-RNase, onconase, binase, and synthetic RNases). Attention is paid to two important, but not always obligatory, aspects of molecules of RNases that have antiviral properties, i.e., catalytic activity and ability to dimerize. The hypothetic scheme of virus elimination by exogenous RNases that reflects possible types of interaction of viruses and RNases with a cell is proposed. The evidence for RNases as classical components of immune defense and thus perspective agents for the development of new antiviral therapeutics is proposed.
Collapse
Affiliation(s)
- O. N. Ilinskaya
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, ul. Kremlevskaya 18, Kazan, 420008 Russia
| | - R. Shah Mahmud
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, ul. Kremlevskaya 18, Kazan, 420008 Russia
| |
Collapse
|
154
|
Jeltsch KM, Hu D, Brenner S, Zöller J, Heinz GA, Nagel D, Vogel KU, Rehage N, Warth SC, Edelmann SL, Gloury R, Martin N, Lohs C, Lech M, Stehklein JE, Geerlof A, Kremmer E, Weber A, Anders HJ, Schmitz I, Schmidt-Supprian M, Fu M, Holtmann H, Krappmann D, Ruland J, Kallies A, Heikenwalder M, Heissmeyer V. Cleavage of roquin and regnase-1 by the paracaspase MALT1 releases their cooperatively repressed targets to promote T(H)17 differentiation. Nat Immunol 2014; 15:1079-89. [PMID: 25282160 DOI: 10.1038/ni.3008] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 09/09/2014] [Indexed: 12/13/2022]
Abstract
Humoral autoimmunity paralleled by the accumulation of follicular helper T cells (T(FH) cells) is linked to mutation of the gene encoding the RNA-binding protein roquin-1. Here we found that T cells lacking roquin caused pathology in the lung and accumulated as cells of the T(H)17 subset of helper T cells in the lungs. Roquin inhibited T(H)17 cell differentiation and acted together with the endoribonuclease regnase-1 to repress target mRNA encoding the T(H)17 cell-promoting factors IL-6, ICOS, c-Rel, IRF4, IκBNS and IκBζ. This cooperation required binding of RNA by roquin and the nuclease activity of regnase-1. Upon recognition of antigen by the T cell antigen receptor (TCR), roquin and regnase-1 proteins were cleaved by the paracaspase MALT1. Thus, this pathway acts as a 'rheostat' by translating TCR signal strength via graded inactivation of post-transcriptional repressors and differential derepression of targets to enhance T(H)17 differentiation.
Collapse
Affiliation(s)
- Katharina M Jeltsch
- 1] Institute for Immunology, Ludwig-Maximilians-Universität München, Munich, Germany. [2] Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Desheng Hu
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Sven Brenner
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Jessica Zöller
- Institute for Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Gitta A Heinz
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Daniel Nagel
- Institute of Toxicology and Pharmacology, Research Unit Cellular Signal Integration, Helmholtz Zentrum München, Neuherberg, Germany
| | - Katharina U Vogel
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Nina Rehage
- 1] Institute for Immunology, Ludwig-Maximilians-Universität München, Munich, Germany. [2] Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Sebastian C Warth
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Stephanie L Edelmann
- 1] Institute for Immunology, Ludwig-Maximilians-Universität München, Munich, Germany. [2] Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Renee Gloury
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Nina Martin
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Claudia Lohs
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Maciej Lech
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jenny E Stehklein
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Achim Weber
- Institute for Surgical Pathology, University Hospital, Zürich, Switzerland
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ingo Schmitz
- 1] Helmholtz Centre for Infection Research, Braunschweig, Germany. [2] Institute for Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | | | - Mingui Fu
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Helmut Holtmann
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany
| | - Daniel Krappmann
- Institute of Toxicology and Pharmacology, Research Unit Cellular Signal Integration, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Axel Kallies
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Mathias Heikenwalder
- Institute for Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Vigo Heissmeyer
- 1] Institute for Immunology, Ludwig-Maximilians-Universität München, Munich, Germany. [2] Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
155
|
Fletcher CE, Dart DA, Bevan CL. Interplay between steroid signalling and microRNAs: implications for hormone-dependent cancers. Endocr Relat Cancer 2014; 21:R409-29. [PMID: 25062737 DOI: 10.1530/erc-14-0208] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hormones are key drivers of cancer development. To date, interest has largely been focussed on the classical model of hormonal gene regulation, but there is increasing evidence for a role of hormone signalling pathways in post-translational regulation of gene expression. In particular, a complex and dynamic network of bi-directional interactions with microRNAs (miRs) at all stages of biogenesis and during target gene repression is emerging. miRs, which act mainly by negatively regulating gene expression through association with 3'-UTRs of mRNA species, are increasingly understood to be important in development, normal physiology and pathogenesis. Given recent demonstrations of altered miR profiles in a diverse range of cancers, their ability to function as oncogenes or tumour suppressors, and hormonal regulation of miRs, understanding mechanisms by which miRs are generated and regulated is vitally important. miRs are transcribed by RNA polymerase II and then processed in the nucleus by the Drosha-containing Microprocessor complex and in the cytoplasm by Dicer, before mature miRs are incorporated into the RNA-induced silencing complex. It is increasingly evident that multiple cellular signalling pathways converge upon the miR biogenesis cascade, adding further layers of regulatory complexity to modulate miR maturation. This review summarises recent advances in identification of novel components and regulators of the Microprocessor and Dicer complexes, with particular emphasis on the role of hormone signalling pathways in regulating their activity. Understanding hormone regulation of miR production and how this is perturbed in cancer are critical for the development of miR-based therapeutics and biomarkers.
Collapse
Affiliation(s)
- Claire E Fletcher
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK
| | - D Alwyn Dart
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK
| | - Charlotte L Bevan
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK
| |
Collapse
|
156
|
Lin RJ, Chu JS, Chien HL, Tseng CH, Ko PC, Mei YY, Tang WC, Kao YT, Cheng HY, Liang YC, Lin SY. MCPIP1 suppresses hepatitis C virus replication and negatively regulates virus-induced proinflammatory cytokine responses. THE JOURNAL OF IMMUNOLOGY 2014; 193:4159-68. [PMID: 25225661 DOI: 10.4049/jimmunol.1400337] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human MCP-1-induced protein 1 (MCPIP1, also known as ZC3H12A and Regnase-1) plays important roles in negatively regulating the cellular inflammatory response. Recently, we found that as an RNase, MCPIP1 has broad-spectrum antiviral effects by targeting viral RNA. In this study, we demonstrated that MCPIP1 expression was induced by hepatitis C virus (HCV) infection in Huh7.5 hepatoma cells. MCPIP1 expression was higher in liver tissue from patients with chronic HCV infection compared with those without chronic HCV infection. Knockdown of MCPIP1 increased HCV replication and HCV-mediated expression of proinflammatory cytokines, such as TNF-α, IL-6, and MCP-1. However, overexpression of MCPIP1 significantly inhibited HCV replication and HCV-mediated expression of proinflammatory cytokines. Various mutants of functional domains of MCPIP1 showed disruption of the RNA binding and oligomerization abilities, as well as RNase activity, but not deubiquitinase activity, which impaired the inhibitory activity against HCV replication. On immunocytochemistry, MCPIP1 colocalized with HCV RNA. Use of a replication-defective HCV John Cunningham 1/AAG mutant and in vitro RNA cleavage assay demonstrated that MCPIP1 could directly degrade HCV RNA. MCPIP1 may suppress HCV replication and HCV-mediated proinflammatory responses with infection, which might contribute to the regulation of host defense against the infection and virus-induced inflammation.
Collapse
Affiliation(s)
- Ren-Jye Lin
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Jan-Show Chu
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Pathology, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Hsu-Ling Chien
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chung-Hsin Tseng
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pin-Chen Ko
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yung-Yu Mei
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Chun Tang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; and
| | - Yu-Ting Kao
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan; and
| | - Hui-Ying Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chih Liang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Shyr-Yi Lin
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan;
| |
Collapse
|
157
|
Xin H, Deng K, Fu M. Post-transcriptional gene regulation by RNA-binding proteins in vascular endothelial dysfunction. SCIENCE CHINA. LIFE SCIENCES 2014; 57:836-44. [PMID: 25104457 PMCID: PMC7089175 DOI: 10.1007/s11427-014-4703-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022]
Abstract
Endothelial cell dysfunction is a term which implies the dysregulation of normal endothelial cell functions, including impairment of the barrier functions, control of vascular tone, disturbance of proliferative and migratory capacity of endothelial cells, as well as control of leukocyte trafficking. Endothelial dysfunction is an early step in vascular inflammatory diseases such as atherosclerosis, diabetic vascular complications, sepsis-induced or severe virus infection-induced organ injuries. The expressions of inflammatory cytokines and vascular adhesion molecules induced by various stimuli, such as modified lipids, smoking, advanced glycation end products and bacteria toxin, significantly contribute to the development of endothelial dysfunction. The transcriptional regulation of inflammatory cytokines and vascular adhesion molecules has been well-studied. However, the regulation of those gene expressions at post-transcriptional level is emerging. RNA-binding proteins have emerged as critical regulators of gene expression acting predominantly at the post-transcriptional level in microRNA-dependent or independent manners. This review summarizes the latest insights into the roles of RNA-binding proteins in controlling vascular endothelial cell functions and their contribution to the pathogenesis of vascular inflammatory diseases.
Collapse
Affiliation(s)
- HongBo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| | - KeYu Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| | - MinGui Fu
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| |
Collapse
|
158
|
Abstract
MicroRNAs (miRNAs) are transcriptional and posttranscriptional regulators involved in nearly all known biological processes in distant eukaryotic clades. Their discovery and functional characterization have broadened our understanding of biological regulatory mechanisms in animals and plants. They show both evolutionary conserved and unique features across Metazoa. Here, we present the current status of the knowledge about the role of miRNA in development, growth, and physiology of teleost fishes, in comparison to other vertebrates. Infraclass Teleostei is the most abundant group among vertebrate lineage. Fish are an important component of aquatic ecosystems and human life, being the prolific source of animal proteins worldwide and a vertebrate model for biomedical research. We review miRNA biogenesis, regulation, modifications, and mechanisms of action. Specific sections are devoted to the role of miRNA in teleost development, organogenesis, tissue differentiation, growth, regeneration, reproduction, endocrine system, and responses to environmental stimuli. Each section discusses gaps in the current knowledge and pinpoints the future directions of research on miRNA in teleosts.
Collapse
Affiliation(s)
| | - Igor Babiak
- Faculty of Aquaculture and Biosciences, University of Nordland, Bodø, Norway
| |
Collapse
|
159
|
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that function as guide molecules in RNA silencing. Targeting most protein-coding transcripts, miRNAs are involved in nearly all developmental and pathological processes in animals. The biogenesis of miRNAs is under tight temporal and spatial control, and their dysregulation is associated with many human diseases, particularly cancer. In animals, miRNAs are ∼22 nucleotides in length, and they are produced by two RNase III proteins--Drosha and Dicer. miRNA biogenesis is regulated at multiple levels, including at the level of miRNA transcription; its processing by Drosha and Dicer in the nucleus and cytoplasm, respectively; its modification by RNA editing, RNA methylation, uridylation and adenylation; Argonaute loading; and RNA decay. Non-canonical pathways for miRNA biogenesis, including those that are independent of Drosha or Dicer, are also emerging.
Collapse
|
160
|
Vindry C, Vo Ngoc L, Kruys V, Gueydan C. RNA-binding protein-mediated post-transcriptional controls of gene expression: integration of molecular mechanisms at the 3' end of mRNAs? Biochem Pharmacol 2014; 89:431-40. [PMID: 24735612 DOI: 10.1016/j.bcp.2014.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 01/17/2023]
Abstract
Initially identified as an occasional and peculiar mode of gene regulation in eukaryotes, RNA-binding protein-mediated post-transcriptional control of gene expression has emerged, over the last two decades, as a major contributor in the control of gene expression. A large variety of RNA-binding proteins (RBPs) allows the recognition of very diverse messenger RNA sequences and participates in the regulation of basically all cellular processes. Nevertheless, the rapid outcome of post-transcriptional regulations on the level of gene expression has favored the expansion of this type of regulation in cellular processes prone to rapid and frequent modulations such as the control of the inflammatory response. At the molecular level, the 3'untranslated region (3'UTR) of mRNA is a favored site of RBP recruitment. RBPs binding to these regions control gene expression through two major modes of regulation, namely mRNA decay and modulation of translational activity. Recent progresses suggest that these two mechanisms are often interdependent and might result one from the other. Therefore, different RBPs binding distinct RNA subsets could share similar modes of action at the molecular level. RBPs are frequent targets of post-translational modifications, thereby disclosing numerous possibilities for pharmacological interventions. However, redundancies of the transduction pathways controlling these modifications have limited the perspectives to define RBPs as new therapeutic targets. Through the analysis of several examples of RBPs binding to 3'untranslated region of mRNA, we present here recent progress and perspectives regarding this rapidly evolving field of molecular biology.
Collapse
Affiliation(s)
- Caroline Vindry
- Laboratoire de Biologie moléculaire du gène, Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, 12 rue des Profs. Jeener et Brachet, Gosselies 6041, Belgium
| | - Long Vo Ngoc
- Laboratoire de Biologie moléculaire du gène, Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, 12 rue des Profs. Jeener et Brachet, Gosselies 6041, Belgium
| | - Véronique Kruys
- Laboratoire de Biologie moléculaire du gène, Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, 12 rue des Profs. Jeener et Brachet, Gosselies 6041, Belgium
| | - Cyril Gueydan
- Laboratoire de Biologie moléculaire du gène, Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, 12 rue des Profs. Jeener et Brachet, Gosselies 6041, Belgium.
| |
Collapse
|
161
|
Van Wynsberghe PM, Finnegan EF, Stark T, Angelus EP, Homan KE, Yeo GW, Pasquinelli AE. The Period protein homolog LIN-42 negatively regulates microRNA biogenesis in C. elegans. Dev Biol 2014; 390:126-35. [PMID: 24699545 DOI: 10.1016/j.ydbio.2014.03.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 03/21/2014] [Accepted: 03/25/2014] [Indexed: 11/18/2022]
Abstract
MicroRNAs (miRNAs) are small RNAs that post-transcriptionally regulate gene expression in many multicellular organisms. They are encoded in the genome and transcribed into primary (pri-) miRNAs before two processing steps that ultimately produce the mature miRNA. In order to generate the appropriate amount of a particular miRNA in the correct location at the correct time, proper regulation of miRNA biogenesis is essential. Here we identify the Period protein homolog LIN-42 as a new regulator of miRNA biogenesis in Caenorhabditis elegans. We mapped a spontaneous suppressor of the normally lethal let-7(n2853) allele to the lin-42 gene. Mutations in this allele (ap201) or a second lin-42 allele (n1089) caused increased mature let-7 miRNA levels at most time points when mature let-7 miRNA is normally expressed. Levels of pri-let-7 and a let-7 transcriptional reporter were also increased in lin-42(n1089) worms. These results indicate that LIN-42 normally represses pri-let-7 transcription and thus the accumulation of let-7 miRNA. This inhibition is not specific to let-7, as pri- and mature levels of lin-4 and miR-35 were also increased in lin-42 mutants. Furthermore, small RNA-seq analysis showed widespread increases in the levels of mature miRNAs in lin-42 mutants. Thus, we propose that the period protein homolog LIN-42 is a global regulator of miRNA biogenesis.
Collapse
Affiliation(s)
- Priscilla M Van Wynsberghe
- Division of Biology, University of California at San Diego, La Jolla, CA 92093-0349, USA; Department of Biology, Colgate University, Hamilton, NY 13323, USA.
| | - Emily F Finnegan
- Division of Biology, University of California at San Diego, La Jolla, CA 92093-0349, USA
| | - Thomas Stark
- Division of Biology, University of California at San Diego, La Jolla, CA 92093-0349, USA; Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, Stem Cell Program, University of California at San Diego, Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA
| | - Evan P Angelus
- Department of Biology, Colgate University, Hamilton, NY 13323, USA
| | - Kathryn E Homan
- Department of Biology, Colgate University, Hamilton, NY 13323, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, Stem Cell Program, University of California at San Diego, Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA
| | - Amy E Pasquinelli
- Division of Biology, University of California at San Diego, La Jolla, CA 92093-0349, USA.
| |
Collapse
|
162
|
Sierra-Filardi E, Nieto C, Domínguez-Soto A, Barroso R, Sánchez-Mateos P, Puig-Kroger A, López-Bravo M, Joven J, Ardavín C, Rodríguez-Fernández JL, Sánchez-Torres C, Mellado M, Corbí AL. CCL2 shapes macrophage polarization by GM-CSF and M-CSF: identification of CCL2/CCR2-dependent gene expression profile. THE JOURNAL OF IMMUNOLOGY 2014; 192:3858-67. [PMID: 24639350 DOI: 10.4049/jimmunol.1302821] [Citation(s) in RCA: 376] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The CCL2 chemokine mediates monocyte egress from bone marrow and recruitment into inflamed tissues through interaction with the CCR2 chemokine receptor, and its expression is upregulated by proinflammatory cytokines. Analysis of the gene expression profile in GM-CSF- and M-CSF-polarized macrophages revealed that a high CCL2 expression characterizes macrophages generated under the influence of M-CSF, whereas CCR2 is expressed only by GM-CSF-polarized macrophages. Analysis of the factors responsible for this differential expression identified activin A as a critical factor controlling the expression of the CCL2/CCR2 pair in macrophages, as activin A increased CCR2 expression but inhibited the acquisition of CCL2 expression by M-CSF-polarized macrophages. CCL2 and CCR2 were found to determine the extent of macrophage polarization because CCL2 enhances the LPS-induced production of IL-10, whereas CCL2 blockade leads to enhanced expression of M1 polarization-associated genes and cytokines, and diminished expression of M2-associated markers in human macrophages. Along the same line, Ccr2-deficient bone marrow-derived murine macrophages displayed an M1-skewed polarization profile at the transcriptomic level and exhibited a significantly higher expression of proinflammatory cytokines (TNF-α, IL-6) in response to LPS. Therefore, the CCL2-CCR2 axis regulates macrophage polarization by influencing the expression of functionally relevant and polarization-associated genes and downmodulating proinflammatory cytokine production.
Collapse
Affiliation(s)
- Elena Sierra-Filardi
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
SKALNIAK ANNA, BORATYN ELŻBIETA, TYRKALSKA SYLWIAD, HORWACIK IRENA, DURBAS MAŁGORZATA, ŁASTOWSKA MARIA, JURA JOLANTA, ROKITA HANNA. Expression of the monocyte chemotactic protein-1-induced protein 1 decreases human neuroblastoma cell survival. Oncol Rep 2014; 31:2385-92. [DOI: 10.3892/or.2014.3076] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/13/2014] [Indexed: 11/05/2022] Open
|
164
|
Garcia-Bonilla L, Benakis C, Moore J, Iadecola C, Anrather J. Immune mechanisms in cerebral ischemic tolerance. Front Neurosci 2014; 8:44. [PMID: 24624056 PMCID: PMC3940969 DOI: 10.3389/fnins.2014.00044] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 02/17/2014] [Indexed: 12/21/2022] Open
Abstract
Stressor-induced tolerance is a central mechanism in the response of bacteria, plants, and animals to potentially harmful environmental challenges. This response is characterized by immediate changes in cellular metabolism and by the delayed transcriptional activation or inhibition of genetic programs that are not generally stressor specific (cross-tolerance). These programs are aimed at countering the deleterious effects of the stressor. While induction of this response (preconditioning) can be established at the cellular level, activation of systemic networks is essential for the protection to occur throughout the organs of the body. This is best signified by the phenomenon of remote ischemic preconditioning, whereby application of ischemic stress to one tissue or organ induces ischemic tolerance (IT) in remote organs through humoral, cellular and neural signaling. The immune system is an essential component in cerebral IT acting simultaneously both as mediator and target. This dichotomy is based on the fact that activation of inflammatory pathways is necessary to establish IT and that IT can be, in part, attributed to a subdued immune activation after index ischemia. Here we describe the components of the immune system required for induction of IT and review the mechanisms by which a reprogrammed immune response contributes to the neuroprotection observed after preconditioning. Learning how local and systemic immune factors participate in endogenous neuroprotection could lead to the development of new stroke therapies.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Corinne Benakis
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Jamie Moore
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Costantino Iadecola
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Josef Anrather
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| |
Collapse
|
165
|
Schwarzenbach H, Nishida N, Calin GA, Pantel K. Clinical relevance of circulating cell-free microRNAs in cancer. Nat Rev Clin Oncol 2014; 11:145-56. [PMID: 24492836 DOI: 10.1038/nrclinonc.2014.5] [Citation(s) in RCA: 821] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Efficient patient management relies on early diagnosis of disease and monitoring of treatment. In this regard, much effort has been made to find informative, blood-based biomarkers for patients with cancer. Owing to their attributes-which are specifically modulated by the tumour-circulating cell-free microRNAs found in the peripheral blood of patients with cancer may provide insights into the biology of the tumour and the effects of therapeutic interventions. Moreover, the role of microRNAs in the regulation of different cellular processes points to their clinical utility as blood-based biomarkers and future therapeutic targets. MicroRNAs are optimal biomarkers owing to high stability under storage and handling conditions and their presence in blood, urine and other body fluids. In particular, detection of levels of microRNAs in blood plasma and serum has the potential for an earlier cancer diagnosis and to predict prognosis and response to therapy. This Review article considers the latest developments in the use of circulating microRNAs as prognostic and predictive biomarkers and discusses their utility in personalized medicine.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Department of Tumour Biology, Centre of Experimental Medicine, University Cancer Centre, University Medical Centre Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | - Naohiro Nishida
- Department of Experimental Therapeutics and the Centre for RNA Interference and Non-Coding RNAs, The University of Texas, MD Anderson Cancer Center, South Campus Research Building 3, 1881 East Road, Houston, TX 77030, USA
| | - George A Calin
- Department of Experimental Therapeutics and the Centre for RNA Interference and Non-Coding RNAs, The University of Texas, MD Anderson Cancer Center, South Campus Research Building 3, 1881 East Road, Houston, TX 77030, USA
| | - Klaus Pantel
- Department of Tumour Biology, Centre of Experimental Medicine, University Cancer Centre, University Medical Centre Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| |
Collapse
|
166
|
Abstract
Epigenetics is one of the most rapidly developing fields of biological research. Breakthroughs in several technologies have enabled the possibility of genome-wide epigenetic research, for example the mapping of human genome-wide DNA methylation. In addition, with the development of various high-throughput and high-resolution sequencing technologies, a large number of functional noncoding RNAs have been identified. Massive studies indicated that these functional ncRNA also play an important role in epigenetics. In this review, we gain inspiration from the recent proposal of the ceRNAs hypothesis. This hypothesis proposes that miRNAs act as a language of communication. Accordingly, we further deduce that all of epigenetics may functionally acquire such a unique language characteristic. In summary, various epigenetic markers may not only participate in regulating cellular processes, but they may also act as the intracellular ‘language’ of communication and are involved in extensive information exchanges within cell.
Collapse
Affiliation(s)
- Biao Huang
- Research Center of Basic Medical Science, Department of Immunology, Basic Medical College, Tianjin Key Laboratory of Cellular & Molecular Immunology, Key Laboratory of Immune Microenvironments & Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Cizhong Jiang
- Department of Bioinformatics, Shanghai Key Laboratory of Signaling & Disease Research, The School of Life Sciences & Technology, Tongji University, Shanghai, China
| | - Rongxin Zhang
- Research Center of Basic Medical Science, Department of Immunology, Basic Medical College, Tianjin Key Laboratory of Cellular & Molecular Immunology, Key Laboratory of Immune Microenvironments & Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
167
|
Podshivalova K, Salomon DR. MicroRNA regulation of T-lymphocyte immunity: modulation of molecular networks responsible for T-cell activation, differentiation, and development. Crit Rev Immunol 2014; 33:435-76. [PMID: 24099302 DOI: 10.1615/critrevimmunol.2013006858] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNA) are a class of small non-coding RNAs that constitute an essential and evolutionarily conserved mechanism for post-transcriptional gene regulation. Multiple miRNAs have been described to play key roles in T-lymphocyte development, differentiation, and function. In this review, we highlight the current literature regarding the differential expression of miRNAs in various models of murine and human T-cell biology. We emphasize mechanistic understandings of miRNA regulation of thymocyte development, T-cell activation, and differentiation into effector and memory subsets. We describe the participation of miRNAs in complex regulatory circuits shaping T-cell proteomes in a context-dependent manner. It is striking that some miRNAs regulate multiple processes, while others only appear in limited functional contexts. It is also evident that the expression and function of specific miRNAs can differ between murine and human systems. Ultimately, it is not always correct to simplify the complex events of T-cell biology into a model driven by only one or two master regulator miRNAs. In reality, T-cell activation and differentiation involve the expression of multiple miRNAs with many mRNA targets; thus, the true extent of miRNA regulation of T-cell biology is likely far more vast than currently appreciated.
Collapse
Affiliation(s)
- Katie Podshivalova
- Laboratory for Functional Genomics, Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | | |
Collapse
|
168
|
Minagawa K, Wakahashi K, Kawano H, Nishikawa S, Fukui C, Kawano Y, Asada N, Sato M, Sada A, Katayama Y, Matsui T. Posttranscriptional modulation of cytokine production in T cells for the regulation of excessive inflammation by TFL. THE JOURNAL OF IMMUNOLOGY 2014; 192:1512-24. [PMID: 24415781 DOI: 10.4049/jimmunol.1301619] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Posttranscriptional machinery regulates inflammation and is associated with autoimmunity as well as tumorigenesis in collaboration with transcription factors. We previously identified the tumor suppressor gene transformed follicular lymphoma (TFL) on 6q25 in a patient with follicular lymphoma, which transformed into diffuse large B cell lymphoma. TFL families have a common RNase domain that governs macrophage-mediated inflammation. In human peripheral blood, TFL is dominantly expressed at the glycine- and tryptophan-rich cytoplasmic processing bodies of T lymphocytes, and it is persistently upregulated in activated T cells. To address its physiological role, we established TFL(-/-) mice in which TFL(-/-) lymphocytes proliferated more rapidly than TFL(+/+) upon stimulation with inappropriate cytokine secretion, including IL-2, IL-6, and IL-10. Moreover, TFL inhibited the synthesis of cytokines such as IL-2, IL-6, IL-10, TNF-α, and IL-17a by 3' untranslated region RNA degradation. Experimental autoimmune encephalitis induced in TFL(-/-) mice demonstrated persistent severe paralysis. CNS-infiltrated CD4(+) T cells in TFL(-/-) mice contained a higher proportion of Th17 cells than did those in TFL(+/+) mice during the resolution phase, and IL-17a mRNA levels were markedly increased in TFL(-/-) cells. These results suggest that TFL may play an important role in attenuating local inflammation by suppressing the infiltration of Th17 cells in the CNS during the resolution phase of experimental autoimmune encephalitis. TFL is a novel gradual and persistent posttranscriptional regulator, and the TFL-driven attenuation of excessive inflammation could contribute to recovery from T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Kentaro Minagawa
- Division of Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Lipert B, Wegrzyn P, Sell H, Eckel J, Winiarski M, Budzynski A, Matlok M, Kotlinowski J, Ramage L, Malecki M, Wilk W, Mitus J, Jura J. Monocyte chemoattractant protein-induced protein 1 impairs adipogenesis in 3T3-L1 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:780-8. [PMID: 24418043 DOI: 10.1016/j.bbamcr.2014.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/13/2013] [Accepted: 01/01/2014] [Indexed: 12/22/2022]
Abstract
Monocyte chemoattractant protein-induced protein 1 (MCPIP1) encoded by the ZC3H12a gene (also known as Regnase-1) is involved in the regulation of degradation of mRNA of inflammatory modulators and for processing of pre-miRNA. These functions depend on the presence of the PIN domain. Moreover, MCPIP1 was described as a negative regulator of NF-κB and AP-1 signaling pathways although mechanisms underlying such activity remain unknown. We aimed at determining the role of MCPIP1 in adipogenesis. Here, we present evidence that Mcpip1 transcription is transiently activated during 3T3-L1 transition from pre- to adipocytes. However Mcpip1 protein expression is also strongly decreased at day one after induction of adipogenesis. Knockdown of Mcpip1 results in an upregulation of C/EBPβ and PPARγ mRNAs, whereas overexpression of MCPIP1 reduces the level of both transcription factors and impairs adipogenesis. MCPIP1-dependend modulation of C/EBPβ and PPARγ levels results in a modulation of the expression of downstream controlled genes. In addition, decreased C/EBPβ, but not PPARγ, depends on the activity of the MCPIP1 PIN domain, which is responsible for RNase properties of this protein. Together, these data confirm that MCPIP1 is a key regulator of adipogenesis.
Collapse
Affiliation(s)
- Barbara Lipert
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Paulina Wegrzyn
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Henrike Sell
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Juergen Eckel
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Düsseldorf, Germany
| | - Marek Winiarski
- 2nd Department of General Surgery, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Andrzej Budzynski
- 2nd Department of General Surgery, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Maciej Matlok
- 2nd Department of General Surgery, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Jerzy Kotlinowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Lindsay Ramage
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Malecki
- Department of Metabolic Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Waclaw Wilk
- The Maria Skłodowska-Curie Institute of Oncology, Krakow, Poland
| | - Jerzy Mitus
- The Maria Skłodowska-Curie Institute of Oncology, Krakow, Poland
| | - Jolanta Jura
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
170
|
Yang K, Wu M, Li M, Li D, Peng A, Nie X, Sun M, Wang J, Wu Y, Deng Q, Zhu M, Chen K, Yuan J, Huang X. miR-155 Suppresses Bacterial Clearance in Pseudomonas aeruginosa–Induced Keratitis by Targeting Rheb. J Infect Dis 2014; 210:89-98. [DOI: 10.1093/infdis/jiu002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Kun Yang
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Minhao Wu
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Meiyu Li
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Dandan Li
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Anping Peng
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Xinxin Nie
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Mingxia Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jinli Wang
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Yongjian Wu
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Qiuchan Deng
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Min Zhu
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Kang Chen
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xi Huang
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
171
|
Identification and characterization of FGF2-dependent mRNA: microRNA networks during lens fiber cell differentiation. G3-GENES GENOMES GENETICS 2013; 3:2239-55. [PMID: 24142921 PMCID: PMC3852386 DOI: 10.1534/g3.113.008698] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) and fibroblast growth factor (FGF) signaling regulate a wide range of cellular functions, including cell specification, proliferation, migration, differentiation, and survival. In lens, both these systems control lens fiber cell differentiation; however, a possible link between these processes remains to be examined. Herein, the functional requirement for miRNAs in differentiating lens fiber cells was demonstrated via conditional inactivation of Dicer1 in mouse (Mus musculus) lens. To dissect the miRNA-dependent pathways during lens differentiation, we used a rat (Rattus norvegicus) lens epithelial explant system, induced by FGF2 to differentiate, followed by mRNA and miRNA expression profiling. Transcriptome and miRNome analysis identified extensive FGF2-regulated cellular responses that were both independent and dependent on miRNAs. We identified 131 FGF2-regulated miRNAs. Seventy-six of these miRNAs had at least two in silico predicted and inversely regulated target mRNAs. Genes modulated by the greatest number of FGF-regulated miRNAs include DNA-binding transcription factors Nfib, Nfat5/OREBP, c-Maf, Ets1, and N-Myc. Activated FGF signaling influenced bone morphogenetic factor/transforming growth factor-β, Notch, and Wnt signaling cascades implicated earlier in lens differentiation. Specific miRNA:mRNA interaction networks were predicted for c-Maf, N-Myc, and Nfib (DNA-binding transcription factors); Cnot6, Cpsf6, Dicer1, and Tnrc6b (RNA to miRNA processing); and Ash1l, Med1/PBP, and Kdm5b/Jarid1b/Plu1 (chromatin remodeling). Three miRNAs, including miR-143, miR-155, and miR-301a, down-regulated expression of c-Maf in the 3′-UTR luciferase reporter assays. These present studies demonstrate for the first time global impact of activated FGF signaling in lens cell culture system and predicted novel gene regulatory networks connected by multiple miRNAs that regulate lens differentiation.
Collapse
|
172
|
Abstract
Regulating the expression of individual miRNAs (microRNAs) is important for cell development and function. The up- or down-regulation of the processing of specific miRNA precursors to the mature active form represents one tool to control miRNA concentration and is mediated by proteins that recognize the terminal loop of the RNA precursors. Terminal loop recognition is achieved by the combined action of several RNA-binding domains. The proteins can then regulate the processing by recruiting RNA enzymes, changing the RNA structure and preventing or enhancing the accessibility and processing activity of the core processing complexes. The present review focuses on how terminal loop-binding proteins recognize their RNA targets and mediate their regulatory function(s), and highlights how terminal loop-mediated regulation relates to the broader regulation of mRNA metabolism.
Collapse
|
173
|
Niu J, Shi Y, Xue J, Miao R, Huang S, Wang T, Wu J, Fu M, Wu ZH. USP10 inhibits genotoxic NF-κB activation by MCPIP1-facilitated deubiquitination of NEMO. EMBO J 2013; 32:3206-19. [PMID: 24270572 DOI: 10.1038/emboj.2013.247] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 10/31/2013] [Indexed: 01/14/2023] Open
Abstract
DNA damage-induced activation of the transcription factor NF-κB plays an important role in the cellular response to genotoxic stress. However, uncontrolled NF-κB activation upon DNA damage may lead to deleterious consequences. Although the mechanisms mediating genotoxic NF-κB activation have been elucidated, how this signalling is terminated remains poorly understood. Here, we show that the CCCH-type zinc finger-containing protein MCPIP1 (monocyte chemotactic protein-1-induced protein-1; also known as ZC3H12A) is induced upon genotoxic treatment in an NF-κB-dependent manner. MCPIP1 upregulation reduces NEMO linear ubiquitylation, resulting in decreased activation of IKK and NF-κB. NEMO ubiquitylation is decreased through the deubiquitinase USP10, which interacts with NEMO via MCPIP1 upon genotoxic stress. USP10 association with NEMO leads to removal of NEMO-attached linear polyubiquitin chains and subsequent inhibition of the genotoxic NF-κB signalling cascade. Consistently, USP10 is required for MCPIP1-mediated inhibition of genotoxic NF-κB activation and promotion of apoptosis. Thus, by mediating USP10-dependent deubiquitination of NEMO, MCPIP1 induction serves as a negative feedback mechanism for attenuating genotoxic NF-κB activation.
Collapse
Affiliation(s)
- Jixiao Niu
- 1] Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, USA [2] Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Yu F, Du F, Wang Y, Huang S, Miao R, Major AS, Murphy EA, Fu M, Fan D. Bone marrow deficiency of MCPIP1 results in severe multi-organ inflammation but diminishes atherogenesis in hyperlipidemic mice. PLoS One 2013; 8:e80089. [PMID: 24223214 PMCID: PMC3819309 DOI: 10.1371/journal.pone.0080089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/08/2013] [Indexed: 11/20/2022] Open
Abstract
Objective MCPIP1 is a newly identified protein that profoundly impacts immunity and inflammation. We aim to test if MCPIP1 deficiency in hematopoietic cells results in systemic inflammation and accelerates atherogenesis in mice. Approach and Results After lethally irradiated, LDLR−/− mice were transplanted with bone marrow cells from either wild-type or MCPIP1−/− mice. These chimeric mice were fed a western-type diet for 7 weeks. We found that bone marrow MCPIP1−/− mice displayed a phenotype similar to that of whole body MCPIP1−/− mice, with severe systemic and multi-organ inflammation. However, MCPIP1−/− bone marrow recipients developed >10-fold less atherosclerotic lesions in the proximal aorta than WT bone marrow recipients, and essentially no lesions in en face aorta. The diminishment in atherosclerosis in bone marrow MCPIP1−/− mice may be partially attributed to the slight decrease in their plasma lipids. Flow cytometric analysis of splenocytes showed that bone marrow MCPIP1−/− mice contained reduced numbers of T cells and B cells, but increased numbers of regulatory T cells, Th17 cells, CD11b+/Gr1+ cells and CD11b+/Ly6Clow cells. This overall anti-atherogenic leukocyte profile may also contribute to the reduced atherogenesis. We also examined the cholesterol efflux capability of MCPIP1 deficient macrophages, and found that MCPIP1deficiency increased cholesterol efflux to apoAI and HDL, due to increased protein levels of ABCA1 and ABCG1. Conclusions Hematopoietic deficiency of MCPIP1 resulted in severe systemic and multi-organ inflammation but paradoxically diminished atherogenesis in mice. The reduced atheroegensis may be explained by the decreased plasma cholesterol levels, the anti-atherogenic leukocyte profile, as well as enhanced cholesterol efflux capability. This study suggests that, while atherosclerosis is a chronic inflammatory disease, the mechanisms underlying atherogenesis-associated inflammation in arterial wall versus the inflammation in solid organs may be substantially different.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nutrition and Food Hygiene, the Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Fen Du
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- Department of Biochemistry and molecular Biology, School of Basic Medicine, Wuhan University, Wuhan, China
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Shengping Huang
- Shock/Trauma Research Center & Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, United States of America
| | - Ruidong Miao
- Shock/Trauma Research Center & Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, United States of America
| | - Amy S. Major
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United State of America
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Mingui Fu
- Shock/Trauma Research Center & Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, United States of America
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
175
|
MCPIP1 restricts HIV infection and is rapidly degraded in activated CD4+ T cells. Proc Natl Acad Sci U S A 2013; 110:19083-8. [PMID: 24191027 DOI: 10.1073/pnas.1316208110] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
HIV-1 primarily infects activated CD4+ T cells and macrophages. Quiescent CD4+ T cells, however, possess cellular factors that limit HIV-1 infection at different postentry steps of the viral life cycle. Here, we show that the previously reported immune regulator monocyte chemotactic protein-induced protein 1 (MCPIP1) restricts HIV-1 production in CD4+ T cells. While the ectopic expression of MCPIP1 in cell lines abolished the production of HIV-1, silencing of MCPIP1 enhanced HIV-1 production. Subsequent analysis indicated that MCPIP1 imposes its restriction by decreasing the steady levels of viral mRNA species through its RNase domain. Remarkably, common T-cell stimuli induced the rapid degradation of MCPIP1 in both T-cell lines and quiescent human CD4+ T cells. Lastly, blocking the proteosomal degradation of MCPIP1 by MG132 abrogated HIV-1 production in phorbol 12-myristate 13-acetate/ionomycin-stimulated human CD4+ T cells isolated from healthy donors. Overall, MCPIP1 poses a potent barrier against HIV-1 infection at a posttranscriptional stage. Although the observed HIV restriction conferred by MCPIP1 does not seem to be overcome by any viral protein, it is removed during cellular stimulation. These findings provide insights into the mechanisms of cellular activation-mediated HIV-1 production in CD4+ T cells.
Collapse
|
176
|
MicroRNA-155 promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb. PLoS Pathog 2013; 9:e1003697. [PMID: 24130493 PMCID: PMC3795043 DOI: 10.1371/journal.ppat.1003697] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 08/26/2013] [Indexed: 02/07/2023] Open
Abstract
Mycobacterium tuberculosis is a hard-to-eradicate intracellular pathogen that infects one-third of the global population. It can live within macrophages owning to its ability to arrest phagolysosome biogenesis. Autophagy has recently been identified as an effective way to control the intracellular mycobacteria by enhancing phagosome maturation. In the present study, we demonstrate a novel role of miR-155 in regulating the autophagy-mediated anti-mycobacterial response. Both in vivo and in vitro studies showed that miR-155 expression was significantly enhanced after mycobacterial infection. Forced expression of miR-155 accelerated the autophagic response in macrophages, thus promoting the maturation of mycobacterial phagosomes and decreasing the survival rate of intracellular mycobacteria, while transfection with miR-155 inhibitor increased mycobacterial survival. However, macrophage-mediated mycobacterial phagocytosis was not affected after miR-155 overexpression or inhibition. Furthermore, blocking autophagy with specific inhibitor 3-methyladenine or silencing of autophagy related gene 7 (Atg7) reduced the ability of miR-155 to promote autophagy and mycobacterial elimination. More importantly, our study demonstrated that miR-155 bound to the 3′-untranslated region of Ras homologue enriched in brain (Rheb), a negative regulator of autophagy, accelerated the process of autophagy and sequential killing of intracellular mycobacteria by suppressing Rheb expression. Our results reveal a novel role of miR-155 in regulating autophagy-mediated mycobacterial elimination by targeting Rheb, and provide potential targets for clinical treatment. microRNA-155 (miR-155) plays an essential role in regulating the host immune response by post-transcriptionally repressing the expression of target genes. However, little is known regarding its activity in modulating autophagy, an important host defense mechanism against intracellular bacterial infection. Mycobacterium tuberculosis is a hard-to-eradicate intracellular pathogen that infects approximately one-third of the global population, and causes 1.5 million deaths annually. The present study explores a novel role of miR-155 in the host response against mycobacterial infection. Our data demonstrates that mycobacterial infection triggers the expression of miR-155, and the induction of miR-155 in turn activates autophagy by targeting Rheb, a negative regulator of autophagy. miR-155-promoted autophagy accelerates the maturation of the mycobacterial phagosome, thus decreasing the survival of intracellular mycobacteria in macrophages. These findings contribute to a better understanding of the host defense mechanisms against mycobacterial infection, providing useful information for development of potential therapeutic interventions against tuberculosis.
Collapse
|
177
|
Shibuta T, Honda E, Shiotsu H, Tanaka Y, Vellasamy S, Shiratsuchi M, Umemura T. Imatinib induces demethylation of miR-203 gene: an epigenetic mechanism of anti-tumor effect of imatinib. Leuk Res 2013; 37:1278-86. [PMID: 23953880 DOI: 10.1016/j.leukres.2013.07.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 07/18/2013] [Indexed: 11/24/2022]
Abstract
MicroRNA (miRNA) is an important regulator of cellular proliferation, differentiation and death. Leukemia-specific signature of miRNAs suggests that epigenetic dysregulation of miRNAs is important for leukemogenesis. We focused on the role of DNA methylation of miR-203 which targets BCR-ABL1 mRNA. The microarray analysis showed that 48 miRNAs of CpG-rich 212 miRNAs were upregulated over 2-fold after imatinib treatment. Imatinib induced the demethylation of the miR-203 promoter region, resulting in low expression of targeted BCR-ABL1 gene, and loss of proliferation of leukemic cells. In conclusion, demethylation of miR-203 is one of the molecular mechanisms of imatinib-induced inhibition of BCR-ABL1-positive leukemic cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Benzamides/pharmacology
- Cell Line, Tumor
- CpG Islands
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methylation/drug effects
- Epigenesis, Genetic/drug effects
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Piperazines/pharmacology
- Promoter Regions, Genetic
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Tatsuki Shibuta
- Department of Health Sciences, Faculty of Medical Sciences, Graduate School of Medical Sciences, Kyushu University, Japan
| | | | | | | | | | | | | |
Collapse
|
178
|
Cellular localization and processing of primary transcripts of exonic microRNAs. PLoS One 2013; 8:e76647. [PMID: 24073292 PMCID: PMC3779153 DOI: 10.1371/journal.pone.0076647] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 08/30/2013] [Indexed: 12/31/2022] Open
Abstract
Processing of miRNAs occurs simultaneous with the transcription and splicing of their primary transcripts. For the small subset of exonic miRNAs it is unclear if the unspliced and/or spliced transcripts are used for miRNA biogenesis. We assessed endogenous levels and cellular location of primary transcripts of three exonic miRNAs. The ratio between unspliced and spliced transcripts varied markedly, i.e. >1 for BIC, <1 for pri-miR-146a and variable for pri-miR-22. Endogenous unspliced transcripts were located almost exclusively in the nucleus and thus available for miRNA processing for all three miRNAs. Endogenous spliced pri-miRNA transcripts were present both in the nucleus and in the cytoplasm and thus only partly available for miRNA processing. Overexpression of constructs containing the 5’ upstream exonic or intronic sequence flanking pre-miR-155 resulted in strongly enhanced miR-155 levels, indicating that the flanking sequence does not affect processing efficiency. Exogenously overexpressed full-length spliced BIC transcripts were present both in the nucleus and in the cytoplasm, were bound by the Microprocessor complex and resulted in enhanced miR-155 levels. We conclude that both unspliced and spliced transcripts of exonic miRNAs can be used for pre-miRNA cleavage. Splicing and cytoplasmic transport of spliced transcripts may present a mechanism to regulate levels of exonic microRNAs.
Collapse
|
179
|
Roy A, Zhang M, Saad Y, Kolattukudy PE. Antidicer RNAse activity of monocyte chemotactic protein-induced protein-1 is critical for inducing angiogenesis. Am J Physiol Cell Physiol 2013; 305:C1021-32. [PMID: 24048733 DOI: 10.1152/ajpcell.00203.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Inflammatory angiogenesis involves the induction of a novel gene ZC3H12A encoding monocyte chemoattractant protein-1 (MCP-1)-induced protein-1 (MCPIP1) that has deubiquitinase and antidicer RNAse activities. If and how these enzymatic activities of MCPIP1 mediate the biological functions of MCPIP1 are unknown. Present studies with human umbilical vein endothelial cells suggest that MCPIP-induced angiogenesis is mediated via hypoxia-inducible factor (HIF-1α), vascular endothelial growth factor (VEGF), and silent information regulator (SIRT-1) induction that results in the inhibition of angiogenesis inhibitor thrombospondin-1. MCPIP1 expression inhibited the production of the antiangiogenic microRNA (miR)-20b and -34a that repress the translation of HIF-1α and SIRT-1, respectively. The RNase-dead MCPIP mutant D141N not only did not induce angiogenesis but also failed to inhibit the production of miR-20b and -34a suggesting that the antidicer RNase activity of MCPIP1 is involved in MCPIP-mediated angiogenesis. Mimetics of miR-20b and -34a inhibited MCPIP1-induced angiogenesis confirming that MCPIP1 suppresses the biogenesis of miR-20b and -34a. Furthermore, our results indicate that MCPIP expression induces nuclear translocation of HIF-1α. We show that under hypoxia angiogenesis is mediated via induction of MCPIP1 and under normoxia, in vitro, MCPIP deubiquitinates ubiquitinated HIF-1α and the stabilized HIF-1α enters the nucleus to promote the transcription of its target genes, cyclooxygenase-2 and VEGF, suggesting that the deubiquitinase activity of MCPIP may also promote angiogenesis. The present results show for the first time that the antidicer RNase activity of MCPIP1 is critical in mediating a biological function of MCPIP, namely angiogenesis.
Collapse
Affiliation(s)
- Arpita Roy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | | | | | | |
Collapse
|
180
|
Kato M, Castro NE, Natarajan R. MicroRNAs: potential mediators and biomarkers of diabetic complications. Free Radic Biol Med 2013; 64:85-94. [PMID: 23770198 PMCID: PMC3762900 DOI: 10.1016/j.freeradbiomed.2013.06.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 06/04/2013] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
Abstract
The incidence of diabetes is escalating worldwide and, consequently, this has become a major health care problem. Moreover, both type 1 and type 2 diabetes are associated with significantly accelerated rates of microvascular complications, including retinopathy, nephropathy, and neuropathy, as well as macrovascular complications such as atherosclerotic cardiovascular and hypertensive diseases. Key factors have been implicated in leading to these complications, including hyperglycemia, insulin resistance, dyslipidemia, advanced glycation end products, growth factors, inflammatory cytokines/chemokines, and related increases in cellular oxidant stress (including mitochondrial) and endoplasmic reticulum stress. However, the molecular mechanisms underlying the high incidence of diabetic complications, which often progress despite glycemic control, are still not fully understood. MicroRNAs (miRNAs) are short noncoding RNAs that have elicited immense interest in recent years. They repress target gene expression via posttranscriptional mechanisms and have diverse cellular and biological functions. Herein, we discuss the role of miRNAs in the pathobiology of various diabetic complications, their involvement in oxidant stress, and also the potential use of differentially expressed miRNAs as novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Mitsuo Kato
- Department of Diabetes, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | | | | |
Collapse
|
181
|
Wan G, Zhang X, Langley RR, Liu Y, Hu X, Han C, Peng G, Ellis LM, Jones SN, Lu X. DNA-damage-induced nuclear export of precursor microRNAs is regulated by the ATM-AKT pathway. Cell Rep 2013; 3:2100-12. [PMID: 23791529 DOI: 10.1016/j.celrep.2013.05.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 04/25/2013] [Accepted: 05/23/2013] [Indexed: 12/13/2022] Open
Abstract
Expression of microRNAs (miRNAs) involves transcription of miRNA genes and maturation of the primary transcripts. Recent studies have shown that posttranscriptional processing of primary and precursor miRNAs is induced after DNA damage through regulatory RNA-binding proteins in the Drosha and Dicer complexes, such as DDX5 and KSRP. However, little is known about the regulation of nuclear export of pre-miRNAs in the DNA-damage response, a critical step in miRNA maturation. Here, we show that nuclear export of pre-miRNAs is accelerated after DNA damage in an ATM-dependent manner. The ATM-activated AKT kinase phosphorylates Nup153, a key component of the nucleopore, leading to enhanced interaction between Nup153 and Exportin-5 (XPO5) and increased nuclear export of pre-miRNAs. These findings define an important role of DNA-damage signaling in miRNA transport and maturation.
Collapse
Affiliation(s)
- Guohui Wan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Uehata T, Akira S. mRNA degradation by the endoribonuclease Regnase-1/ZC3H12a/MCPIP-1. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:708-13. [DOI: 10.1016/j.bbagrm.2013.03.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/04/2013] [Accepted: 03/04/2013] [Indexed: 01/14/2023]
|
183
|
Kasza A. Signal-dependent Elk-1 target genes involved in transcript processing and cell migration. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1026-33. [PMID: 23711433 DOI: 10.1016/j.bbagrm.2013.05.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 05/16/2013] [Accepted: 05/17/2013] [Indexed: 11/25/2022]
Abstract
Elk-1 was regarded as a transcription factor engaged mainly in the regulation of cell growth, differentiation, and survival. Recent findings show the engagement of Elk-1 in the control of expression of genes encoding proteins involved in transcript turnover, such as MCPIP1/ZC3H12A and tristetraprolin (TTP/ZFP36). Thus, Elk-1 plays an important role in the control of gene expression not only through the stimulation of expression of transcription factors, but also through regulation of transcript half-live. Moreover, Elk-1 is engaged in the regulation of expression of genes encoding proteins that control proteolytic activity, such as inhibitor of plasminogen activator-1 (PAI-1) and metalloproteinases-2 and -9 (MMP-2 and MMP-9). This review summarizes the biological roles of proteins with expression regulated by Elk-1, involved in transcripts turnover or in cell migration. The broad range of function of these proteins illustrates the complex role of Elk-1 in the regulation of cancer and inflammation.
Collapse
Affiliation(s)
- Aneta Kasza
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland.
| |
Collapse
|
184
|
Dhamija S, Winzen R, Doerrie A, Behrens G, Kuehne N, Schauerte C, Neumann E, Dittrich-Breiholz O, Kracht M, Holtmann H. Interleukin-17 (IL-17) and IL-1 activate translation of overlapping sets of mRNAs, including that of the negative regulator of inflammation, MCPIP1. J Biol Chem 2013; 288:19250-9. [PMID: 23658019 DOI: 10.1074/jbc.m113.452649] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Changes in gene expression during inflammation are in part caused by post-transcriptional mechanisms. A transcriptome-wide screen for changes in ribosome occupancy indicated that the inflammatory cytokine IL-17 activates translation of a group of mRNAs that overlaps partially with those affected similarly by IL-1. Included are mRNAs of IκBζ and of MCPIP1, important regulators of the quality and course of immune and inflammatory responses. Evidence for increased ribosome association of these mRNAs was also obtained in LPS-activated RAW264.7 macrophages and human peripheral blood mononuclear cells. Like IL-1, IL-17 activated translation of IκBζ mRNA by counteracting the function of a translational silencing element in its 3'-UTR defined previously. Translational silencing of MCPIP1 mRNA in unstimulated cells resulted from the combined suppressive activities of its 5'-UTR, which contains upstream open reading frames, and of its 3'-UTR, which silences independently of the 5'-UTR. Only the silencing function of the 3'-UTR was counteracted by IL-17 as well as by IL-1. Translational silencing by the 3'-UTR was dependent on a putative stem-loop-forming region previously associated with rapid degradation of the mRNA. The results suggest that translational control exerted by IL-1 and IL-17 plays an important role in the coordination of an inflammatory reaction.
Collapse
Affiliation(s)
- Sonam Dhamija
- Institute of Biochemistry, Hannover Medical School, D-30623 Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Skalniak L, Koj A, Jura J. Proteasome inhibitor MG-132 induces MCPIP1 expression. FEBS J 2013; 280:2665-74. [PMID: 23551903 PMCID: PMC3806276 DOI: 10.1111/febs.12264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/19/2013] [Accepted: 03/22/2013] [Indexed: 01/14/2023]
Abstract
The proteasome is a protein complex responsible for the degradation of polyubiquitin-tagged proteins. Besides the removal of target proteins, the proteasome also participates in the regulation of gene transcription in both proteolytic and non-proteolytic fashion. In this study the effect of proteasome inhibition on the basal expression of monocyte chemotactic protein-1 induced protein 1 (MCPIP1) was examined. Treatment of HepG2 or HeLa cells with proteasome inhibitor MG-132 resulted in a significant increase of MCPIP1 expression, both at mRNA and protein level. Interestingly, MG-132 did not alter MCPIP1 stability. Instead, the observed protein increase was blocked by actinomycin D, suggesting the involvement of de novo mRNA synthesis in the increase of MCPIP1 protein following MG-132 treatment. Using several inhibitors we determined the participation of extracellular-signal-regulated kinase 1/2 and p38 kinases in MCPIP1 upregulation by MG-132. Our findings show for the first time the impact of proteasome inhibition on MCPIP1 protein expression by modulation of the activity of intracellular signaling pathways. Overexpression of MCPIP1-myc protein decreased the viability of HeLa cells but not HepG2 cells, which correlates with the increased susceptibility of HeLa cells to MG-132 toxicity. Notably, both MG-132 treatment and MCPIP1-myc overexpression led to the activation of apoptosis, as revealed by the induction of caspases 3/7 in both types of cell lines. This suggests the involvement of MCPIP1 upregulation in toxic properties of proteasome inhibition, which is an acknowledged approach to the treatment of several cancer types.
Collapse
Affiliation(s)
- Lukasz Skalniak
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | | | | |
Collapse
|
186
|
Abstract
RNA interference (RNAi) is an ancient process by which non-coding RNAs regulate gene expression in a sequence-specific manner. The core components of RNAi are small regulatory RNAs, approximately 21-30 nucleotides in length, including small interfering RNAs (siRNAs) and microRNAs (miRNAs). The past two decades have seen considerable progress in our understanding of the molecular mechanisms underlying the biogenesis of siRNAs and miRNAs. Recent advances have also revealed the crucial regulatory roles played by small RNAs in such diverse processes as development, homeostasis, innate immunity, and oncogenesis. Accumulating evidence indicates that RNAi initially evolved as a host defense mechanism against viruses and transposons. The ability of the host small RNA biogenesis machinery to recognize viral double-stranded RNA replication intermediates and transposon transcripts is critical to this process, as is small RNA-guided targeting of RNAs via complementary base pairing. Collectively, these properties confer unparalleled specificity and precision to RNAi-mediated gene silencing as an effective antiviral mechanism.
Collapse
Affiliation(s)
- Rui Zhou
- Program for RNA Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
187
|
RNA decay tolerizes: MCPIP1 (Zc3h12a) keeps inflammation in check by cleaving 3' UTRs. Immunol Cell Biol 2013; 91:331-2. [PMID: 23628803 DOI: 10.1038/icb.2013.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
188
|
Tahara H, Kay MA, Yasui W, Tahara E. MicroRNAs in Cancer: The 22nd Hiroshima Cancer Seminar/The 4th Japanese Association for RNA Interference Joint International Symposium, 30 August 2012, Grand Prince Hotel Hiroshima. Jpn J Clin Oncol 2013; 43:579-82. [DOI: 10.1093/jjco/hyt037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
189
|
Lin RJ, Chien HL, Lin SY, Chang BL, Yu HP, Tang WC, Lin YL. MCPIP1 ribonuclease exhibits broad-spectrum antiviral effects through viral RNA binding and degradation. Nucleic Acids Res 2013; 41:3314-26. [PMID: 23355615 PMCID: PMC3597685 DOI: 10.1093/nar/gkt019] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 12/21/2012] [Accepted: 12/21/2012] [Indexed: 12/31/2022] Open
Abstract
Monocyte chemoattractant protein 1-induced protein 1 (MCPIP1), belonging to the MCPIP family with highly conserved CCCH-type zinc finger and Nedd4-BP1, YacP Nuclease domains, has been implicated in negative regulation of the cellular inflammatory responses. In this report, we demonstrate for the first time that this RNA-binding nuclease also targets viral RNA and possesses potent antiviral activities. Overexpression of the human MCPIP1, but not MCPIP2, MCPIP3 or MCPIP4, inhibited Japanese encephalitis virus (JEV) and dengue virus (DEN) replication. The functional analysis of MCPIP1 revealed that the activities of RNase, RNA binding and oligomerization, but not deubiqutinase, are required for its antiviral potential. Furthermore, infection of other positive-sense RNA viruses, such as sindbis virus and encephalomyocarditis virus, and negative-sense RNA virus, such as influenza virus, as well as DNA virus, such as adenovirus, can also be blocked by MCPIP1. Moreover, the endogenous MCPIP1 gene expression was induced by JEV and DEN infection, and knockdown of MCPIP1 expression enhanced the replication of JEV and DEN in human cells. Thus, MCPIP1 can act as a host innate defense via RNase activity for targeting and degrading viral RNA.
Collapse
Affiliation(s)
- Ren-Jye Lin
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Hsu-Ling Chien
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Shyr-Yi Lin
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Bi-Lan Chang
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Han-Pang Yu
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Wei-Chun Tang
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Yi-Ling Lin
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
190
|
Libri V, Miesen P, van Rij RP, Buck AH. Regulation of microRNA biogenesis and turnover by animals and their viruses. Cell Mol Life Sci 2013; 70:3525-44. [PMID: 23354060 PMCID: PMC3771402 DOI: 10.1007/s00018-012-1257-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 12/27/2012] [Accepted: 12/27/2012] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) are a ubiquitous component of gene regulatory networks that modulate the precise amounts of proteins expressed in a cell. Despite their small size, miRNA genes contain various recognition elements that enable specificity in when, where and to what extent they are expressed. The importance of precise control of miRNA expression is underscored by functional studies in model organisms and by the association between miRNA mis-expression and disease. In the last decade, identification of the pathways by which miRNAs are produced, matured and turned-over has revealed many aspects of their biogenesis that are subject to regulation. Studies in viral systems have revealed a range of mechanisms by which viruses target these pathways through viral proteins or non-coding RNAs in order to regulate cellular gene expression. In parallel, a field of study has evolved around the activation and suppression of antiviral RNA interference (RNAi) by viruses. Virus encoded suppressors of RNAi can impact miRNA biogenesis in cases where miRNA and small interfering RNA pathways converge. Here we review the literature on the mechanisms by which miRNA biogenesis and turnover are regulated in animals and the diverse strategies that viruses use to subvert or inhibit these processes.
Collapse
Affiliation(s)
- Valentina Libri
- Centre for Immunity, Infection and Evolution, University of Edinburgh, King's Buildings, West Mains Road, Edinburgh, EH9 3JT, UK
| | | | | | | |
Collapse
|
191
|
Liang C, Xiong K, Szulwach KE, Zhang Y, Wang Z, Peng J, Fu M, Jin P, Suzuki HI, Liu Q. Sjogren syndrome antigen B (SSB)/La promotes global microRNA expression by binding microRNA precursors through stem-loop recognition. J Biol Chem 2013; 288:723-36. [PMID: 23129761 PMCID: PMC3537071 DOI: 10.1074/jbc.m112.401323] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 10/23/2012] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNA) control numerous physiological and pathological processes. Typically, the primary miRNA (pri-miRNA) transcripts are processed by nuclear Drosha complex into ~70-nucleotide stem-loop precursor miRNAs (pre-miRNA), which are further cleaved by cytoplasmic Dicer complex into ~21-nucleotide mature miRNAs. However, it is unclear how nascent pre-miRNAs are protected from ribonucleases, such as MCPIP1, that degrade pre-miRNAs to abort miRNA production. Here, we identify Sjögren syndrome antigen B (SSB)/La as a pre-miRNA-binding protein that regulates miRNA processing in vitro. All three RNA-binding motifs (LAM, RRM1, and RRM2) of La/SSB are required for efficient pre-miRNA binding. Intriguingly, La/SSB recognizes the characteristic stem-loop structure of pre-miRNAs, of which the majority lack a 3' UUU terminus. Moreover, La/SSB associates with endogenous pri-/pre-miRNAs and promotes miRNA biogenesis by stabilizing pre-miRNAs from nuclease (e.g. MCPIP1)-mediated decay in mammalian cells. Accordingly, we observed positive correlations between the expression status of La/SSB and Dicer in human cancer transcriptome and prognosis. These studies identify an important function of La/SSB as a global regulator of miRNA expression, and implicate stem-loop recognition as a major mechanism that mediates association between La/SSB and diverse RNA molecules.
Collapse
Affiliation(s)
- Chunyang Liang
- From the Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ke Xiong
- From the Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
- the College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Keith E. Szulwach
- the Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Yi Zhang
- From the Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Zhaohui Wang
- From the Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Junmin Peng
- the Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Mingui Fu
- the Department of Basic Medical Science, University of Missouri School of Medicine, Kansas, Missouri 64108, and
| | - Peng Jin
- the Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Hiroshi I. Suzuki
- the Department of Molecular Pathology, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Qinghua Liu
- From the Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
192
|
O'Carroll D, Schaefer A. General principals of miRNA biogenesis and regulation in the brain. Neuropsychopharmacology 2013; 38:39-54. [PMID: 22669168 PMCID: PMC3521995 DOI: 10.1038/npp.2012.87] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/02/2012] [Accepted: 05/02/2012] [Indexed: 02/08/2023]
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that mediate posttranscriptional gene suppression in a sequence-specific manner. The ability of a single miRNA species to target multiple messenger RNAs (mRNAs) makes miRNAs exceptionally important regulators of various cellular functions. The regulatory capacity of miRNAs is increased further by the miRNA ability to suppress gene expression using multiple mechanisms that range from translational inhibition to mRNA degradation. The high miRNA diversity multiplied by the large number of individual miRNA targets generates a vast regulatory RNA network than enables flexible control of mRNA expression. The gene-regulatory capacity and diversity of miRNAs is particularly valuable in the brain, where functional specialization of neurons and persistent flow of information requires constant neuronal adaptation to environmental cues. In this review we will summarize the current knowledge about miRNA biogenesis and miRNA expression regulation with a focus on the role of miRNAs in the mammalian nervous system.
Collapse
Affiliation(s)
- Dónal O'Carroll
- European Molecular Biology Laboratory, Mouse Biology Unit, Monterotondo Scalo, Italy
| | - Anne Schaefer
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
193
|
Suzuki HI, Mihira H, Watabe T, Sugimoto K, Miyazono K. Widespread inference of weighted microRNA-mediated gene regulation in cancer transcriptome analysis. Nucleic Acids Res 2012; 41:e62. [PMID: 23275554 PMCID: PMC3597654 DOI: 10.1093/nar/gks1439] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) comprise a gene-regulatory network through sequence complementarity with target mRNAs. Previous studies have shown that mammalian miRNAs decrease many target mRNA levels and reduce protein production predominantly by target mRNA destabilization. However, it has not yet been fully assessed whether this scheme is widely applicable to more realistic conditions with multiple miRNA fluctuations. By combining two analytical frameworks for detecting the enrichment of gene sets, Gene Set Enrichment Analysis (GSEA) and Functional Assignment of miRNAs via Enrichment (FAME), we developed GSEA–FAME analysis (GFA), which enables the prediction of miRNA activities from mRNA expression data using rank-based enrichment analysis and weighted evaluation of miRNA–mRNA interactions. This cooperative approach delineated a better widespread correlation between miRNA expression levels and predicted miRNA activities in cancer transcriptomes, thereby providing proof-of-concept of the mRNA-destabilization scenario. In an integrative analysis of The Cancer Genome Atlas (TCGA) multidimensional data including profiles of both mRNA and miRNA, we also showed that GFA-based inference of miRNA activity could be used for the selection of prognostic miRNAs in the development of cancer survival prediction models. This approach proposes a next-generation strategy for the interpretation of miRNA function and identification of target miRNAs as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Hiroshi I Suzuki
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo 113-0033, Japan.
| | | | | | | | | |
Collapse
|
194
|
Morrison BJ, Hastie ML, Grewal YS, Bruce ZC, Schmidt C, Reynolds BA, Gorman JJ, Lopez JA. Proteomic comparison of mcf-7 tumoursphere and monolayer cultures. PLoS One 2012; 7:e52692. [PMID: 23285151 PMCID: PMC3527578 DOI: 10.1371/journal.pone.0052692] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/21/2012] [Indexed: 01/09/2023] Open
Abstract
Breast cancer is a heterogenous disease, composed of tumour cells with differing gene expressions and phenotypes. Very few antigens have been identified and a better understanding of tumour initiating-cells as targets for therapy is critically needed. Recently, a rare subpopulation of cells within tumours has been described with the ability to: (i) initiate and sustain tumour growth; (ii) resist traditional therapies and allow for secondary tumour dissemination; and (iii) display some of the characteristics of stem cells such as self-renewal. These cells are termed tumour-initiating cells or cancer stem cells, or alternatively, in the case of breast cancer, breast cancer stem cells. Previous studies have demonstrated that breast cancer stem cells can be enriched for in “tumoursphere” culture. Proteomics represents a novel way to investigate protein expression between cells. We hypothesise that characterisation of the proteome of the breast cancer line MCF-7 tumourspheres compared to adherent/differentiated cells identifies proteins of novel interest for further isolating or targeting breast cancer stem cells. We present evidence that: (i) the proteome of adherent cells is different to the proteome of cells grown in sphere medium from either early passage (passage 2) or late passage (passage 5) spheres; (ii) that spheres are enriched in expression of a variety of tumour-relevant proteins (including MUC1 and Galectin-3); and (iii) that targeting of one of these identified proteins (galectin-3) using an inhibitor (N-acetyllactosamine) decreases sphere formation/self-renewal of MCF-7 cancer stem cells in vitro and tumourigenicity in vivo. Hence, proteomic analysis of tumourspheres may find use in identifying novel targets for future therapy. The therapeutic targeting of breast cancer stem cells, a highly clinically relevant sub-population of tumour cells, has the potential to eliminate residual disease and may become an important component of a multi-modality treatment of cancer.
Collapse
Affiliation(s)
- Brian J. Morrison
- School of Biomolecular and Physical Sciences, Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Marcus L. Hastie
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Yadveer S. Grewal
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Zara C. Bruce
- School of Biomolecular and Physical Sciences, Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Chris Schmidt
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Brent A. Reynolds
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| | - Jeffrey J. Gorman
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - J. Alejandro Lopez
- School of Biomolecular and Physical Sciences, Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
195
|
Wang Y, Taniguchi T. MicroRNAs and DNA damage response: implications for cancer therapy. Cell Cycle 2012; 12:32-42. [PMID: 23255103 DOI: 10.4161/cc.23051] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The DNA damage response (DDR) pathways play critical roles in protecting the genome from DNA damage. Abrogation of DDR often results in elevated genomic instability and cellular sensitivity to DNA damaging agents. Many proteins involved in DDR are subjected to precise regulation at multiple levels, such as transcriptional control and posttranslational modifications, in response to DNA damage. MicroRNAs (miRNAs) are a class of small non-coding RNAs that negatively regulate gene expression at the post-transcriptional level. The expression levels of some miRNAs change in response to DNA damage. Some miRNAs, such as miR-24, 138, 96 and 182, have been implicated in DDR and/or DNA repair and affect cellular sensitivity to DNA damaging agents. In this review, we summarize recent findings related to the emerging roles of miRNAs in regulating DDR and DNA repair and discuss their potential in cancer therapy.
Collapse
Affiliation(s)
- Yemin Wang
- Howard Hughes Medical Institute, Human Biology and Public Health Sciences Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | |
Collapse
|
196
|
Oestrogen-dependent regulation of miRNA biogenesis: many ways to skin the cat. Biochem Soc Trans 2012; 40:752-8. [PMID: 22817728 DOI: 10.1042/bst20110763] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The steroid hormone oestrogen is central to normal female physiology, reproduction and behaviour, through its effects on cellular processes including cell proliferation and cell survival. The effects of oestrogen are mediated by nuclear ERs (oestrogen receptors). ER status is important for the development, progression and treatment of breast cancer. miRNAs (microRNAs) are small non-coding RNAs that bind the 3'-UTR (untranslated region) of target mRNAs to reduce their stability and/or translation. miRNAs participate in oestrogen signalling by regulating oestrogen-responsive genes and pathways. Interestingly expression and maturation of miRNAs can also be regulated by ER signalling at multiple levels. In addition to regulating the expression of miRNAs at the transcriptional level, ER appears to be able to regulate the biogenesis of miRNAs. In the present review, we summarize recent findings on miRNA biogenesis and describe various mechanisms by which oestrogen signalling can modulate the production of miRNAs.
Collapse
|
197
|
Abstract
Regulation of miRNA (microRNA) biogenesis shapes the profiles of miRNAs in the living cell, contributing to cell identity and function. Importantly, aberrant miRNA levels have been linked to a variety of human pathological states. In recent years, a number of proteins have been shown to regulate the miRNA biogenesis at the level of Drosha and Dicer cleavage. A large proportion of these factors regulate miRNA production through binding to the TL (terminal loop) regions of miRNA progenitors. In the present paper, we review the current knowledge about pri-miRNA (primary miRNA) and pre-miRNA (precursor miRNA) TL involvement in the regulation of miRNA biogenesis.
Collapse
|
198
|
Li M, Cao W, Liu H, Zhang W, Liu X, Cai Z, Guo J, Wang X, Hui Z, Zhang H, Wang J, Wang L. MCPIP1 down-regulates IL-2 expression through an ARE-independent pathway. PLoS One 2012; 7:e49841. [PMID: 23185455 PMCID: PMC3504106 DOI: 10.1371/journal.pone.0049841] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 10/12/2012] [Indexed: 01/14/2023] Open
Abstract
IL-2 plays a key role in the survival and proliferation of immune cells, especially T lymphocytes. Its expression is precisely regulated at transcriptional and posttranscriptional level. IL-2 is known to be regulated by RNA binding proteins, such as tristetraprolin (TTP), via an AU-rich element (ARE) in the 3'-untranslated region (3'UTR) to influence the stability of mRNA. MCPIP1, identified as a novel RNase, can degrade IL-6, IL-12 and TNF-α mRNA by an ARE-independent pathway in the activation of macrophages. Here, we reported that MCPIP1 was induced in the activation of T lymphocytes and negatively regulated IL-2 gene expression in both mouse and human primary T lymphocytes through destabilizing its mRNA. A set of Luciferase reporter assay demonstrated that a non-ARE conserved element in IL-2 3'UTR, which formed a stem-loop structure, responded to MCPIP1 activity.RNA immunoprecipitation and Biotin pulldown experiments further suggested that MCPIP1 could modestly bind to IL-2 mRNA. Taken together, these data demonstrate that MCPIP1 down-regulates IL-2 via an ARE-independent pathway.
Collapse
Affiliation(s)
- Min Li
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenqiang Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Haifeng Liu
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Guo
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuelian Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaoyuan Hui
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Zhang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianli Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lie Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
199
|
Abstract
MicroRNAs (miRNAs) function as 21-24 nucleotide guide RNAs that use partial base-pairing to recognize target messenger RNAs and repress their expression. As a large fraction of protein-coding genes are under miRNA control, production of the appropriate level of specific miRNAs at the right time and in the right place is integral to most gene regulatory pathways. MiRNA biogenesis initiates with transcription, followed by multiple processing steps to produce the mature miRNA. Every step of miRNA production is subject to regulation and disruption of these control mechanisms has been linked to numerous human diseases, where the balance between the expression of miRNAs and their targets becomes distorted. Here we review the basic steps of miRNA biogenesis and describe the various factors that control miRNA transcription, processing, and stability in animal cells. The tremendous effort put into producing the appropriate type and level of specific miRNAs underscores the critical role of these small RNAs in gene regulation.
Collapse
Affiliation(s)
- Emily F Finnegan
- Division of Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
200
|
Manavella PA, Hagmann J, Ott F, Laubinger S, Franz M, Macek B, Weigel D. Fast-forward genetics identifies plant CPL phosphatases as regulators of miRNA processing factor HYL1. Cell 2012; 151:859-870. [PMID: 23141542 DOI: 10.1016/j.cell.2012.09.039] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 06/08/2012] [Accepted: 09/30/2012] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) are processed from primary transcripts that contain partially self-complementary foldbacks. As in animals, the core microprocessor in plants is a Dicer protein, DICER-LIKE1 (DCL1). Processing accuracy and strand selection is greatly enhanced through the RNA binding protein HYPONASTIC LEAVES 1 (HYL1) and the zinc finger protein SERRATE (SE). We have combined a luciferase-based genetic screen with whole-genome sequencing for rapid identification of new regulators of miRNA biogenesis and action. Among the first six mutants analyzed were three alleles of C-TERMINAL DOMAIN PHOSPHATASE-LIKE 1 (CPL1)/FIERY2 (FRY2). In the miRNA processing complex, SE functions as a scaffold to mediate CPL1 interaction with HYL1, which needs to be dephosphorylated for optimal activity. In the absence of CPL1, HYL1 dephosphorylation and hence accurate processing and strand selection from miRNA duplexes are compromised. Our findings thus define a new regulatory step in plant miRNA biogenesis.
Collapse
Affiliation(s)
- Pablo A Manavella
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Jörg Hagmann
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Felix Ott
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Sascha Laubinger
- Center for Plant Molecular Biology (ZMBP), University of Tübingen, 72076 Tübingen, Germany
| | - Mirita Franz
- Proteome Center, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Boris Macek
- Proteome Center, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Detlef Weigel
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany.
| |
Collapse
|