151
|
Athanasopoulos V, Ramiscal RR, Vinuesa CG. ROQUIN signalling pathways in innate and adaptive immunity. Eur J Immunol 2016; 46:1082-90. [PMID: 27060455 DOI: 10.1002/eji.201545956] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/15/2016] [Accepted: 03/30/2016] [Indexed: 12/25/2022]
Abstract
ROQUIN is an RNA-binding protein that plays important roles in both the innate and adaptive immune systems. ROQUIN binds to several key immune-relevant messenger RNA (mRNA) targets through its ROQ domain modulating their stability and influencing macrophage function and the peripheral homeostasis of T cells and B cells. More recently, the E3 ubiquitin ligase activity of the ROQUIN RING domain has been shown to be crucial for T-cell-dependent B-cell responses against infection. Defective ROQUIN activity can culminate in a range of diseases, such as systemic autoimmunity, immunodeficiency, and inflammatory bowel disorder. Here, we provide a current overview of the immunomodulatory role of ROQUIN defined by its ribonucleoprotein-like structure, its repertoire of mRNA targets shared by related RNA-binding enzymes, and its involvement in a range of intracellular signalling pathways central to shaping immune responses.
Collapse
Affiliation(s)
- Vicki Athanasopoulos
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Roybel R Ramiscal
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Carola G Vinuesa
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| |
Collapse
|
152
|
Emerging Roles for MicroRNAs in T Follicular Helper Cell Differentiation. Trends Immunol 2016; 37:297-309. [PMID: 27068008 DOI: 10.1016/j.it.2016.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 03/03/2016] [Accepted: 03/13/2016] [Indexed: 01/07/2023]
Abstract
T follicular helper (Tfh) cells are essential for the formation of germinal centers (GCs) and the development of long-lived humoral immunity. Tfh cell differentiation is a multistep process driven by the balanced expression of key transcription factors that form a regulatory network in which small changes in gene expression determine the Tfh cell fate decision. Here, we review recent findings that have revealed that certain microRNAs act as important mediators within this network, with roles in tuning gene expression. We integrate these findings into the current understanding of the mechanisms governing T helper cell differentiation, and propose a model in which the establishment of Tfh cell identity is dependent on the differential expression and concerted action of distinct microRNAs and transcription factors.
Collapse
|
153
|
Contreras JR, Palanichamy JK, Tran TM, Fernando TR, Rodriguez-Malave NI, Goswami N, Arboleda VA, Casero D, Rao DS. MicroRNA-146a modulates B-cell oncogenesis by regulating Egr1. Oncotarget 2016; 6:11023-37. [PMID: 25906746 PMCID: PMC4484436 DOI: 10.18632/oncotarget.3433] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 02/24/2015] [Indexed: 11/25/2022] Open
Abstract
miR-146a is a NF-κB induced microRNA that serves as a feedback regulator of this critical pathway. In mice, deficiency of miR-146a results in hematolymphoid cancer at advanced ages as a consequence of constitutive NF-κB activity. In this study, we queried whether the deficiency of miR-146a contributes to B-cell oncogenesis. Combining miR-146a deficiency with transgenic expression of c-Myc led to the development of highly aggressive B-cell malignancies. Mice transgenic for c-Myc and deficient for miR-146a were characterized by significantly shortened survival, increased lymph node involvement, differential involvement of the spleen and a mature B-cell phenotype. High-throughput sequencing of the tumors revealed significant dysregulation of approximately 250 genes. Amongst these, the transcription factor Egr1 was consistently upregulated in mice deficient for miR-146a. Interestingly, transcriptional targets of Egr1 were enriched in both the high-throughput dataset and in a larger set of miR-146a-deficient tumors. miR-146a overexpression led to downregulation of Egr1 and downstream targets with concomitant decrease in cell growth. Direct targeting of the human EGR1 by miR-146a was seen by luciferase assay. Together our findings illuminate a bona fide role for miR-146a in the modulation of B-cell oncogenesis and reveal the importance of understanding microRNA function in a cell- and disease-specific context.
Collapse
Affiliation(s)
- Jorge R Contreras
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, CA, USA
| | | | - Tiffany M Tran
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
| | - Thilini R Fernando
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
| | - Norma I Rodriguez-Malave
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, CA, USA
| | - Neha Goswami
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
| | - Valerie A Arboleda
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
| | - David Casero
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.,Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA
| |
Collapse
|
154
|
Ji Y, Hocker JD, Gattinoni L. Enhancing adoptive T cell immunotherapy with microRNA therapeutics. Semin Immunol 2015; 28:45-53. [PMID: 26710685 DOI: 10.1016/j.smim.2015.11.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 12/15/2022]
Abstract
Adoptive T cell-based immunotherapies can mediate complete and durable regressions in patients with advanced cancer, but current response rates remain inadequate. Maneuvers to improve the fitness and antitumor efficacy of transferred T cells have been under extensive exploration in the field. Small non-coding microRNAs have emerged as critical modulators of immune system homeostasis and T cell immunity. Here, we summarize recent advances in our understanding of the role of microRNAs in regulating T cell activation, differentiation, and function. We also discuss how microRNA therapeutics could be employed to fine-tune T cell receptor signaling and enhance T cell persistence and effector functions, paving the way for the next generation of adoptive immunotherapies.
Collapse
Affiliation(s)
- Yun Ji
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA.
| | - James D Hocker
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA
| | - Luca Gattinoni
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA.
| |
Collapse
|
155
|
Samadi AK, Bilsland A, Georgakilas AG, Amedei A, Amin A, Bishayee A, Azmi AS, Lokeshwar BL, Grue B, Panis C, Boosani CS, Poudyal D, Stafforini DM, Bhakta D, Niccolai E, Guha G, Vasantha Rupasinghe HP, Fujii H, Honoki K, Mehta K, Aquilano K, Lowe L, Hofseth LJ, Ricciardiello L, Ciriolo MR, Singh N, Whelan RL, Chaturvedi R, Ashraf SS, Shantha Kumara HMC, Nowsheen S, Mohammed SI, Keith WN, Helferich WG, Yang X. A multi-targeted approach to suppress tumor-promoting inflammation. Semin Cancer Biol 2015; 35 Suppl:S151-S184. [PMID: 25951989 PMCID: PMC4635070 DOI: 10.1016/j.semcancer.2015.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.
Collapse
Affiliation(s)
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Asfar S Azmi
- Department of Pathology, Wayne State Univeristy, Karmanos Cancer Center, Detroit, MI, USA
| | - Bal L Lokeshwar
- Department of Urology, University of Miami, Miller School of Medicine, Miami, FL, United States; Miami Veterans Administration Medical Center, Miami, FL, United States
| | - Brendan Grue
- Department of Environmental Science, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada.
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
156
|
Wissink EM, Smith NL, Spektor R, Rudd BD, Grimson A. MicroRNAs and Their Targets Are Differentially Regulated in Adult and Neonatal Mouse CD8+ T Cells. Genetics 2015; 201:1017-30. [PMID: 26416483 PMCID: PMC4649632 DOI: 10.1534/genetics.115.179176] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 09/18/2015] [Indexed: 11/30/2022] Open
Abstract
Immunological memory, which protects organisms from re-infection, is a hallmark of the mammalian adaptive immune system and the underlying principle of vaccination. In early life, however, mice and other mammals are deficient at generating memory CD8+ T cells, which protect organisms from intracellular pathogens. The molecular basis that differentiates adult and neonatal CD8+ T cells is unknown. MicroRNAs (miRNAs) are both developmentally regulated and required for normal adult CD8+ T cell functions. We used next-generation sequencing to identify mouse miRNAs that are differentially regulated in adult and neonatal CD8+ T cells, which may contribute to the impaired development of neonatal memory cells. The miRNA profiles of adult and neonatal cells were surprisingly similar during infection; however, we observed large differences prior to infection. In particular, miR-29 and miR-130 have significant differential expression between adult and neonatal cells before infection. Importantly, using RNA-Seq, we detected reciprocal changes in expression of messenger RNA targets for both miR-29 and miR-130. Moreover, targets that we validated include Eomes and Tbx21, key genes that regulate the formation of memory CD8+ T cells. Notably, age-dependent changes in miR-29 and miR-130 are conserved in human CD8+ T cells, further suggesting that these developmental differences are biologically relevant. Together, these results demonstrate that miR-29 and miR-130 are likely important regulators of memory CD8+ T cell formation and suggest that neonatal cells are committed to a short-lived effector cell fate prior to infection.
Collapse
Affiliation(s)
- Erin M Wissink
- Graduate Field of Biochemistry, Molecular, and Cell Biology, Cornell University, Ithaca, New York 14853
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853
| | - Roman Spektor
- Graduate Field of Genetics, Genomics, and Development, Cornell University, Ithaca, New York 14853
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York 14853
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| |
Collapse
|
157
|
Dysregulated co-stimulatory molecule expression in a Sjögren's syndrome mouse model with potential implications by microRNA-146a. Mol Immunol 2015; 68:606-16. [PMID: 26505653 DOI: 10.1016/j.molimm.2015.09.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 09/22/2015] [Accepted: 09/30/2015] [Indexed: 01/04/2023]
Abstract
Sjögren's syndrome (SjS) is an autoimmune condition that primarily affects salivary and lacrimal glands, causing loss of secretion. We have previously shown that microRNA-146a (miR-146a) is over-expressed in the salivary glands and peripheral blood mononuclear cells (PBMC) of SjS-prone mice (C57BL/6.NOD-Aec1Aec2, B6DC) and in PBMC of SjS patients. The purpose of this research was to identify a target molecule of miR-146a and identify subpopulations of cells affected by altered miR-146a in the salivary glands of SjS-prone mice. In silico analyses identified costimulatory molecule CD80 as a potential target of miR-146a. Luciferase assay of the human CD80 3'untranslated region demonstrated miR-146a directly inhibited CD80 protein expression as indicated by reduced luciferase reporter expression and an examination of B6DC salivary glands revealed a reduction in CD80 protein. More interestingly, the specific reduction in CD80 protein was detected from the salivary gland epithelial cell population and in interstitial dendritic cells in the glands as well. The reduction in CD80 protein levels in salivary gland epithelial cells were negatively associated with elevated miR-146a expression. Therefore, this study provides the first indication that salivary gland epithelial cells may be critically involved in SjS progression by altering CD86:CD80 protein ratio in response to miR-146a upregulation.
Collapse
|
158
|
Leonov G, Shah K, Yee D, Timmis J, Sharp TV, Lagos D. Suppression of AGO2 by miR-132 as a determinant of miRNA-mediated silencing in human primary endothelial cells. Int J Biochem Cell Biol 2015; 69:75-84. [PMID: 26475020 PMCID: PMC4679077 DOI: 10.1016/j.biocel.2015.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/08/2015] [Accepted: 10/07/2015] [Indexed: 01/07/2023]
Abstract
The abundance of miR-132 ranges from constitutively high in the brain where it is necessary for neuronal development and function, to inducible expression in haematopoietic and endothelial cells where it controls angiogenesis and immune activation. We show that expression of AGO2, a protein central to miRNA-mediated gene silencing and miRNA biogenesis, is negatively regulated by miR-132. Using HeLa cells, we demonstrate that miR-132 interacts with the AGO2 mRNA 3'UTR and suppresses AGO2 expression and AGO2-dependent small RNA-mediated silencing. Similarly, miR-132 over-expression leads to AGO2 suppression in primary human dermal lymphatic endothelial cells (HDLECs). During phorbol myristate acetate (PMA)-activation of HDLECs, miR-132 is induced in a CREB-dependent manner and inhibition of miR-132 results in increased AGO2 expression. In agreement with the role of AGO2 in maintenance of miRNA expression, AGO2 suppression by miR-132 affects the steady state levels of miR-221 and miR-146a, two miRNAs involved in angiogenesis and inflammation, respectively. Our data demonstrate that the miRNA-silencing machinery is subject to autoregulation during primary cell activation through direct suppression of AGO2 by miR-132.
Collapse
Affiliation(s)
- German Leonov
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School University of York, Wentworth Way, York YO10 5DD, UK
| | - Kunal Shah
- Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University London, London EC1M 6BQ, UK
| | - Daniel Yee
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School University of York, Wentworth Way, York YO10 5DD, UK
| | - Jon Timmis
- Department of Electronics, Wentworth Way, York YO10 5DD, UK
| | - Tyson V Sharp
- Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University London, London EC1M 6BQ, UK
| | - Dimitris Lagos
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School University of York, Wentworth Way, York YO10 5DD, UK.
| |
Collapse
|
159
|
Runtsch MC, Hu R, Alexander M, Wallace J, Kagele D, Petersen C, Valentine JF, Welker NC, Bronner MP, Chen X, Smith DP, Ajami NJ, Petrosino JF, Round JL, O'Connell RM. MicroRNA-146a constrains multiple parameters of intestinal immunity and increases susceptibility to DSS colitis. Oncotarget 2015; 6:28556-72. [PMID: 26456940 PMCID: PMC4745677 DOI: 10.18632/oncotarget.5597] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/22/2015] [Indexed: 12/12/2022] Open
Abstract
Host-microbial interactions within the mammalian intestines must be properly regulated in order to promote host health and limit disease. Because the microbiota provide constant immunological signals to intestinal tissues, a variety of regulatory mechanisms have evolved to ensure proper immune responses to maintain homeostasis. However, many of the genes that comprise these regulatory pathways, including immune-modulating microRNAs (miRNAs), have not yet been identified or studied in the context of intestinal homeostasis. Here, we investigated the role of microRNA-146a (miR-146a) in regulating intestinal immunity and barrier function and found that this miRNA is expressed in a variety of gut tissues in adult mice. By comparing intestinal gene expression in WT and miR-146a-/- mice, we demonstrate that miR-146a represses a subset of gut barrier and inflammatory genes all within a network of immune-related signaling pathways. We also found that miR-146a restricts the expansion of intestinal T cell populations, including Th17, Tregs, and Tfh cells. GC B cells, Tfh ICOS expression, and the production of luminal IgA were also reduced by miR-146a in the gut. Consistent with an enhanced intestinal barrier, we found that miR-146a-/- mice are resistant to DSS-induced colitis, a model of Ulcerative Colitis (UC), and this correlated with elevated colonic miR-146a expression in human UC patients. Taken together, our data describe a role for miR-146a in constraining intestinal barrier function, a process that alters gut homeostasis and enhances at least some forms of intestinal disease in mice.
Collapse
Affiliation(s)
- Marah C. Runtsch
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Ruozhen Hu
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | | | - Jared Wallace
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Dominique Kagele
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Charisse Petersen
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - John F. Valentine
- Department of Medicine, Division of Gastroenterology, University of Utah, Salt Lake City, UT, USA
| | - Noah C. Welker
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Mary P. Bronner
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Xinjian Chen
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Daniel P. Smith
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nadim J. Ajami
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph F. Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - June L. Round
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Ryan M. O'Connell
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
160
|
Jiang SH, Shen N, Vinuesa CG. Posttranscriptional T cell gene regulation to limit Tfh cells and autoimmunity. Curr Opin Immunol 2015; 37:21-7. [PMID: 26432764 DOI: 10.1016/j.coi.2015.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/23/2015] [Accepted: 09/10/2015] [Indexed: 01/06/2023]
Abstract
T follicular helper (Tfh) cells are crucial to induce protective extrafollicular and germinal center antibody responses against protein antigens. Over the last decade, control of Tfh cell numbers has emerged as an important regulatory checkpoint which, when perturbed, may lead to production of autoantibodies. Recent progress in understanding how Tfh cells are kept limiting has revealed an important role for posttranscriptional control of gene expression mediated by microRNAs such as miR-17 ∼ 92, miR-155 and miR-146a, and the RNA-binding proteins Roquin and Regnase. Additionally, T cell microRNAs dysregulated in patients with systemic lupus erythematosus have been shown to influence processes such as DNA hypomethylation, IL-2 and CCL5 secretion, and Treg function, which contribute to autoantibody formation and tissue damage.
Collapse
Affiliation(s)
- Simon H Jiang
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research and Centre for Personalised Immunology, Australian National University, Canberra, Australia; Department of Renal Medicine, The Canberra Hospital, Canberra, Australia; China Australia Centre for Personalised Immunology, Shanghai Renji Hospital, China and Australian National University, Australia.
| | - Nan Shen
- China Australia Centre for Personalised Immunology, Shanghai Renji Hospital, China and Australian National University, Australia; Joint Molecular Rheumatology Laboratory of the Institute of Health Sciences and Shanghai Renji Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research and Centre for Personalised Immunology, Australian National University, Canberra, Australia; China Australia Centre for Personalised Immunology, Shanghai Renji Hospital, China and Australian National University, Australia.
| |
Collapse
|
161
|
Abstract
Systemic lupus erythematosus is a prototypic autoimmune disease characterized by the production of an array of pathogenic autoantibodies, including high-affinity anti-dsDNA IgG antibodies, which play an important role in disease development and progression. Lupus preferentially affects women during their reproductive years. The pathogenesis of lupus is contributed by both genetic factors and epigenetic modifications that arise from exposure to the environment. Epigenetic marks, including DNA methylation, histone post-translational modifications and microRNAs (miRNAs), interact with genetic programs to regulate immune responses. Epigenetic modifications influence gene expression and modulate B cell functions, such as class-switch DNA recombination, somatic hypermutation and plasma cell differentiation, thereby informing the antibody response. Epigenetic dysregulation can result in aberrant antibody responses to exogenous antigens or self-antigens, such as chromatin, histones and dsDNA in lupus. miRNAs play key roles in the post-transcriptional regulation of most gene-regulatory pathways and regulate both the innate and adaptive immune responses. In mice, dysregulation of miRNAs leads to aberrant immune responses and development of systemic autoimmunity. Altered miRNA expression has been reported in human autoimmune diseases, including lupus. The dysregulation of miRNAs in lupus could be the result of multiple environmental factors, such as sex hormones and viral or bacterial infection. Modulation of miRNA is a potential therapeutic strategy for lupus.
Collapse
Affiliation(s)
- Hong Zan
- Department of Microbiology and Immunology, School of Medicine, University of Texas Health Science Center , San Antonio, TX , USA
| | | | | |
Collapse
|
162
|
MiR-146b Mediates Endotoxin Tolerance in Human Phagocytes. Mediators Inflamm 2015; 2015:145305. [PMID: 26451077 PMCID: PMC4584235 DOI: 10.1155/2015/145305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/26/2015] [Accepted: 07/12/2015] [Indexed: 01/06/2023] Open
Abstract
A proper regulation of the innate immune response is fundamental to keep the immune system in check and avoid a chronic status of inflammation. As they act as negative modulators of TLR signaling pathways, miRNAs have been recently involved in the control of the inflammatory response. However, their role in the context of endotoxin tolerance is just beginning to be explored. We here show that miR-146b is upregulated in human monocytes tolerized by LPS, IL-10, or TGFβ priming and demonstrate that its transcription is driven by STAT3 and RUNX3, key factors downstream of IL-10 and TGFβ signaling. Our study also found that IFNγ, known to revert LPS tolerant state, inhibits miR-146b expression. Finally, we provide evidence that miR-146b levels have a profound effect on the tolerant state, thus candidating miR-146b as a molecular mediator of endotoxin tolerance.
Collapse
|
163
|
Hakimi MA, Bougdour A. Toxoplasma 's ways of manipulating the host transcriptome via secreted effectors. Curr Opin Microbiol 2015; 26:24-31. [DOI: 10.1016/j.mib.2015.04.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/06/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022]
|
164
|
Zhou Q, Haupt S, Kreuzer JT, Hammitzsch A, Proft F, Neumann C, Leipe J, Witt M, Schulze-Koops H, Skapenko A. Decreased expression of miR-146a and miR-155 contributes to an abnormal Treg phenotype in patients with rheumatoid arthritis. Ann Rheum Dis 2015; 74:1265-74. [PMID: 24562503 DOI: 10.1136/annrheumdis-2013-204377] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/24/2014] [Indexed: 12/31/2022]
Abstract
OBJECTIVES MicroRNAs (miRNAs) have been implicated in the pathogenesis of autoimmune diseases, not least for their critical role in the regulation of regulatory T cell (Treg) function. Deregulated expression of miR-146a and miR-155 has been associated with rheumatoid arthritis (RA). We therefore investigated miR-146a and miR-155 expression in Tregs of patients with RA and their possible impact on Treg function and disease activity. METHODS Expression of miR-146a and miR-155 was assessed in RA patients and controls. MiRNA expression was correlated with disease activity and expression of target genes. Interference with biological activity of miRNAs was evaluated in functional Treg assays. RESULTS Diminished upregulation of miR-146a and miR-155 in response to T cell stimulation was found in Tregs of RA patients. Diminution of miR-146a expression was observed in particular in patients with active disease, and correlated with joint inflammation. In patients with active RA, Tregs demonstrated a pro-inflammatory phenotype characterised by inflammatory cytokine expression. This was due to an augmented expression and activation of signal transducer and activator transcription 1 (STAT1), a direct target of miR-146a. CONCLUSIONS Our results suggest that in RA miR-146a facilitates a pro-inflammatory phenotype of Tregs via increased STAT1 activation, and contributes thereby to RA pathogenesis.
Collapse
Affiliation(s)
- Qihui Zhou
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Sonja Haupt
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Johannes T Kreuzer
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Ariane Hammitzsch
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Fabian Proft
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Carla Neumann
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Jan Leipe
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Matthias Witt
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Alla Skapenko
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| |
Collapse
|
165
|
Amado T, Schmolka N, Metwally H, Silva-Santos B, Gomes AQ. Cross-regulation between cytokine and microRNA pathways in T cells. Eur J Immunol 2015; 45:1584-95. [PMID: 25865116 DOI: 10.1002/eji.201545487] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/25/2015] [Accepted: 04/08/2015] [Indexed: 01/27/2023]
Abstract
microRNA (miRNA) mediated regulation of protein expression has emerged as an important mechanism in T-cell physiology, from development and survival to activation, proliferation, and differentiation. One of the major classes of proteins involved in these processes are cytokines, which are both key input signals and major products of T-cell function. Here, we summarize the current data on the molecular cross-talk between cytokines and miRNAs: how cytokines regulate miRNA expression, and how specific miRNAs control cytokine production in T cells. We also describe the inflammatory consequences of deregulating the miRNA/cytokine axis in mice and humans. We believe this topical area will have key implications for immune modulation and treatment of autoimmune pathology.
Collapse
Affiliation(s)
- Tiago Amado
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nina Schmolka
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Hozaifa Metwally
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Anita Q Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Escola Superior de Tecnologia da Saúde de Lisboa, Lisboa, Portugal
| |
Collapse
|
166
|
Li P, Spolski R, Liao W, Leonard WJ. Complex interactions of transcription factors in mediating cytokine biology in T cells. Immunol Rev 2015; 261:141-56. [PMID: 25123282 DOI: 10.1111/imr.12199] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
T-helper (Th) cells play critical roles within the mammalian immune system, and the differentiation of naive CD4(+) T cells into distinct T-helper subsets is critical for normal immunoregulation and host defense. These carefully regulated differentiation processes are controlled by networks of cytokines, transcription factors, and epigenetic modifications, resulting in the generation of multiple CD4(+) T-cell subsets, including Th1, Th2, Th9, Th17, Treg, and Tfh cells. In this review, we discuss the roles of transcription factors in determining the specific type of differentiation and in particular the role of interleukin-2 (IL-2) in promoting or inhibiting Th differentiation. In addition to discussing master regulators and subset-specific transcription factors for distinct T-helper cell populations, we focus on signal transducer and activator of transcription (STAT) proteins and on the cooperative action of interferon regulatory factor 4 (IRF4) with activator protein 1 (AP-1) family proteins and STAT3 in the assembly of complexes that broadly influence T-cell differentiation.
Collapse
Affiliation(s)
- Peng Li
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
167
|
Song J, Lee JE. miR-155 is involved in Alzheimer's disease by regulating T lymphocyte function. Front Aging Neurosci 2015; 7:61. [PMID: 25983691 PMCID: PMC4415416 DOI: 10.3389/fnagi.2015.00061] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 04/09/2015] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is considered the most common cause of sporadic dementia. In AD, adaptive and innate immune responses play a crucial role in clearance of amyloid beta and maintenance of cognitive functions. In addition to other changes in the immune system, AD alters the T-cell responses that affect activation of glial cells, neuronal cells, macrophages, and secretion of pro-inflammatory cytokines. These changes in the immune system influence AD pathogenesis. Micro-RNA (miRNA)-155 is a multifunctional miRNA with a distinct expression profile. It is involved in diverse physiological and pathological mechanisms, such as immunity and inflammation. Recent studies indicate that miR-155 regulates T-cell functions during inflammation. In this article, we summarize recent studies describing the therapeutic potential of miR-155 via regulation of T cells in AD. Further, we propose that regulation of miR-155 might be a new protective approach against AD pathogenesis.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine Seoul, South Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine Seoul, South Korea ; Brain Korea 21 Plus Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine Seoul, South Korea
| |
Collapse
|
168
|
Xu HY, Wang ZY, Chen JF, Wang TY, Wang LL, Tang LL, Lin XY, Zhang CW, Chen BC. Association between ankylosing spondylitis and the miR-146a and miR-499 polymorphisms. PLoS One 2015; 10:e0122055. [PMID: 25836258 PMCID: PMC4383612 DOI: 10.1371/journal.pone.0122055] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 02/06/2015] [Indexed: 01/07/2023] Open
Abstract
miRNAs are small, non-coding RNAs that regulate the expression of multiple target genes at the post-transcriptional level. Single-nucleotide polymorphisms (SNPs) in miRNA sequences may alter miRNA expression and have been implicated in the pathogenesis of multiple forms of arthritis, including rheumatoid arthritis (RA) and osteoarthritis. The present study explored the association between ankylosing spondylitis (AS) and two single nucleotide polymorphisms (SNPs), miR-146a rs2910164G>C and miR-499 rs3746444T>C, in a Han Chinese population. A case-control study consisting of 102 subjects with AS and 105 healthy controls was designed. The two miRNA SNPs were identified by direct sequencing. Subsequently, their gene and genotype frequencies were compared with healthy controls. A significant difference was observed in the miR-146a rs2910164G>C SNP. The frequency of the G allele was markedly higher in the AS patients than in the healthy controls (P = 0.005, Pc = 0.01, OR = 1.787), and the frequency of the GG genotype was higher in AS patients than in controls (P = 0.014, Pc = 0.042, OR = 2.516). However, no significant association was found between the miR-499 rs3746444T>C variant and susceptibility to AS. This is the first study to address the association between the miR-146a rs2910164G>C and miR-499 rs3746444T>C polymorphisms and AS, and it suggests a potential pathogenic factor for AS. Further studies are needed to validate our findings in a larger series, as well as in other ethnic backgrounds.
Collapse
Affiliation(s)
- Hui Ying Xu
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhang Yang Wang
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jing Feng Chen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Tian Yang Wang
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ling Ling Wang
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Li Li Tang
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xian-yang Lin
- Injury Orthopaedics of Traditional Chinese medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Chun-wu Zhang
- Injury Orthopaedics of Traditional Chinese medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- * E-mail:
| | - Bi-cheng Chen
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
169
|
Mele F, Basso C, Leoni C, Aschenbrenner D, Becattini S, Latorre D, Lanzavecchia A, Sallusto F, Monticelli S. ERK phosphorylation and miR-181a expression modulate activation of human memory TH17 cells. Nat Commun 2015; 6:6431. [DOI: 10.1038/ncomms7431] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 01/28/2015] [Indexed: 12/19/2022] Open
|
170
|
Andreou I, Sun X, Stone PH, Edelman ER, Feinberg MW. miRNAs in atherosclerotic plaque initiation, progression, and rupture. Trends Mol Med 2015; 21:307-18. [PMID: 25771097 DOI: 10.1016/j.molmed.2015.02.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 01/28/2015] [Accepted: 02/10/2015] [Indexed: 02/03/2023]
Abstract
Atherosclerosis is a chronic immune-inflammatory disorder that integrates multiple cell types and a diverse set of inflammatory mediators. miRNAs are emerging as important post-transcriptional regulators of gene expression in most, if not all, vertebrate cells, and constitute central players in many physiological and pathological processes. Rapidly accumulating experimental studies reveal their key role in cellular and molecular processes related to the development of atherosclerosis. We review current evidence for the involvement of miRNAs in early atherosclerotic lesion formation and in plaque rupture and erosion. We conclude with a perspective on the clinical relevance, therapeutic opportunities, and future challenges of miRNA biology in understanding the pathogenesis of this complex disease.
Collapse
Affiliation(s)
- Ioannis Andreou
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xinghui Sun
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter H Stone
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elazer R Edelman
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mark W Feinberg
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
171
|
Pratama A, Srivastava M, Williams NJ, Papa I, Lee SK, Dinh XT, Hutloff A, Jordan MA, Zhao JL, Casellas R, Athanasopoulos V, Vinuesa CG. MicroRNA-146a regulates ICOS-ICOSL signalling to limit accumulation of T follicular helper cells and germinal centres. Nat Commun 2015; 6:6436. [PMID: 25743066 PMCID: PMC4366510 DOI: 10.1038/ncomms7436] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/28/2015] [Indexed: 12/22/2022] Open
Abstract
Tight control of T follicular helper (Tfh) cells is required for optimal maturation of the germinal centre (GC) response. The molecular mechanisms controlling Tfh-cell differentiation remain incompletely understood. Here we show that microRNA-146a (miR-146a) is highly expressed in Tfh cells and peak miR-146a expression marks the decline of the Tfh response after immunization. Loss of miR-146a causes cell-intrinsic accumulation of Tfh and GC B cells. MiR-146a represses several Tfh-cell-expressed messenger RNAs, and of these, ICOS is the most strongly cell autonomously upregulated target in miR-146a-deficient T cells. In addition, miR-146a deficiency leads to increased ICOSL expression on GC B cells and antigen-presenting cells. Partial blockade of ICOS signalling, either by injections of low dose of ICOSL blocking antibody or by halving the gene dose of Icos in miR-146a-deficient T cells, prevents the Tfh and GC B-cell accumulation. Collectively, miR-146a emerges as a post-transcriptional brake to limit Tfh cells and GC responses. Maturation of antibody-producing B cells in germinal centers is orchestrated by T follicular helper cells. Here Pratama et al. show that miR-146a negatively regulates T follicular helper cells by targeting ICOS-ICOS ligand signaling in germinal centers.
Collapse
Affiliation(s)
- Alvin Pratama
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| | - Monika Srivastava
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| | - Naomi J Williams
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| | - Ilenia Papa
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| | - Sau K Lee
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| | - Xuyen T Dinh
- Comparative Genomics Centre, James Cook University, Townsville, Queensland 4811, Australia
| | - Andreas Hutloff
- Chronic Immune Reactions Group, German Rheumatism Research Centre Berlin (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Margaret A Jordan
- Comparative Genomics Centre, James Cook University, Townsville, Queensland 4811, Australia
| | - Jimmy L Zhao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Rafael Casellas
- Genomics and Immunity Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Vicki Athanasopoulos
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| | - Carola G Vinuesa
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Canberra, Australian Capital Territory 0200, Australia
| |
Collapse
|
172
|
Yang M, Ye L, Wang B, Gao J, Liu R, Hong J, Wang W, Gu W, Ning G. Decreased miR-146 expression in peripheral blood mononuclear cells is correlated with ongoing islet autoimmunity in type 1 diabetes patients 1miR-146. J Diabetes 2015; 7:158-65. [PMID: 24796653 DOI: 10.1111/1753-0407.12163] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/26/2014] [Accepted: 04/27/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1D) is a common autoimmune disease mediated by autoimmune attack against pancreatic β-cells. It has been reported that dysregulation of microRNAs (miRNAs) may contribute to the pathogenesis of autoimmune diseases, including T1D. The aim of the present study was to identify pathogenic miRNAs in peripheral blood mononuclear cells (PBMC) of T1D patients. METHODS Global miRNA and mRNA expression was profiled in PBMC from 12 patients with newly diagnosed T1D and 10 normal controls. Differently expressed miRNAs were validated in an independent set of patients and controls. The dynamic changes in miRNA and target gene expression were analyzed in T1D patients treated with either a short (6 months) or long (12-24 months) course of insulin. The association between miRNA expression and serum glutamic acid decarboxylase antibody (GADA) titers was also investigated. RESULTS Compared with normal controls, there were 26 miRNAs and 1218 genes differently expressed in PBMC of patients with newly diagnosed T1D. The greatest downregulation was for miR-146a (48% decrease; P < 0.05). Expression of its target genes, predicted to be tumor necrosis factor receptor-associated factor 6 (TRAF6), B cell CLL/lymphoma 11A (BCL11A), syntaxin 3 (STX3) and numb homolog (NUMB), was upregulated. Moreover, T1D patients on long-course insulin and optimized glucose control had sustained low expression of miR-146. Interestingly, decreased miR-146a expression was significantly associated with high serum GADA titers (P < 0.05). CONCLUSIONS The results suggest that dysregulation of miR-146 expression in PBMC may be associated with the ongoing autoimmune imbalance in T1D patients.
Collapse
Affiliation(s)
- Minglan Yang
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, School of Medicine, Shanghai Key Laboratory for Endocrine Tumors, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai JiaoTong University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Brain endothelial miR-146a negatively modulates T-cell adhesion through repressing multiple targets to inhibit NF-κB activation. J Cereb Blood Flow Metab 2015; 35:412-23. [PMID: 25515214 PMCID: PMC4348377 DOI: 10.1038/jcbfm.2014.207] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 09/19/2014] [Accepted: 10/20/2014] [Indexed: 12/24/2022]
Abstract
Pro-inflammatory cytokine-induced activation of nuclear factor, NF-κB has an important role in leukocyte adhesion to, and subsequent migration across, brain endothelial cells (BECs), which is crucial for the development of neuroinflammatory disorders such as multiple sclerosis (MS). In contrast, microRNA-146a (miR-146a) has emerged as an anti-inflammatory molecule by inhibiting NF-κB activity in various cell types, but its effect in BECs during neuroinflammation remains to be evaluated. Here, we show that miR-146a was upregulated in microvessels of MS-active lesions and the spinal cord of mice with experimental autoimmune encephalomyelitis. In vitro, TNFα and IFNγ treatment of human cerebral microvascular endothelial cells (hCMEC/D3) led to upregulation of miR-146a. Brain endothelial overexpression of miR-146a diminished, whereas knockdown of miR-146a augmented cytokine-stimulated adhesion of T cells to hCMEC/D3 cells, nuclear translocation of NF-κB, and expression of adhesion molecules in hCMEC/D3 cells. Furthermore, brain endothelial miR-146a modulates NF-κB activity upon cytokine activation through targeting two novel signaling transducers, RhoA and nuclear factor of activated T cells 5, as well as molecules previously identified, IL-1 receptor-associated kinase 1, and TNF receptor-associated factor 6. We propose brain endothelial miR-146a as an endogenous NF-κB inhibitor in BECs associated with decreased leukocyte adhesion during neuroinflammation.
Collapse
|
174
|
Next-generation sequencing identifies deregulation of microRNAs involved in both innate and adaptive immune response in ALK+ ALCL. PLoS One 2015; 10:e0117780. [PMID: 25688981 PMCID: PMC4331429 DOI: 10.1371/journal.pone.0117780] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/02/2015] [Indexed: 12/21/2022] Open
Abstract
Anaplastic large cell lymphoma (ALCL) is divided into two systemic diseases according to the expression of the anaplastic lymphoma kinase (ALK). We investigated the differential expression of miRNAs between ALK+ ALCL, ALK- ALCL cells and normal T-cells using next generation sequencing (NGS). In addition, a C/EBPβ-dependent miRNA profile was generated. The data were validated in primary ALCL cases. NGS identified 106 miRNAs significantly differentially expressed between ALK+ and ALK- ALCL and 228 between ALK+ ALCL and normal T-cells. We identified a signature of 56 miRNAs distinguishing ALK+ ALCL, ALK- ALCL and T-cells. The top candidates significant differentially expressed between ALK+ and ALK- ALCL included 5 upregulated miRNAs: miR-340, miR-203, miR-135b, miR-182, miR-183; and 7 downregulated: miR-196b, miR-155, miR-146a, miR-424, miR-503, miR-424*, miR-542-3p. The miR-17-92 cluster was also upregulated in ALK+ cells. Additionally, we identified a signature of 3 miRNAs significantly regulated by the transcription factor C/EBPβ, which is specifically overexpressed in ALK+ ALCL, including the miR-181 family. Of interest, miR-181a, which regulates T-cell differentiation and modulates TCR signalling strength, was significantly downregulated in ALK+ ALCL cases. In summary, our data reveal a miRNA signature linking ALK+ ALCL to a deregulated immune response and may reflect the abnormal TCR antigen expression known in ALK+ ALCL.
Collapse
|
175
|
Kroesen BJ, Teteloshvili N, Smigielska-Czepiel K, Brouwer E, Boots AMH, van den Berg A, Kluiver J. Immuno-miRs: critical regulators of T-cell development, function and ageing. Immunology 2015; 144:1-10. [PMID: 25093579 DOI: 10.1111/imm.12367] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are instrumental to many aspects of immunity, including various levels of T-cell immunity. Over the last decade, crucial immune functions were shown to be regulated by specific miRNAs. These 'immuno-miRs' regulate generic cell biological processes in T cells, such as proliferation and apoptosis, as well as a number of T-cell-specific features that are fundamental to the development, differentiation and function of T cells. In this review, we give an overview of the current literature with respect to the role of miRNAs at various stages of T-cell development, maturation, differentiation, activation and ageing. Little is known about the involvement of miRNAs in thymic T-cell development, although miR-181a and miR-150 have been implicated herein. In contrast, several broadly expressed miRNAs including miR-21, miR-155 and miR-17~92, have now been shown to regulate T-cell activation. Other miRNAs, including miR-146a, show a more T-cell-subset-specific expression pattern and are involved in the regulation of processes unique to that specific T-cell subset. Importantly, differences in the miRNA target gene repertoires of different T-cell subsets allow similar miRNAs to control different T-cell-subset-specific functions. Interestingly, several of the here described immuno-miRs have also been implicated in T-cell ageing and there are clear indications for causal involvement of miRNAs in immunosenescence. It is concluded that immuno-miRs have a dynamic regulatory role in many aspects of T-cell differentiation, activation, function and ageing. An important notion when studying miRNAs in relation to T-cell biology is that specific immuno-miRs may have quite unrelated functions in closely related T-cell subsets.
Collapse
Affiliation(s)
- Bart-Jan Kroesen
- Department of Laboratory Medicine, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
176
|
Genetic engineering of hematopoietic stem cells to generate invariant natural killer T cells. Proc Natl Acad Sci U S A 2015; 112:1523-8. [PMID: 25605948 DOI: 10.1073/pnas.1424877112] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Invariant natural killer T (iNKT) cells comprise a small population of αβ T lymphocytes. They bridge the innate and adaptive immune systems and mediate strong and rapid responses to many diseases, including cancer, infections, allergies, and autoimmunity. However, the study of iNKT cell biology and the therapeutic applications of these cells are greatly limited by their small numbers in vivo (∼0.01-1% in mouse and human blood). Here, we report a new method to generate large numbers of iNKT cells in mice through T-cell receptor (TCR) gene engineering of hematopoietic stem cells (HSCs). We showed that iNKT TCR-engineered HSCs could generate a clonal population of iNKT cells. These HSC-engineered iNKT cells displayed the typical iNKT cell phenotype and functionality. They followed a two-stage developmental path, first in thymus and then in the periphery, resembling that of endogenous iNKT cells. When tested in a mouse melanoma lung metastasis model, the HSC-engineered iNKT cells effectively protected mice from tumor metastasis. This method provides a powerful and high-throughput tool to investigate the in vivo development and functionality of clonal iNKT cells in mice. More importantly, this method takes advantage of the self-renewal and longevity of HSCs to generate a long-term supply of engineered iNKT cells, thus opening up a new avenue for iNKT cell-based immunotherapy.
Collapse
|
177
|
Abstract
MicroRNA (miR)-146a-deficient mice develop low-grade, chronic, and systemic inflammation, similar to inflammaging. Hu et al. (2014) demonstrate that the lack of miR-155 prevents the accumulation of Tfh cells and inflammaging in miR-146a-deficient mice. They identify Fosl2 as a functionally important target of miR-155 in Tfh cells.
Collapse
|
178
|
Hu R, Kagele DA, Huffaker TB, Runtsch MC, Alexander M, Liu J, Bake E, Su W, Williams MA, Rao DS, Möller T, Garden GA, Round JL, O'Connell RM. miR-155 promotes T follicular helper cell accumulation during chronic, low-grade inflammation. Immunity 2015; 41:605-19. [PMID: 25367574 DOI: 10.1016/j.immuni.2014.09.015] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 09/12/2014] [Indexed: 12/23/2022]
Abstract
Chronic inflammation is a contributing factor to most life-shortening human diseases. However, the molecular and cellular mechanisms that sustain chronic inflammatory responses remain poorly understood, making it difficult to treat this deleterious condition. Using a mouse model of age-dependent inflammation that results from a deficiency in miR-146a, we demonstrate that miR-155 contributed to the progressive inflammatory disease that emerged as Mir146a(-/-) mice grew older. Upon analyzing lymphocytes from inflamed versus healthy middle-aged mice, we found elevated numbers of T follicular helper (Tfh) cells, germinal center (GC) B cells, and autoantibodies, all occurring in a miR-155-dependent manner. Further, Cd4-cre Mir155(fl/fl) mice were generated and demonstrated that miR-155 functions in T cells, in addition to its established role in B cells, to promote humoral immunity in a variety of contexts. Taken together, our study discovers that miR-146a and miR-155 counterregulate Tfh cell development that drives aberrant GC reactions during chronic inflammation.
Collapse
Affiliation(s)
- Ruozhen Hu
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Dominique A Kagele
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Thomas B Huffaker
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Marah C Runtsch
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Margaret Alexander
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Jin Liu
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Erin Bake
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Wei Su
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Matthew A Williams
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas Möller
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, 4280 JMRB, 15 North Medical Dr. East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
179
|
Abstract
During an immune response, CD8(+)T cells can differentiate into multiple types of effector and memory cells that are important components of immune surveillance. However, their dysregulation has been implicated in infection with viruses or intracellular bacteria and tumorigenesis. miRNAs have been identified as crucial regulators of gene expression, and they perform this function by repressing specific target genes at the post-transcriptional level. Most miRNAs expressed in a given cell type serve the function to impede broadly cell-type-inappropriate gene expression and potently deepen a pre-existing differentiation program. It is increasingly recognized that miRNAs directly modulate the concentration of many regulatory proteins that are required for the development of immune cells in the thymus and their responses in the periphery. This review outlines our current understanding of the function of miRNAs in CD8(+)T cell biology as it impacts expression of protein-coding genes in the context of proper development, infection, as well as oncogenesis. In addition, we conclude with a perspective on future challenges and the clinical relevance of miRNA biology.
Collapse
Affiliation(s)
- Yan Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| |
Collapse
|
180
|
Inducible RNAi in vivo reveals that the transcription factor BATF is required to initiate but not maintain CD8+ T-cell effector differentiation. Proc Natl Acad Sci U S A 2014; 112:512-7. [PMID: 25548173 DOI: 10.1073/pnas.1413291112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The differentiation of effector CD8(+) T cells is critical for the development of protective responses to pathogens and for effective vaccines. In the first few hours after activation, naive CD8(+) T cells initiate a transcriptional program that leads to the formation of effector and memory T cells, but the regulation of this process is poorly understood. Investigating the role of specific transcription factors (TFs) in determining CD8(+) effector T-cell fate by gene knockdown with RNAi is challenging because naive T cells are refractory to transduction with viral vectors without extensive ex vivo stimulation, which obscures the earliest events in effector differentiation. To overcome this obstacle, we developed a novel strategy to test the function of genes in naive CD8(+) T cells in vivo by creating bone marrow chimera from hematopoietic progenitors transduced with an inducible shRNA construct. Following hematopoietic reconstitution, this approach allowed inducible in vivo gene knockdown in any cell type that developed from this transduced progenitor pool. We demonstrated that lentivirus-transduced progenitor cells could reconstitute normal hematopoiesis and develop into naive CD8(+) T cells that were indistinguishable from wild-type naive T cells. This experimental system enabled induction of efficient gene knockdown in vivo without subsequent manipulation. We applied this strategy to show that the TF BATF is essential for initial commitment of naive CD8(+) T cells to effector development but becomes dispensable by 72h. This approach makes possible the study of gene function in vivo in unperturbed cells of hematopoietic origin that are refractory to viral transduction.
Collapse
|
181
|
Park H, Huang X, Lu C, Cairo MS, Zhou X. MicroRNA-146a and microRNA-146b regulate human dendritic cell apoptosis and cytokine production by targeting TRAF6 and IRAK1 proteins. J Biol Chem 2014; 290:2831-41. [PMID: 25505246 DOI: 10.1074/jbc.m114.591420] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We have previously reported 27 differentially expressed microRNAs (miRNAs) during human monocyte differentiation into immature dendritic cells (imDCs) and mature DCs (mDCs). However, their roles in DC differentiation and function remain largely elusive. Here, we report that microRNA (miR)-146a and miR-146b modulate DC apoptosis and cytokine production. Expression of miR-146a and miR-146b was significantly increased upon monocyte differentiation into imDCs and mDCs. Silencing of miR-146a and/or miR-146b in imDCs and mDCs significantly prevented DC apoptosis, whereas overexpressing miR-146a and/or miR-146b increased DC apoptosis. miR-146a and miR-146b expression in imDCs and mDCs was inversely correlated with TRAF6 and IRAK1 expression. Furthermore, siRNA silencing of TRAF6 and/or IRAK1 in imDCs and mDCs enhanced DC apoptosis. By contrast, lentivirus overexpression of TRAF6 and/or IRAK1 promoted DC survival. Moreover, silencing of miR-146a and miR-146b expression had little effect on DC maturation but enhanced IL-12p70, IL-6, and TNF-α production as well as IFN-γ production by IL-12p70-mediated activation of natural killer cells, whereas miR-146a and miR-146b overexpression in mDCs reduced cytokine production. Silencing of miR-146a and miR-146b in DCs also down-regulated NF-κB inhibitor IκBα and increased Bcl-2 expression. Our results identify a new negative feedback mechanism involving the miR-146a/b-TRAF6/IRAK1-NF-κB axis in promoting DC apoptosis.
Collapse
Affiliation(s)
- Haein Park
- From the Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, and
| | - Xin Huang
- From the Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, and
| | - Changming Lu
- the Institute of Oral Health, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35233
| | - Mitchell S Cairo
- From the Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, and the Departments of Microbiology and Immunology, Cell Biology and Anatomy, Pathology, and Medicine, New York Medical College, Valhalla, New York 10595 and
| | - Xianzheng Zhou
- From the Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, and the Departments of Microbiology and Immunology, Cell Biology and Anatomy,
| |
Collapse
|
182
|
Cheng HS, Njock MS, Khyzha N, Dang LT, Fish JE. Noncoding RNAs regulate NF-κB signaling to modulate blood vessel inflammation. Front Genet 2014; 5:422. [PMID: 25540650 PMCID: PMC4261819 DOI: 10.3389/fgene.2014.00422] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 11/17/2014] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular diseases such as atherosclerosis are one of the leading causes of morbidity and mortality worldwide. The clinical manifestations of atherosclerosis, which include heart attack and stroke, occur several decades after initiation of the disease and become more severe with age. Inflammation of blood vessels plays a prominent role in atherogenesis. Activation of the endothelium by inflammatory mediators leads to the recruitment of circulating inflammatory cells, which drives atherosclerotic plaque formation and progression. Inflammatory signaling within the endothelium is driven predominantly by the pro-inflammatory transcription factor, NF-κB. Interestingly, activation of NF-κB is enhanced during the normal aging process and this may contribute to the development of cardiovascular disease. Importantly, studies utilizing mouse models of vascular inflammation and atherosclerosis are uncovering a network of noncoding RNAs, particularly microRNAs, which impinge on the NF-κB signaling pathway. Here we summarize the literature regarding the control of vascular inflammation by microRNAs, and provide insight into how these microRNA-based pathways might be harnessed for therapeutic treatment of disease. We also discuss emerging areas of endothelial cell biology, including the involvement of long noncoding RNAs and circulating microRNAs in the control of vascular inflammation.
Collapse
Affiliation(s)
- Henry S Cheng
- Toronto General Research Institute, University Health Network Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada ; Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research Toronto, ON, Canada
| | - Makon-Sébastien Njock
- Toronto General Research Institute, University Health Network Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada ; Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research Toronto, ON, Canada
| | - Nadiya Khyzha
- Toronto General Research Institute, University Health Network Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada ; Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research Toronto, ON, Canada
| | - Lan T Dang
- Toronto General Research Institute, University Health Network Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada ; Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research Toronto, ON, Canada
| | - Jason E Fish
- Toronto General Research Institute, University Health Network Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada ; Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research Toronto, ON, Canada
| |
Collapse
|
183
|
Möhnle P, Schütz SV, van der Heide V, Hübner M, Luchting B, Sedlbauer J, Limbeck E, Hinske LC, Briegel J, Kreth S. MicroRNA-146a controls Th1-cell differentiation of human CD4+ T lymphocytes by targeting PRKCε. Eur J Immunol 2014; 45:260-72. [PMID: 25308712 DOI: 10.1002/eji.201444667] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 08/28/2014] [Accepted: 10/07/2014] [Indexed: 12/31/2022]
Abstract
T-cell functions must be tightly controlled to keep the balance between vital proinflammatory activity and detrimental overactivation. MicroRNA-146a (miR-146a) has been identified as a key negative regulator of T-cell responses in mice. Its role in human T cells and its relevance to human inflammatory disease, however, remains poorly defined. In this study, we have characterized miR-146a-driven pathways in primary human T cells. Our results identify miR-146a as a critical gatekeeper of Th1-cell differentiation processes acting via molecular mechanisms not uncovered so far. MiR-146a targets protein kinase C epsilon (PRKCε), which is part of a functional complex consisting of PRKCε and signal transducer and activator of transcription 4 (STAT4). Within this complex, PRKCε phosphorylates STAT4, which in turn is capable of promoting Th1-cell differentiation processes in human CD4(+) T lymphocytes. In addition, we observed that T cells of sepsis patients had reduced levels of miR-146a and an increased PRKCε expression in the initial hyperinflammatory phase of the disease. Collectively, our results identify miR-146a as a potent inhibitor of Th1-cell differentiation in human T cells and suggest that dysregulation of miR-146a contributes to the pathogenesis of sepsis.
Collapse
Affiliation(s)
- Patrick Möhnle
- Research Group Molecular Medicine, Department of Anesthesiology, University of Munich (LMU), Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Khalaj M, Tavakkoli M, Stranahan AW, Park CY. Pathogenic microRNA's in myeloid malignancies. Front Genet 2014; 5:361. [PMID: 25477897 PMCID: PMC4237136 DOI: 10.3389/fgene.2014.00361] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/27/2014] [Indexed: 12/21/2022] Open
Abstract
Recent studies have significantly improved our understanding of the role microRNAs (miRNAs) play in regulating normal hematopoiesis. miRNAs are critical for maintaining hematopoietic stem cell function and the development of mature progeny. Thus, perhaps it is not surprising that miRNAs serve as oncogenes and tumor suppressors in hematologic malignancies arising from hematopoietic stem and progenitor cells, such as the myeloid disorders. A number of studies have extensively documented the widespread dysregulation of miRNA expression in human acute myeloid leukemia (AML), inspiring numerous explorations of the functional role of miRNAs in myeloid leukemogenesis. While these investigations have confirmed that a large number of miRNAs exhibit altered expression in AML, only a small fraction has been confirmed as functional mediators of AML development or maintenance. Herein, we summarize the miRNAs for which strong experimental evidence supports their functional roles in AML pathogenesis. We also discuss the implications of these studies on the development of miRNA-directed therapies in AML.
Collapse
Affiliation(s)
- Mona Khalaj
- Weill Graduate School of Medical Sciences, Cornell University NY, USA ; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA
| | - Montreh Tavakkoli
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA
| | - Alec W Stranahan
- Weill Graduate School of Medical Sciences, Cornell University NY, USA ; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA
| | - Christopher Y Park
- Weill Graduate School of Medical Sciences, Cornell University NY, USA ; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA ; Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center NY, USA
| |
Collapse
|
185
|
Sheppard HM, Verdon D, Brooks AES, Feisst V, Ho YYJ, Lorenz N, Fan V, Birch NP, Didsbury A, Dunbar PR. MicroRNA regulation in human CD8+ T cell subsets--cytokine exposure alone drives miR-146a expression. J Transl Med 2014; 12:292. [PMID: 25331734 PMCID: PMC4219087 DOI: 10.1186/s12967-014-0292-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/08/2014] [Indexed: 12/21/2022] Open
Abstract
Background microRNAs (miRNAs) are emerging as key regulators of the immune system, but their role in CD8+ T cell differentiation is not well explored. Some evidence suggests that signals from cell surface receptors influence the expression of miRNAs in CD8+ T cells, and may have consequent effects on cell phenotype and function. We set out to investigate whether common gamma chain cytokines modulated human CD8+ T cell expression of miR-146a, which previous studies have associated with different stages of CD8+ differentiation. We also investigated how changes in miR-146a related to other miRNAs that alter with CD8+ differentiation status. Methods We treated human CD8+ T cells with the cytokines IL-2, IL-7 or IL-15 either at rest or after stimulation with anti-CD3 and anti-CD28. For some experiments we also purified human CD8+ T cell subsets ex vivo. Flow cytometry was used in parallel to assess cell surface memory marker expression. Total RNA from these cells was subjected to microarray analysis and real-time PCR for miRNA expression. Nucleofection studies were performed to assess potential mRNA targets of miR-146a. Results We find that miR-146a is up-regulated in naïve CD8+ T cells exposed to IL-2 or IL-15, even in the absence of an activating T cell receptor stimulus, but not when IL-7 is also present. miR-146a expression correlates with a memory phenotype in both ex vivo and in vitro cultured cells although in our hands overexpression of miR-146a was not sufficient alone to drive a full memory phenotype. In ex vivo analysis, miR-146a was one of a small number of miRNAs that was differentially expressed between naïve and memory CD8+ T cells. Conclusions miR-146a is emerging as a critical regulator of immune system. Our data shows that miR-146a expression is strongly influenced by the cytokine milieu even in the absence of a T cell receptor stimulus. Our results have implications for studies designed to assess the function of miR-146a, help to define a fingerprint of miRNA expression in CD8+ T cell subsets and may be useful when designing optimal protocols for T cell expansion as efficacy of T cell immunotherapy is correlated with an ‘early’ memory phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0292-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hilary M Sheppard
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, NZ, New Zealand.
| | - Daniel Verdon
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, NZ, New Zealand.
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, NZ, New Zealand.
| | - Vaughan Feisst
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, NZ, New Zealand.
| | - Yu-Yu Joyce Ho
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, NZ, New Zealand.
| | - Natalie Lorenz
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand.
| | - Vicky Fan
- Bioinformatics Institute, University of Auckland, Auckland, NZ, New Zealand.
| | - Nigel P Birch
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Centre for Brain Research, University of Auckland, Auckland, NZ, New Zealand.
| | - Alicia Didsbury
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand.
| | - P Rod Dunbar
- School of Biological Sciences, University of Auckland, Thomas Building, Auckland, NZ, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, NZ, New Zealand.
| |
Collapse
|
186
|
Abstract
Key Points
This study deciphers the regulatory role of miR-146a during GVHD in mice. In humans, the minor genotype of the SNP rs2910164, which reduces expression of miR-146a, was associated with higher GVHD severity.
Collapse
|
187
|
Marcais A, Blevins R, Graumann J, Feytout A, Dharmalingam G, Carroll T, Amado IF, Bruno L, Lee K, Walzer T, Mann M, Freitas AA, Boothby M, Fisher AG, Merkenschlager M. microRNA-mediated regulation of mTOR complex components facilitates discrimination between activation and anergy in CD4 T cells. ACTA ACUST UNITED AC 2014; 211:2281-95. [PMID: 25311506 PMCID: PMC4203951 DOI: 10.1084/jem.20132059] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Using Dicer-deficient CD4 T cells, Marcais et al. show that microRNAs regulate the expression of mTOR components that are needed to discriminate between activating and anergy-inducing stimuli. T cell receptor (TCR) signals can elicit full activation with acquisition of effector functions or a state of anergy. Here, we ask whether microRNAs affect the interpretation of TCR signaling. We find that Dicer-deficient CD4 T cells fail to correctly discriminate between activating and anergy-inducing stimuli and produce IL-2 in the absence of co-stimulation. Excess IL-2 production by Dicer-deficient CD4 T cells was sufficient to override anergy induction in WT T cells and to restore inducible Foxp3 expression in Il2-deficient CD4 T cells. Phosphorylation of Akt on S473 and of S6 ribosomal protein was increased and sustained in Dicer-deficient CD4 T cells, indicating elevated mTOR activity. The mTOR components Mtor and Rictor were posttranscriptionally deregulated, and the microRNAs Let-7 and miR-16 targeted the Mtor and Rictor mRNAs. Remarkably, returning Mtor and Rictor to normal levels by deleting one allele of Mtor and one allele of Rictor was sufficient to reduce Akt S473 phosphorylation and to reduce co-stimulation–independent IL-2 production in Dicer-deficient CD4 T cells. These results show that microRNAs regulate the expression of mTOR components in T cells, and that this regulation is critical for the modulation of mTOR activity. Hence, microRNAs contribute to the discrimination between T cell activation and anergy.
Collapse
Affiliation(s)
- Antoine Marcais
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK CIRI, International Center for Infectiology Research, Université de Lyon, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Ecole Normale Supérieure, 69007 Lyon, France
| | - Rory Blevins
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Johannes Graumann
- Department of Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, 82152 Martinsried, Germany Weill Cornell Medical College in Qatar, Qatar Foundation, Education City, Doha, State of Qatar
| | - Amelie Feytout
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Gopuraja Dharmalingam
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Thomas Carroll
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Inês F Amado
- Unité de Biologie des Populations Lymphocytaires, Department of Immunology, Institut Pasteur, and Centre National pour la Recherche Scientifique (Centre National de la Recherche Scientifique), URA1961, 75724 Paris, France Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Ludovica Bruno
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Keunwook Lee
- Department of Pathology, Microbiology, and Immunology and Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Thierry Walzer
- CIRI, International Center for Infectiology Research, Université de Lyon, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Ecole Normale Supérieure, 69007 Lyon, France
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Antonio A Freitas
- Unité de Biologie des Populations Lymphocytaires, Department of Immunology, Institut Pasteur, and Centre National pour la Recherche Scientifique (Centre National de la Recherche Scientifique), URA1961, 75724 Paris, France
| | - Mark Boothby
- Department of Pathology, Microbiology, and Immunology and Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Pathology, Microbiology, and Immunology and Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Amanda G Fisher
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Matthias Merkenschlager
- Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK Lymphocyte Development Group and Epigenetics Section, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| |
Collapse
|
188
|
Shi YL, Weiland M, Lim HW, Mi QS, Zhou L. Serum miRNA expression profiles change in autoimmune vitiligo in mice. Exp Dermatol 2014; 23:140-2. [PMID: 24401108 DOI: 10.1111/exd.12319] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2014] [Indexed: 12/18/2022]
Abstract
It is widely believed that non-segmental vitiligo results from the autoimmune destruction of melanocytes. MicroRNAs (miRNAs), a class of small non-coding RNAs that negatively regulate gene expression, are involved in the immune cell development and function and regulate the development of autoimmune diseases. Recent studies demonstrate that functional miRNAs can be detected in the serum and serve as biomarkers of various diseases. In the present study, we used a mouse autoimmune vitiligo model, in which melanocyte autoreactive CD4+ T cells were adoptively transferred into Rag1(-/-) host mice. Serum miRNA expression was profiled in vitiligo developed mice and control mice using TaqMan RT-PCR arrays. We have found that the expressions of 20 serum miRNAs were changed in vitiligo mice compared to control mice. Three increased miRNAs, miR-146a, miR-191, and miR-342-3p, were further confirmed by a single TaqMan RT-PCR. Our findings suggest that miRNAs may be involved in vitiligo development and serum miRNAs could serve as serum biomarkers for vitiligo in mice.
Collapse
Affiliation(s)
- Yu-Ling Shi
- Henry Ford Immunology Program, Henry Ford Hospital, Detroit, MI, USA; Multicultural Dermatology Center, Department of Dermatology, Henry Ford Hospital, Detroit, MI, USA; Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
189
|
Pivarcsi A, Ståhle M, Sonkoly E. Genetic polymorphisms altering microRNA activity in psoriasis - a key to solve the puzzle of missing heritability? Exp Dermatol 2014; 23:620-4. [DOI: 10.1111/exd.12469] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Andor Pivarcsi
- Unit of Dermatology and Venerology; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| | - Mona Ståhle
- Unit of Dermatology and Venerology; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| | - Enikő Sonkoly
- Unit of Dermatology and Venerology; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
190
|
Shih HY, Sciumè G, Poholek AC, Vahedi G, Hirahara K, Villarino AV, Bonelli M, Bosselut R, Kanno Y, Muljo SA, O'Shea JJ. Transcriptional and epigenetic networks of helper T and innate lymphoid cells. Immunol Rev 2014; 261:23-49. [PMID: 25123275 PMCID: PMC4321863 DOI: 10.1111/imr.12208] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of the specification of CD4(+) helper T cells to discrete effector 'lineages' represented a watershed event in conceptualizing mechanisms of host defense and immunoregulation. However, our appreciation for the actual complexity of helper T-cell subsets continues unabated. Just as the Sami language of Scandinavia has 1000 different words for reindeer, immunologists recognize the range of fates available for a CD4(+) T cell is numerous and may be underestimated. Added to the crowded scene for helper T-cell subsets is the continuously growing family of innate lymphoid cells (ILCs), endowed with common effector responses and the previously defined 'master regulators' for CD4(+) helper T-cell subsets are also shared by ILC subsets. Within the context of this extraordinary complexity are concomitant advances in the understanding of transcriptomes and epigenomes. So what do terms like 'lineage commitment' and helper T-cell 'specification' mean in the early 21st century? How do we put all of this together in a coherent conceptual framework? It would be arrogant to assume that we have a sophisticated enough understanding to seriously answer these questions. Instead, we review the current status of the flexibility of helper T-cell responses in relation to their genetic regulatory networks and epigenetic landscapes. Recent data have provided major surprises as to what master regulators can or cannot do, how they interact with other transcription factors and impact global genome-wide changes, and how all these factors come together to influence helper cell function.
Collapse
Affiliation(s)
- Han-Yu Shih
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Transcriptomics identified a critical role for Th2 cell-intrinsic miR-155 in mediating allergy and antihelminth immunity. Proc Natl Acad Sci U S A 2014; 111:E3081-90. [PMID: 25024218 DOI: 10.1073/pnas.1406322111] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Allergic diseases, orchestrated by hyperactive CD4(+) Th2 cells, are some of the most common global chronic diseases. Therapeutic intervention relies upon broad-scale corticosteroids with indiscriminate impact. To identify targets in pathogenic Th2 cells, we took a comprehensive approach to identify the microRNA (miRNA) and mRNA transcriptome of highly purified cytokine-expressing Th1, Th2, Th9, Th17, and Treg cells both generated in vitro and isolated ex vivo from allergy, infection, and autoimmune disease models. We report here that distinct regulatory miRNA networks operate to regulate Th2 cells in house dust mite-allergic or helminth-infected animals and in vitro Th2 cells, which are distinguishable from other T cells. We validated several miRNA (miR) candidates (miR-15a, miR-20b, miR-146a, miR-155, and miR-200c), which targeted a suite of dynamically regulated genes in Th2 cells. Through in-depth studies using miR-155(-/-) or miR-146a(-/-) T cells, we identified that T-cell-intrinsic miR-155 was required for type-2 immunity, in part through regulation of S1pr1, whereas T-cell-intrinsic miR-146a was required to prevent overt Th1/Th17 skewing. These data identify miR-155, but not miR-146a, as a potential therapeutic target to alleviate Th2-medited inflammation and allergy.
Collapse
|
192
|
Zhao JL, Starczynowski DT. Role of microRNA-146a in normal and malignant hematopoietic stem cell function. Front Genet 2014; 5:219. [PMID: 25071842 PMCID: PMC4087671 DOI: 10.3389/fgene.2014.00219] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 06/24/2014] [Indexed: 01/12/2023] Open
Abstract
Regulation of hematopoiesis is controlled by microRNAs (miRNAs). In this review, we focus on miR-146a, and its role in regulating normal and malignant hematopoiesis. miR-146a is a negative regulator of immune cell activation by repressing two targets, TRAF6 and IRAK1. Genetic deletion of miR-146a confirmed a role of miR-146a during innate immune signaling as well as for hematopoietic stem cell function. miR-146a is also implicated in the pathogenesis of human myelodysplastic syndromes (MDSs) as it is located within a commonly deleted region on chromosome 5, and miR-146a-deficient mice exhibit features of an MDS-like disease. With new insight into miR-146a through genetic and expression analyses, we highlight and discuss the recent advances in the understanding of miR-146a in physiological hematopoiesis during steady-state and inflammation, as well as in MDS.
Collapse
Affiliation(s)
- Jimmy L Zhao
- Division of Biology and Biological Engineering, California Institute of Technology Pasadena, CA, USA ; David Geffen School of Medicine, University of California Los Angeles, CA, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA ; Department of Cancer Biology, University of Cincinnati Cincinnati, OH, USA
| |
Collapse
|
193
|
Rebane A, Runnel T, Aab A, Maslovskaja J, Rückert B, Zimmermann M, Plaas M, Kärner J, Treis A, Pihlap M, Haljasorg U, Hermann H, Nagy N, Kemeny L, Erm T, Kingo K, Li M, Boldin MP, Akdis CA. MicroRNA-146a alleviates chronic skin inflammation in atopic dermatitis through suppression of innate immune responses in keratinocytes. J Allergy Clin Immunol 2014; 134:836-847.e11. [PMID: 24996260 DOI: 10.1016/j.jaci.2014.05.022] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/24/2014] [Accepted: 05/06/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chronic skin inflammation in atopic dermatitis (AD) is associated with elevated expression of proinflammatory genes and activation of innate immune responses in keratinocytes. microRNAs (miRNAs) are short, single-stranded RNA molecules that silence genes via the degradation of target mRNAs or inhibition of translation. OBJECTIVE The aim of this study was to investigate the role of miR-146a in skin inflammation in AD. METHODS RNA and protein expression was analyzed using miRNA and mRNA arrays, RT-quantitative PCR, Western blotting, and immunonohistochemistry. Transfection of miR-146a precursors and inhibitors into human primary keratinocytes, luciferase assays, and MC903-dependent mouse model of AD were used to study miR-146a function. RESULTS We show that miR-146a expression is increased in keratinocytes and chronic lesional skin of patients with AD. miR-146a inhibited the expression of numerous proinflammatory factors, including IFN-γ-inducible and AD-associated genes CCL5, CCL8, and ubiquitin D (UBD) in human primary keratinocytes stimulated with IFN-γ, TNF-α, or IL-1β. In a mouse model of AD, miR-146a-deficient mice developed stronger inflammation characterized by increased accumulation of infiltrating cells in the dermis, elevated expression of IFN-γ, CCL5, CCL8, and UBD in the skin, and IFN-γ, IL-1β, and UBD in draining lymph nodes. Both tissue culture and in vivo experiments in mice demonstrated that miR-146a-mediated suppression in allergic skin inflammation partially occurs through direct targeting of upstream nuclear factor kappa B signal transducers caspase recruitment domain-containing protein 10 and IL-1 receptor-associated kinase 1. In addition, human CCL5 was determined as a novel, direct target of miR-146a. CONCLUSION Our data demonstrate that miR-146a controls nuclear factor kappa B-dependent inflammatory responses in keratinocytes and chronic skin inflammation in AD.
Collapse
Affiliation(s)
- Ana Rebane
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
| | - Toomas Runnel
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Institute of Molecular and Cellular Biology, University of Tartu, Tartu, Estonia
| | - Alar Aab
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Julia Maslovskaja
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Maya Zimmermann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Mario Plaas
- Transgenic Technology Core Laboratory, University of Tartu, Tartu, Estonia
| | - Jaanika Kärner
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Angela Treis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Maire Pihlap
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Uku Haljasorg
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Helen Hermann
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Nikoletta Nagy
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; Dermatological Research Group of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Lajos Kemeny
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; Dermatological Research Group of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Triin Erm
- Department of Pathology, Tartu University Hospital, Tartu, Estonia
| | - Külli Kingo
- Department of Dermatology and Venereology, University of Tartu, Tartu, Estonia; Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Mei Li
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université de Strasbourg, Illkirch, France
| | - Mark P Boldin
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, Calif
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| |
Collapse
|
194
|
Lochhead RB, Ma Y, Zachary JF, Baltimore D, Zhao JL, Weis JH, O'Connell RM, Weis JJ. MicroRNA-146a provides feedback regulation of lyme arthritis but not carditis during infection with Borrelia burgdorferi. PLoS Pathog 2014; 10:e1004212. [PMID: 24967703 PMCID: PMC4072785 DOI: 10.1371/journal.ppat.1004212] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 05/13/2014] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs have been shown to be important regulators of inflammatory and immune responses and are implicated in several immune disorders including systemic lupus erythematosus and rheumatoid arthritis, but their role in Lyme borreliosis remains unknown. We performed a microarray screen for expression of miRNAs in joint tissue from three mouse strains infected with Borrelia burgdorferi. This screen identified upregulation of miR-146a, a key negative regulator of NF-κB signaling, in all three strains, suggesting it plays an important role in the in vivo response to B. burgdorferi. Infection of B6 miR-146a-/- mice with B. burgdorferi revealed a critical nonredundant role of miR-146a in modulating Lyme arthritis without compromising host immune response or heart inflammation. The impact of miR-146a was specifically localized to the joint, and did not impact lesion development or inflammation in the heart. Furthermore, B6 miR-146a-/- mice had elevated levels of NF-κB-regulated products in joint tissue and serum late in infection. Flow cytometry analysis of various lineages isolated from infected joint tissue of mice showed that myeloid cell infiltration was significantly greater in B6 miR-146a-/- mice, compared to B6, during B. burgdorferi infection. Using bone marrow-derived macrophages, we found that TRAF6, a known target of miR-146a involved in NF-κB activation, was dysregulated in resting and B. burgdorferi-stimulated B6 miR-146a-/- macrophages, and corresponded to elevated IL-1β, IL-6 and CXCL1 production. This dysregulated protein production was also observed in macrophages treated with IL-10 prior to B. burgdorferi stimulation. Peritoneal macrophages from B6 miR-146a-/- mice also showed enhanced phagocytosis of B. burgdorferi. Together, these data show that miR-146a-mediated regulation of TRAF6 and NF-κB, and downstream targets such as IL-1β, IL-6 and CXCL1, are critical for modulation of Lyme arthritis during chronic infection with B. burgdorferi.
Collapse
Affiliation(s)
- Robert B. Lochhead
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ying Ma
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - James F. Zachary
- Department of Veterinary Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - David Baltimore
- Department of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Jimmy L. Zhao
- Department of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - John H. Weis
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ryan M. O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Janis J. Weis
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
195
|
Abstract
MicroRNAs (miRNAs) are a class of small noncoding RNA which exert post-transcriptional gene regulation activity by targeting messenger RNAs. miRNAs have been found to be involved in various fundamental biological processes and deregulation of miRNAs is known to result in pathological conditions. In this review, we provide an overview of recent discoveries on the role played by this class of molecules in lung development and in pulmonary diseases, such as asthma, cystic fibrosis, chronic obstructive pulmonary disease, and pulmonary artery hypertension. Considering the relevant role of these miRNAs under physiological and pathological conditions, they represent new clinical targets as well as diagnostic and prognostic tools. Therefore, this review pays special attention to recent advances and possible future directions for the use of miRNAs for clinical applications.
Collapse
Affiliation(s)
- Roberto Sessa
- Cardiovascular research institute, University of California San Francisco, CA 94158, USA
| | | |
Collapse
|
196
|
Berrih-Aknin S. Myasthenia Gravis: paradox versus paradigm in autoimmunity. J Autoimmun 2014; 52:1-28. [PMID: 24934596 DOI: 10.1016/j.jaut.2014.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Myasthenia Gravis (MG) is a paradigm of organ-specific autoimmune disease (AID). It is mediated by antibodies that target the neuromuscular junction. The purpose of this review is to place MG in the general context of autoimmunity, to summarize the common mechanisms between MG and other AIDs, and to describe the specific mechanisms of MG. We have chosen the most common organ-specific AIDs to compare with MG: type 1 diabetes mellitus (T1DM), autoimmune thyroid diseases (AITD), multiple sclerosis (MS), some systemic AIDs (systemic lupus erythematous (SLE), rheumatoid arthritis (RA), Sjogren's syndrome (SS)), as well as inflammatory diseases of the gut and liver (celiac disease (CeD), Crohn's disease (CD), and primary biliary cirrhosis (PBC)). Several features are similar between all AIDs, suggesting that common pathogenic mechanisms lead to their development. In this review, we address the predisposing factors (genetic, epigenetic, hormones, vitamin D, microbiota), the triggering components (infections, drugs) and their interactions with the immune system [1,2]. The dysregulation of the immune system is detailed and includes the role of B cells, Treg cells, Th17 and cytokines. We particularly focused on the role of TNF-α and interferon type I whose role in MG is very analogous to that in several other AIDS. The implication of AIRE, a key factor in central tolerance is also discussed. Finally, if MG is a prototype of AIDS, it has a clear specificity compared to the other AIDS, by the fact that the target organ, the muscle, is not the site of immune infiltration and B cell expansion, but exclusively that of antibody-mediated pathogenic mechanisms. By contrast, the thymus in the early onset subtype frequently undergoes tissue remodeling, resulting in the development of ectopic germinal centers surrounded by high endothelial venules (HEV), as observed in the target organs of many other AIDs.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center UM76, F-75013 Paris, France; INSERM U974, F-75013 Paris, France; CNRS FRE 3617, F-75013 Paris, France; Institute of Myology, F-75013 Paris, France.
| |
Collapse
|
197
|
Xu Z, Ho S, Chang CC, Liu Z, Li M, Vasilescu ER, Clynes RA, Vlad G, Suciu-Foca N. ILT3.Fc inhibits the production of exosomes containing inflammatory microRNA in supernatants of alloactivated T cells. Hum Immunol 2014; 75:756-9. [PMID: 24862932 DOI: 10.1016/j.humimm.2014.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 12/12/2013] [Accepted: 05/13/2014] [Indexed: 10/25/2022]
Abstract
Immune activation needs to be tightly regulated to control immune-mediated tissue damage. Inhibitory pathways serve to terminate an immune response and resolve inflammation. Persistent exposure to antigens can drive development of adaptive regulatory cells. Similarly exposure of activated T cells to the recombinant ILT3-Fc molecule during priming triggers the differentiation of CD8 T suppressor cells and the induction of CD4 T helper anergy. Ts express high levels of immunoregulatory signature genes together with low levels of microRNA which control their function. Analysis of microRNA contained by exosomes from cultures in which T cells were alloactivated in the presence or absence of ILT3.Fc, demonstrated that this agent inhibits the release of inflammatory microRNA. The source of such inflammatory microRNA was found to reside in alloactivated CD4 T cells, since exosomes from MLC primed CD4 T cells were shown to diminish the suppressive activity of ILT3-Fc-induced CD8(+) Ts at high effector to suppressor T cell ratios. This indicates that inflammatory exosomes can swing the balance between effector and regulatory T cells in favor of immunity. These data suggest that isolation and characterization of micro-RNA containing exosomes in patients' circulation may be of use for treatment, prevention and monitoring of immune activation.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Sophey Ho
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Chih-Chao Chang
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Zhuoru Liu
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Muyang Li
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Elena R Vasilescu
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Raphael A Clynes
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - George Vlad
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA
| | - Nicole Suciu-Foca
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
198
|
Li K, Du Y, Jiang BL, He JF. Increased microRNA-155 and decreased microRNA-146a may promote ocular inflammation and proliferation in Graves' ophthalmopathy. Med Sci Monit 2014; 20:639-43. [PMID: 24743332 PMCID: PMC3999163 DOI: 10.12659/msm.890686] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Graves' ophthalmopathy is an inflammatory autoimmune disease of the orbit, characterized by inflammation and proliferation of the orbital tissue caused by CD4+T cells and orbital fibroblasts. Despite recent substantial findings regarding its cellular and molecular foundations, the pathogenesis of Graves' ophthalmopathy remains unclear. Accumulating data suggest that microRNAs play important roles in the pathophysiology of autoimmunity and proliferation. Specifically, microRNA-155 (miR-155) can promote autoimmune inflammation by enhancing inflammatory T cell development. In contrast to miR-155, microRNA-146a (miR-146a) can inhibit the immune response by suppressing T cell activation. Furthermore, miR-155 and miR-146a are involved in cell proliferation, differentiation, and many other life processes. Thus, miR-155 and miR-146a, with opposite impacts on inflammatory responses carried out by T lymphocytes, appear to have multiple targets in the pathogenesis of Graves' ophthalmopathy. Our previous work showed that the expression of miR-146a was significantly decreased in peripheral blood mononuclear cells from Graves' ophthalmopathy patients compared with normal subjects. Accordingly, we proposed that the expression of miR-155 increased and the expression of miR-146a decreased in the target cells (CD4+T cells and orbital fibroblasts), thus promoting ocular inflammation and proliferation in Graves' ophthalmopathy. The proposed hypothesis warrants further investigation of the function of the differentially expressed microRNAs, which may shed new light on the pathogenesis of Graves' ophthalmopathy and lead to new strategies for its management.
Collapse
Affiliation(s)
- Kaijun Li
- Department of Ophthalmology, First Affiliated Hospital of Guangxi Medical University, Nanning, China (mainland)
| | - Yi Du
- Department of Ophthalmology, First Affiliated Hospital of Guangxi Medical University, Nanning, China (mainland)
| | - Ben-Li Jiang
- Department of Ophthalmology, First People's Hospital of Nanning, Nanning, China (mainland)
| | - Jian-Feng He
- Department of Ophthalmology, First Affiliated Hospital of Guangxi Medical University, Nanning, China (mainland)
| |
Collapse
|
199
|
Premalignant PTEN-deficient thymocytes activate microRNAs miR-146a and miR-146b as a cellular defense against malignant transformation. Blood 2014; 123:4089-100. [PMID: 24735967 DOI: 10.1182/blood-2013-11-539411] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer develops by a multistep process during which cells acquire characteristics that allow them to evade apoptosis and proliferate unchecked. Sequential acquisition of genetic alterations drives this process but also causes cellular stress, frequently prompting cells to enter a premalignant period during which they mount a defense against transformation. T cell-specific deletion of the tumor suppressor PTEN in mice induces premalignancy in the thymus and development of CD4(+) T-cell lymphomas in the periphery. Here we sought to identify factors mediating the cellular defense against transformation during the premalignant period. We identified several microRNAs upregulated specifically in premalignant thymocytes, including miR-146a, miR-146b, and the miR-183/96/182 cluster. CD4-driven T cell-specific transgenic overexpression of mir-146a and mir-146b significantly delayed PTEN-deficient lymphomagenesis and delayed c-myc oncogene induction, a key driver of transformation in PTEN-deficient T-cell malignancies. We found that miR-146a and miR-146b targeting of Traf6 attenuates TCR signaling in the thymus and inhibits downstream NF-κB-dependent induction of c-myc. Additionally, c-myc repression in mature CD4 T cells by miR-146b impaired TCR-mediated proliferation. Hence, we have identified 2 miRNAs that are upregulated as part of the cellular response against transformation that, when overrepresented, can effectively inhibit progression to malignancy in the context of PTEN deficiency.
Collapse
|
200
|
Lindner JM, Kayo H, Hedlund S, Fukuda Y, Fukao T, Nielsen PJ. Cutting edge: The transcription factor Bob1 counteracts B cell activation and regulates miR-146a in B cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:4483-6. [PMID: 24719463 DOI: 10.4049/jimmunol.1303022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Mice lacking the lymphocyte-specific transcription factor Bob1 (also called OBF-1 or OCA-B) fail to generate germinal centers and a robust Ig response. We show that peripheral B cells in Bob1(-/-) mice bear characteristics of chronically activated or anergic-like B cells and identify the immunosuppressive microRNA-146a, together with other microRNAs, as novel transcriptional targets of Bob1. The inability to restrict B cell signaling could contribute to the immunodeficient phenotype of these mice and is consistent with an important role for Bob1 in suppressing B cell activation in vivo.
Collapse
Affiliation(s)
- John M Lindner
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|