Targeting translational control as a novel way to treat inflammatory disease: the emerging role of MicroRNAs
M. Plank
Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Search for more papers by this authorS. Maltby
Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Search for more papers by this authorJ. Mattes
Priority Research Centre for Asthma and Respiratory Disease, Experimental & Translational Respiratory Group, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Paediatric Respiratory and Sleep Medicine Unit, Newcastle Children's Hospital, Newcastle, NSW, Australia
Search for more papers by this authorCorresponding Author
P. S. Foster
Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Correspondence:
Paul S. Foster, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.
E-mail: [email protected]
Search for more papers by this authorM. Plank
Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Search for more papers by this authorS. Maltby
Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Search for more papers by this authorJ. Mattes
Priority Research Centre for Asthma and Respiratory Disease, Experimental & Translational Respiratory Group, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Paediatric Respiratory and Sleep Medicine Unit, Newcastle Children's Hospital, Newcastle, NSW, Australia
Search for more papers by this authorCorresponding Author
P. S. Foster
Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
Correspondence:
Paul S. Foster, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.
E-mail: [email protected]
Search for more papers by this authorSummary
Chronic inflammatory diseases (e.g. asthma and chronic obstructive pulmonary disease) are leading causes of morbidity and mortality world-wide and effective treatments are limited. These disorders can often be attributed to abnormal immune responses to environmental stimuli and infections. Mechanisms leading to inflammation are complex, resulting from interactions of structural cells and activation of both the adaptive and innate arms of the immune system. The activation of structural and immune cells involves both temporary and permanent changes in gene expression in these cells, which underpin chronic inflammation and tissue dysfunction. miRNAs are small non-coding RNAs increasingly being recognized to play important roles in the post-transcriptional regulation of gene expression in mammalian cells by regulating translation. Individual miRNAs can exert their effects by directly inhibiting the translation or stability of multiple mRNAs simultaneously. Thus, the expression or blockade of function of a single miRNA (miR) can result in pronounced alterations in protein expression within a given cell. Dysregulation of miRNA expression may subsequently alter cellular function, and in certain situations predispose to disease. Our current understanding of the role of miRNA in the regulation of inflammatory disease (e.g. allergic diseases) remains limited. In this review, we provide an overview of the current understanding of miRNA biogenesis and function, the roles miRNA play in the regulation of immune cell function and their potential contribution to inflammatory diseases. We also highlight strategies to alter miRNA function for experimental or therapeutic gain, and discuss the potential utility and limitations of targeting these molecules as anti-inflammatory strategies.
References
- 1Schickel R, Boyerinas B, Park SM, Peter ME. MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene 2008; 27: 5959–74.
- 2Griffiths-Jones S. The microRNA Registry. Nucleic Acids Res 2004; 32: D109–11.
- 3Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res 2006; 34: D140–4.
- 4Weiland M, Gao XH, Zhou L, Mi QS. Small RNAs have a large impact: circulating microRNAs as biomarkers for human diseases. RNA Biol 2012; 9: 850–9.
- 5Lai EC, Tomancak P, Williams RW, Rubin GM. Computational identification of Drosophila microRNA genes. Genome Biol 2003; 4: R42.
- 6Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005; 120: 15–20.
- 7Lim LP, Lau NC, Weinstein EG et al. The microRNAs of Caenorhabditis elegans. Genes Dev 2003; 17: 991–1008.
- 8Bentwich I, Avniel A, Karov Y et al. Identification of hundreds of conserved and nonconserved human microRNAs. Nat Genet 2005; 37: 766–70.
- 9Berezikov E, Guryev V, van de Belt J, Wienholds E, Plasterk RH, Cuppen E. Phylogenetic shadowing and computational identification of human microRNA genes. Cell 2005; 120: 21–4.
- 10Borchert GM, Lanier W, Davidson BL. RNA polymerase III transcribes human microRNAs. Nat Struct Mol Biol 2006; 13: 1097–101.
- 11Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science 2004; 303: 83–6.
- 12Cullen BR. Transcription and processing of human microRNA precursors. Mol Cell 2004; 16: 861–5.
- 13Lee Y, Ahn C, Han J et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003; 425: 415–9.
- 14Landthaler M, Yalcin A, Tuschl T. The human DiGeorge syndrome critical region gene 8 and Its D. melanogaster homolog are required for miRNA biogenesis. Curr Biol 2004; 14: 2162–7.
- 15Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 2003; 17: 3011–6.
- 16Bohnsack MT, Czaplinski K, Gorlich D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 2004; 10: 185–91.
- 17Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science 2004; 303: 95–8.
- 18Grishok A, Pasquinelli AE, Conte D et al. Genes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing. Cell 2001; 106: 23–34.
- 19Hutvagner G, McLachlan J, Pasquinelli AE, Balint E, Tuschl T, Zamore PD. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 2001; 293: 834–8.
- 20Zhang H, Kolb FA, Brondani V, Billy E, Filipowicz W. Human Dicer preferentially cleaves dsRNAs at their termini without a requirement for ATP. EMBO J 2002; 21: 5875–85.
- 21Hammond SM, Boettcher S, Caudy AA, Kobayashi R, Hannon GJ. Argonaute2, a link between genetic and biochemical analyses of RNAi. Science 2001; 293: 1146–50.
- 22Mallory AC, Reinhart BJ, Jones-Rhoades MW et al. MicroRNA control of PHABULOSA in leaf development: importance of pairing to the microRNA 5′ region. EMBO J 2004; 23: 3356–64.
- 23Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell 2003; 115: 787–98.
- 24Baek D, Villen J, Shin C, Camargo FD, Gygi SP, Bartel DP. The impact of microRNAs on protein output. Nature 2008; 455: 64–71.
- 25Selbach M, Schwanhausser B, Thierfelder N, Fang Z, Khanin R, Rajewsky N. Widespread changes in protein synthesis induced by microRNAs. Nature 2008; 455: 58–63.
- 26Nottrott S, Simard MJ, Richter JD. Human let-7a miRNA blocks protein production on actively translating polyribosomes. Nat Struct Mol Biol 2006; 13: 1108–14.
- 27Petersen CP, Bordeleau ME, Pelletier J, Sharp PA. Short RNAs repress translation after initiation in mammalian cells. Mol Cell 2006; 21: 533–42.
- 28Pillai RS, Bhattacharyya SN, Artus CG et al. Inhibition of translational initiation by Let-7 MicroRNA in human cells. Science 2005; 309: 1573–6.
- 29Bagga S, Bracht J, Hunter S et al. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell 2005; 122: 553–63.
- 30Giraldez AJ, Mishima Y, Rihel J et al. Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science 2006; 312: 75–9.
- 31Guo H, Ingolia NT, Weissman JS, Bartel DP. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 2010; 466: 835–40.
- 32Lim LP, Lau NC, Garrett-Engele P et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 2005; 433: 769–73.
- 33Wu L, Fan J, Belasco JG. MicroRNAs direct rapid deadenylation of mRNA. Proc Natl Acad Sci U S A 2006; 103: 4034–9.
- 34Orom UA, Nielsen FC, Lund AH. MicroRNA-10a binds the 5′UTR of ribosomal protein mRNAs and enhances their translation. Mol Cell 2008; 30: 460–71.
- 35Huang S, Wu S, Ding J et al. MicroRNA-181a modulates gene expression of zinc finger family members by directly targeting their coding regions. Nucleic Acids Res 2010; 38: 7211–8.
- 36Lytle JR, Yario TA, Steitz JA. Target mRNAs are repressed as efficiently by microRNA-binding sites in the 5′ UTR as in the 3′ UTR. Proc Natl Acad Sci U S A 2007; 104: 9667–72.
- 37Shin C, Nam JW, Farh KK, Chiang HR, Shkumatava A, Bartel DP. Expanding the microRNA targeting code: functional sites with centered pairing. Mol Cell 2010; 38: 789–802.
- 38Zardo G, Ciolfi A, Vian L et al. Polycombs and microRNA-223 regulate human granulopoiesis by transcriptional control of target gene expression. Blood 2012; 119: 4034–46.
- 39Kim DH, Saetrom P, Snove O Jr, Rossi JJ. MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci U S A 2008; 105: 16230–5.
- 40Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 2008; 9: 102–14.
- 41Doench JG, Petersen CP, Sharp PA. siRNAs can function as miRNAs. Genes Dev 2003; 17: 438–42.
- 42Ameres SL, Horwich MD, Hung JH et al. Target RNA-directed trimming and tailing of small silencing RNAs. Science 2010; 328: 1534–9.
- 43Cazalla D, Yario T, Steitz J. Down-regulation of a host microRNA by a Herpesvirus saimiri noncoding RNA. Science 2010; 328: 1563–6.
- 44Kuchen S, Resch W, Yamane A et al. Regulation of MicroRNA Expression and Abundance during Lymphopoiesis. Immunity 2010; 32: 828–39.
- 45Krutzfeldt J, Rajewsky N, Braich R et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 2005; 438: 685–9.
- 46Esau C, Davis S, Murray SF et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab 2006; 3: 87–98.
- 47Ebert PJ, Jiang S, Xie J, Li QJ, Davis MM. An endogenous positively selecting peptide enhances mature T cell responses and becomes an autoantigen in the absence of microRNA miR-181a. Nat Immunol 2009; 10: 1162–9.
- 48Li QJ, Chau J, Ebert PJ et al. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 2007; 129: 147–61.
- 49Ebert MS, Neilson JR, Sharp PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods 2007; 4: 721–6.
- 50Gentner B, Schira G, Giustacchini A et al. Stable knockdown of microRNA in vivo by lentiviral vectors. Nat Methods 2009; 6: 63–6.
- 51Lin SL, Chang SJ, Ying SY. Transgene-like animal models using intronic microRNAs. Methods Mol Biol 2006; 342: 321–34.
- 52Xiao C, Calado DP, Galler G et al. MiR-150 controls B cell differentiation by targeting the transcription factor c-Myb. Cell 2007; 131: 146–59.
- 53Johnnidis JB, Harris MH, Wheeler RT et al. Regulation of progenitor cell proliferation and granulocyte function by microRNA-223. Nature 2008; 451: 1125–9.
- 54Rodriguez A, Vigorito E, Clare S et al. Requirement of bic/microRNA-155 for normal immune function. Science 2007; 316: 608–11.
- 55Muljo SA, Ansel KM, Kanellopoulou C, Livingston DM, Rao A, Rajewsky K. Aberrant T cell differentiation in the absence of Dicer. J Exp Med 2005; 202: 261–9.
- 56Koralov SB, Muljo SA, Galler GR et al. Dicer ablation affects antibody diversity and cell survival in the B lymphocyte lineage. Cell 2008; 132: 860–74.
- 57Chong MM, Rasmussen JP, Rudensky AY, Littman DR. The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease. J Exp Med 2008; 205: 2005–17.
- 58O'Carroll D, Mecklenbrauker I, Das PP et al. A Slicer-independent role for Argonaute 2 in hematopoiesis and the microRNA pathway. Genes Dev 2007; 21: 1999–2004.
- 59Cobb BS, Nesterova TB, Thompson E et al. T cell lineage choice and differentiation in the absence of the RNase III enzyme Dicer. J Exp Med 2005; 201: 1367–73.
- 60Fazi F, Rosa A, Fatica A et al. A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis. Cell 2005; 123: 819–31.
- 61Monticelli S, Ansel KM, Xiao C et al. MicroRNA profiling of the murine hematopoietic system. Genome Biol 2005; 6: R71.
- 62Poltorak A, He X, Smirnova I et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 1998; 282: 2085–8.
- 63Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A 2006; 103: 12481–6.
- 64Eissmann P, Evans JH, Mehrabi M, Rose EL, Nedvetzki S, Davis DM. Multiple mechanisms downstream of TLR-4 stimulation allow expression of NKG2D ligands to facilitate macrophage/NK cell crosstalk. J Immunol 2010; 184: 6901–9.
- 65Bazzoni F, Rossato M, Fabbri M et al. Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals. Proc Natl Acad Sci U S A 2009; 106: 5282–7.
- 66Liu G, Friggeri A, Yang Y, Park YJ, Tsuruta Y, Abraham E.. miR-147, a microRNA that is induced upon Toll-like receptor stimulation, regulates murine macrophage inflammatory responses. Proc Natl Acad Sci U S A 2009; 106: 15819–24.
- 67Alsaleh G, Suffert G, Semaan N et al. Bruton's tyrosine kinase is involved in miR-346-related regulation of IL-18 release by lipopolysaccharide-activated rheumatoid fibroblast-like synoviocytes. J Immunol 2009; 182: 5088–97.
- 68Chen XM, Splinter PL, O'Hara SP, LaRusso NF. A cellular micro-RNA, let-7i, regulates Toll-like receptor 4 expression and contributes to cholangiocyte immune responses against Cryptosporidium parvum infection. J Biol Chem 2007; 282: 28929–38.
- 69Sheedy FJ, Palsson-McDermott E, Hennessy EJ et al. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat Immunol 2010; 11: 141–7.
- 70Tili E, Michaille JJ, Cimino A et al. Modulation of miR-155 and miR-125b levels following lipopolysaccharide/TNF-alpha stimulation and their possible roles in regulating the response to endotoxin shock. J Immunol 2007; 179: 5082–9.
- 71Androulidaki A, Iliopoulos D, Arranz A et al. The kinase Akt1 controls macrophage response to lipopolysaccharide by regulating microRNAs. Immunity 2009; 31: 220–31.
- 72Brown BD, Gentner B, Cantore A et al. Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nat Biotechnol 2007; 25: 1457–67.
- 73Nahid MA, Satoh M, Chan EK. Mechanistic Role of MicroRNA-146a in Endotoxin-Induced Differential Cross-Regulation of TLR Signaling. J Immunol 2011; 186: 1723–34.
- 74Jurkin J, Schichl YM, Koeffel R et al. miR-146a is differentially expressed by myeloid dendritic cell subsets and desensitizes cells to TLR2-dependent activation. J Immunol 2010; 184: 4955–65.
- 75Perry MM, Moschos SA, Williams AE, Shepherd NJ, Larner-Svensson HM, Lindsay MA. Rapid changes in microRNA-146a expression negatively regulate the IL-1beta-induced inflammatory response in human lung alveolar epithelial cells. J Immunol 2008; 180: 5689–98.
- 76O'Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci U S A 2007; 104: 1604–9.
- 77McCoy CE, Sheedy FJ, Qualls JE et al. IL-10 inhibits miR-155 induction by toll-like receptors. J Biol Chem 2010; 285: 20492–8.
- 78Kurowska-Stolarska M, Alivernini S, Ballantine LE et al. MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis. Proc Natl Acad Sci U S A 2011; 108: 11193–8.
- 79Zhou H, Huang X, Cui H et al. miR-155 and its star-form partner miR-155* cooperatively regulate type I interferon production by human plasmacytoid dendritic cells. Blood 2010; 116: 5885–94.
- 80Yin Q, McBride J, Fewell C et al. MicroRNA-155 is an Epstein-Barr virus-induced gene that modulates Epstein-Barr virus-regulated gene expression pathways. J Virol 2008; 82: 5295–306.
- 81Martinez-Nunez RT, Louafi F, Sanchez-Elsner T. The interleukin 13 (IL-13) pathway in human macrophages is modulated by microRNA-155 via direct targeting of interleukin 13 receptor alpha1 (IL13Ralpha1). J Biol Chem 2010; 286: 1786–94.
- 82Xue Q, Guo ZY, Li W et al. Human activated CD4(+) T lymphocytes increase IL-2 expression by downregulating microRNA-181c. Mol Immunol 2011; 48: 592–9.
- 83Stittrich AB, Haftmann C, Sgouroudis E et al. The microRNA miR-182 is induced by IL-2 and promotes clonal expansion of activated helper T lymphocytes. Nat Immunol 2010; 11: 1057–62.
- 84Nakayamada S, Takahashi H, Kanno Y, O'Shea JJ. Helper T cell diversity and plasticity. Curr Opin Immunol 2012; 24: 297–302.
- 85O'Shea JJ, Lahesmaa R, Vahedi G, Laurence A, Kanno Y. Genomic views of STAT function in CD4 + T helper cell differentiation. Nat Rev Immunol 2011; 11: 239–50.
- 86Steiner DF, Thomas MF, Hu JK et al. MicroRNA-29 Regulates T-Box Transcription Factors and Interferon-gamma Production in Helper T Cells. Immunity 2011; 35: 169–81.
- 87Ma F, Xu S, Liu X et al. The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-gamma. Nat Immunol 2011; 12: 861–9.
- 88Rossi RL, Rossetti G, Wenandy L et al. Distinct microRNA signatures in human lymphocyte subsets and enforcement of the naive state in CD4 + T cells by the microRNA miR-125b. Nat Immunol 2011; 12: 796–803.
- 89Almanza G, Fernandez A, Volinia S, Cortez-Gonzalez X, Croce CM, Zanetti M. Selected microRNAs define cell fate determination of murine central memory CD8 T cells. PLoS ONE 2010; 5: e11243.
- 90Lu LF, Thai TH, Calado DP et al. Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein. Immunity 2009; 30: 80–91.
- 91Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky AY. Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature 2007; 445: 936–40.
- 92Thai TH, Calado DP, Casola S et al. Regulation of the germinal center response by microRNA-155. Science 2007; 316: 604–8.
- 93Vigorito E, Perks KL, Abreu-Goodger C et al. microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells. Immunity 2007; 27: 847–59.
- 94Curtale G, Citarella F, Carissimi C et al. An emerging player in the adaptive immune response: microRNA-146a is a modulator of IL-2 expression and activation-induced cell death in T lymphocytes. Blood 2010; 115: 265–73.
- 95Lu LF, Boldin MP, Chaudhry A et al. Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses. Cell 2010; 142: 914–29.
- 96Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J, Benz CC. Expression of microRNA-146 suppresses NF-kappaB activity with reduction of metastatic potential in breast cancer cells. Oncogene 2008; 27: 5643–7.
- 97Kumar MS, Erkeland SJ, Pester RE et al. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc Natl Acad Sci U S A 2008; 105: 3903–8.
- 98Tam W, Dahlberg JE. miR-155/BIC as an oncogenic microRNA. Genes Chromosom Cancer 2006; 45: 211–2.
- 99Eis PS, Tam W, Sun L et al. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc Natl Acad Sci U S A 2005; 102: 3627–32.
- 100Gregersen LH, Jacobsen AB, Frankel LB, Wen J, Krogh A, Lund AH. MicroRNA-145 targets YES and STAT1 in colon cancer cells. PLoS ONE 2010; 5: e8836.
- 101Asangani IA, Rasheed SA, Nikolova DA et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008; 27: 2128–36.
- 102Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A, Lund AH. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem 2008; 283: 1026–33.
- 103Garofalo M, Di Leva G, Romano G et al. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell 2009; 16: 498–509.
- 104Nakasa T, Miyaki S, Okubo A et al. Expression of microRNA-146 in rheumatoid arthritis synovial tissue. Arthritis Rheum 2008; 58: 1284–92.
- 105Stanczyk J, Pedrioli DM, Brentano F et al. Altered expression of MicroRNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis. Arthritis Rheum 2008; 58: 1001–9.
- 106Pauley KM, Satoh M, Chan AL, Bubb MR, Reeves WH, Chan EK. Upregulated miR-146a expression in peripheral blood mononuclear cells from rheumatoid arthritis patients. Arthritis Res Ther 2008; 10: R101.
- 107Nakamachi Y, Kawano S, Takenokuchi M et al. MicroRNA-124a is a key regulator of proliferation and monocyte chemoattractant protein 1 secretion in fibroblast-like synoviocytes from patients with rheumatoid arthritis. Arthritis Rheum 2009; 60: 1294–304.
- 108Fulci V, Scappucci G, Sebastiani GD et al. miR-223 is overexpressed in T-lymphocytes of patients affected by rheumatoid arthritis. Hum Immunol 2010; 71: 206–11.
- 109Shibuya H, Nakasa T, Adachi N et al. Overexpression of microRNA-223 in rheumatoid arthritis synovium controls osteoclast differentiation. Mod Rheumatol 2012; doi: 10.1007/s10165-012-0710-1. [Epub ahead of print].
10.1007/s10165‐012‐0710‐1 Google Scholar
- 110Li YT, Chen SY, Wang CR et al. Brief report: amelioration of collagen-induced arthritis in mice by lentivirus-mediated silencing of microRNA-223. Arthritis Rheum 2012; 64: 3240–5.
- 111Niimoto T, Nakasa T, Ishikawa M et al. MicroRNA-146a expresses in interleukin-17 producing T cells in rheumatoid arthritis patients. BMC Musculoskelet Disord 2010; 11: 209.
- 112Nakasa T, Shibuya H, Nagata Y, Niimoto T, Ochi M. The inhibitory effect of microRNA-146a expression on bone destruction in collagen-induced arthritis. Arthritis Rheum 2011; 63: 1582–90.
- 113Semaan N, Frenzel L, Alsaleh G et al. miR-346 controls release of TNF-alpha protein and stability of its mRNA in rheumatoid arthritis via tristetraprolin stabilization. PLoS ONE 2011; 6: e19827.
- 114Nagata Y, Nakasa T, Mochizuki Y et al. Induction of apoptosis in the synovium of mice with autoantibody-mediated arthritis by the intraarticular injection of double-stranded MicroRNA-15a. Arthritis Rheum 2009; 60: 2677–83.
- 115Zhu S, Pan W, Song X et al. The microRNA miR-23b suppresses IL-17-associated autoimmune inflammation by targeting TAB 2, TAB 3 and IKK-alpha. Nat Med 2012; 18: 1077–86.
- 116Philippe L, Alsaleh G, Suffert G et al. TLR2 expression is regulated by microRNA miR-19 in rheumatoid fibroblast-like synoviocytes. J Immunol 2012; 188: 454–61.
- 117Trenkmann M, Brock M, Gay RE, Michel BA, Gay S, Huber LC. The TNFalpha-induced miR-18a activates rheumatoid arthritis synovial fibroblasts through a feedback loop in NF-kappaB signaling. Arthritis Rheum 2013; 65: 916–27.
- 118Keller A, Leidinger P, Lange J et al. Multiple sclerosis: microRNA expression profiles accurately differentiate patients with relapsing-remitting disease from healthy controls. PLoS ONE 2009; 4: e7440.
- 119Cox MB, Cairns MJ, Gandhi KS et al. MicroRNAs miR-17 and miR-20a inhibit T cell activation genes and are under-expressed in MS whole blood. PLoS ONE 2010; 5: e12132.
- 120De Santis G, Ferracin M, Biondani A et al. Altered miRNA expression in T regulatory cells in course of multiple sclerosis. J Neuroimmunol 2010; 226: 165–71.
- 121Lindberg RL, Hoffmann F, Mehling M, Kuhle J, Kappos L. Altered expression of miR-17-5p in CD4 + lymphocytes of relapsing-remitting multiple sclerosis patients. Eur J Immunol 2010; 40: 888–98.
- 122Du C, Liu C, Kang J et al. MicroRNA miR-326 regulates TH-17 differentiation and is associated with the pathogenesis of multiple sclerosis. Nat Immunol 2009; 10: 1252–9.
- 123Otaegui D, Baranzini SE, Armananzas R et al. Differential micro RNA expression in PBMC from multiple sclerosis patients. PLoS ONE 2009; 4: e6309.
- 124Junker A, Krumbholz M, Eisele S et al. MicroRNA profiling of multiple sclerosis lesions identifies modulators of the regulatory protein CD47. Brain 2009; 132: 3342–52.
- 125Siegel SR, Mackenzie J, Chaplin G, Jablonski NG, Griffiths L. Circulating microRNAs involved in multiple sclerosis. Mol Biol Rep 2012; 39: 6219–25.
- 126Sievers C, Meira M, Hoffmann F, Fontoura P, Kappos L, Lindberg RL. Altered microRNA expression in B lymphocytes in multiple sclerosis: towards a better understanding of treatment effects. Clin Immunol 2012; 144: 70–9.
- 127Haghikia A, Haghikia A, Hellwig K et al. Regulated microRNAs in the CSF of patients with multiple sclerosis: a case-control study. Neurology 2012; 79: 2166–70.
- 128Ponomarev ED, Veremeyko T, Barteneva N, Krichevsky AM, Weiner HL. MicroRNA-124 promotes microglia quiescence and suppresses EAE by deactivating macrophages via the C/EBP-alpha-PU.1 pathway. Nat Med 2010; 17: 64–70.
- 129Lorenzi JC, Brum DG, Zanette DL et al. miR-15a and 16-1 are downregulated in CD4 + T cells of multiple sclerosis relapsing patients. Int J Neurosci 2012; 122: 466–71.
- 130Smith KM, Guerau-de-Arellano M, Costinean S et al. miR-29ab1 deficiency identifies a negative feedback loop controlling Th1 bias that is dysregulated in multiple sclerosis. J Immunol 2012; 189: 1567–76.
- 131Murugaiyan G, Beynon V, Mittal A, Joller N, Weiner HL. Silencing microRNA-155 ameliorates experimental autoimmune encephalomyelitis. J Immunol 2011; 187: 2213–21.
- 132Guan H, Fan D, Mrelashvili D et al. MicroRNA let-7e is associated with the pathogenesis of experimental autoimmune encephalomyelitis. Eur J Immunol 2012; 43: 104–14.
- 133Lescher J, Paap F, Schultz V et al. MicroRNA regulation in experimental autoimmune encephalomyelitis in mice and marmosets resembles regulation in human multiple sclerosis lesions. J Neuroimmunol 2012; 246: 27–33.
- 134McKenna LB, Schug J, Vourekas A et al. MicroRNAs control intestinal epithelial differentiation, architecture, and barrier function. Gastroenterology 2010; 139: 1654–64.
- 135Wu F, Zikusoka M, Trindade A et al. MicroRNAs are differentially expressed in ulcerative colitis and alter expression of macrophage inflammatory peptide-2 alpha. Gastroenterology 2008; 135: 1624–35.
- 136Wu F, Zhang S, Dassopoulos T et al. Identification of microRNAs associated with ileal and colonic Crohn's disease. Inflamm Bowel Dis 2010; 16: 1729–38.
- 137Wu F, Guo NJ, Tian H et al. Peripheral blood microRNAs distinguish active ulcerative colitis and Crohn's disease. Inflamm Bowel Dis 2011; 17: 241–50.
- 138Zahm AM, Thayu M, Hand NJ, Horner A, Leonard MB, Friedman JR. Circulating microRNA is a biomarker of pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2011; 53: 26–33.
- 139Paraskevi A, Theodoropoulos G, Papaconstantinou I, Mantzaris G, Nikiteas N, Gazouli M. Circulating MicroRNA in inflammatory bowel disease. J Crohns Colitis 2012; 6: 900–4.
- 140Duttagupta R, DiRienzo S, Jiang R et al. Genome-wide maps of circulating miRNA biomarkers for ulcerative colitis. PLoS ONE 2012; 7: e31241.
- 141Prescott NJ, Dominy KM, Kubo M et al. Independent and population-specific association of risk variants at the IRGM locus with Crohn's disease. Hum Mol Genet 2010; 19: 1828–39.
- 142Brest P, Lapaquette P, Souidi M et al. A synonymous variant in IRGM alters a binding site for miR-196 and causes deregulation of IRGM-dependent xenophagy in Crohn's disease. Nat Genet 2011; 43: 242–5.
- 143Zwiers A, Kraal L, van de Pouw Kraan TC, Wurdinger T, Bouma G, Kraal G. Cutting edge: a variant of the IL-23R gene associated with inflammatory bowel disease induces loss of microRNA regulation and enhanced protein production. J Immunol 2011; 188: 1573–7.
- 144Oertli M, Engler DB, Kohler E, Koch M, Meyer TF, Muller A. MicroRNA-155 is essential for the T cell-mediated control of Helicobacter pylori infection and for the induction of chronic Gastritis and Colitis. J Immunol 2011; 187: 3578–86.
- 145Sonkoly E, Wei T, Janson PC et al. MicroRNAs: novel regulators involved in the pathogenesis of psoriasis? PLoS ONE 2007; 2: e610.
- 146Joyce CE, Zhou X, Xia J et al. Deep sequencing of small RNAs from human skin reveals major alterations in the psoriasis miRNAome. Hum Mol Genet 2011; 20: 4025–40.
- 147Ichihara A, Jinnin M, Yamane K et al. microRNA-mediated keratinocyte hyperproliferation in psoriasis vulgaris. Br J Dermatol 2011; 165: 1003–10.
- 148Lerman G, Avivi C, Mardoukh C et al. MiRNA expression in psoriatic skin: reciprocal regulation of hsa-miR-99a and IGF-1R. PLoS ONE 2011; 6: e20916.
- 149Zibert JR, Lovendorf MB, Litman T, Olsen J, Kaczkowski B, Skov L. MicroRNAs and potential target interactions in psoriasis. J Dermatol Sci 2010; 58: 177–85.
- 150Xu N, Meisgen F, Butler LM et al. MicroRNA-31 Is Overexpressed in Psoriasis and Modulates Inflammatory Cytokine and Chemokine Production in Keratinocytes via Targeting Serine/Threonine Kinase 40. J Immunol 2012; 190: 678–88.
- 151Peng H, Kaplan N, Hamanaka RB et al. microRNA-31/factor-inhibiting hypoxia-inducible factor 1 nexus regulates keratinocyte differentiation. Proc Natl Acad Sci U S A 2012; 109: 14030–4.
- 152Xu N, Brodin P, Wei T et al. MiR-125b, a microRNA downregulated in psoriasis, modulates keratinocyte proliferation by targeting FGFR2. J Invest Dermatol 2011; 131: 1521–9.
- 153Meisgen F, Xu N, Wei T et al. MiR-21 is up-regulated in psoriasis and suppresses T cell apoptosis. Exp Dermatol 2012; 21: 312–4.
- 154Sano S, Chan KS, Carbajal S et al. Stat3 links activated keratinocytes and immunocytes required for development of psoriasis in a novel transgenic mouse model. Nat Med 2005; 11: 43–9.
- 155Lena AM, Shalom-Feuerstein R, Rivetti di Val Cervo P et al. miR-203 represses ‘stemness’ by repressing DeltaNp63. Cell Death Differ 2008; 15: 1187–95.
- 156Yi R, Poy MN, Stoffel M, Fuchs E. A skin microRNA promotes differentiation by repressing ‘stemness’. Nature 2008; 452: 225–9.
- 157Sonkoly E, Janson P, Majuri ML et al. MiR-155 is overexpressed in patients with atopic dermatitis and modulates T-cell proliferative responses by targeting cytotoxic T lymphocyte-associated antigen 4. J Allergy Clin Immunol 2010; 126: 581–9. e1-20.
- 158Vennegaard MT, Bonefeld CM, Hagedorn PH et al. Allergic contact dermatitis induces upregulation of identical microRNAs in humans and mice. Contact Dermatitis 2012; 67: 298–305.
- 159Shaoqing Y, Ruxin Z, Guojun L et al. Microarray analysis of differentially expressed microRNAs in allergic rhinitis. Am J Rhinol Allergy 2012; 25: e242–6.
- 160Williams AE, Larner-Svensson H, Perry MM et al. MicroRNA expression profiling in mild asthmatic human airways and effect of corticosteroid therapy. PLoS ONE 2009; 4: e5889.
- 161Solberg OD, Ostrin EJ, Love MI et al. Airway epithelial miRNA expression is altered in asthma. Am J Respir Crit Care Med 2012; 186: 965–74.
- 162Jardim MJ, Dailey L, Silbajoris R, Diaz-Sanchez D. Distinct microRNA expression in human airway cells of asthmatic donors identifies a novel asthma-associated gene. Am J Respir Cell Mol Biol 2012; 47: 536–42.
- 163Yamamoto M, Singh A, Ruan J et al. Decreased miR-192 expression in peripheral blood of asthmatic individuals undergoing an allergen inhalation challenge. BMC Genomics 2012; 13: 655.
- 164Moschos SA, Williams AE, Perry MM, Birrell MA, Belvisi MG, Lindsay MA. Expression profiling in vivo demonstrates rapid changes in lung microRNA levels following lipopolysaccharide-induced inflammation but not in the anti-inflammatory action of glucocorticoids. BMC Genomics 2007; 8: 240.
- 165Mattes J, Collison A, Plank M, Phipps S, Foster PS. Antagonism of microRNA-126 suppresses the effector function of TH2 cells and the development of allergic airways disease. Proc Natl Acad Sci U S A 2009; 106: 18704–9.
- 166Brunner C, Sindrilaru A, Girkontaite I, Fischer KD, Sunderkotter C, Wirth T. BOB.1/OBF.1 controls the balance of TH1 and TH2 immune responses. EMBO J 2007; 26: 3191–202.
- 167Chang HC, Zhang S, Thieu VT et al. PU.1 expression delineates heterogeneity in primary Th2 cells. Immunity 2005; 22: 693–703.
- 168Collison A, Herbert C, Siegle JS, Mattes J, Foster PS, Kumar RK. Altered expression of microRNA in the airway wall in chronic asthma: miR-126 as a potential therapeutic target. BMC Pulm Med 2011; 11: 29.
- 169Collison A, Mattes J, Plank M, Foster PS. Inhibition of house dust mite-induced allergic airways disease by antagonism of microRNA-145 is comparable to glucocorticoid treatment. J Allergy Clin Immunol 2011; 128: 160–7.
- 170Lu TX, Munitz A, Rothenberg ME. MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. J Immunol 2009; 182: 4994–5002.
- 171Lu TX, Hartner J, Lim EJ et al. MicroRNA-21 limits in vivo immune response-mediated activation of the IL-12/IFN-gamma pathway, Th1 polarization, and the severity of delayed-type hypersensitivity. J Immunol 2011; 187: 3362–73.
- 172Feng MJ, Shi F, Qiu C, Peng WK. MicroRNA-181a, -146a and -146b in spleen CD4 + T lymphocytes play proinflammatory roles in a murine model of asthma. Int Immunopharmacol 2012; 13: 347–53.
- 173Sharma A, Kumar M, Aich J et al. Posttranscriptional regulation of interleukin-10 expression by hsa-miR-106a. Proc Natl Acad Sci U S A 2009; 106: 5761–6.
- 174Sharma A, Kumar M, Ahmad T et al. Antagonism of mmu-mir-106a attenuates asthma features in allergic murine model. J Appl Physiol 2012; 113: 459–64.
- 175Liu F, Qin HB, Xu B, Zhou H, Zhao DY. Profiling of miRNAs in pediatric asthma: upregulation of miRNA-221 and miRNA-485-3p. Mol Med Report 2012; 6: 1178–82.
- 176Qin HB, Xu B, Mei JJ et al. Inhibition of miRNA-221 suppresses the airway inflammation in asthma. Inflammation 2012; 35: 1595–9.
- 177Garbacki N, Di Valentin E, Huynh-Thu VA et al. MicroRNAs profiling in murine models of acute and chronic asthma: a relationship with mRNAs targets. PLoS ONE 2011; 6: e16509.
- 178Polikepahad S, Knight JM, Naghavi AO et al. Proinflammatory role for let-7 microRNAS in experimental asthma. J Biol Chem 2010; 285: 30139–49.
- 179Kumar M, Ahmad T, Sharma A et al. Let-7 microRNA-mediated regulation of IL-13 and allergic airway inflammation. J Allergy Clin Immunol 2011; 128: 1077–85.
- 180Oglesby IK, Bray IM, Chotirmall SH et al. miR-126 is downregulated in cystic fibrosis airway epithelial cells and regulates TOM1 expression. J Immunol 2010; 184: 1702–9.
- 181Xie T, Liang J, Liu N et al. MicroRNA-127 Inhibits Lung Inflammation by Targeting IgG Fcgamma Receptor I. J Immunol 2012; 188: 2437–44.
- 182Sato T, Liu X, Nelson A et al. Reduced MiR-146a Increases Prostaglandin E2 in Chronic Obstructive Pulmonary Disease Fibroblasts. Am J Respir Crit Care Med 2010; 182: 1020–9.
- 183Larner-Svensson HM, Williams AE, Tsitsiou E et al. Pharmacological studies of the mechanism and function of interleukin-1beta-induced miRNA-146a expression in primary human airway smooth muscle. Respir Res 2010; 11: 68.
- 184Graff JW, Powers LS, Dickson AM et al. Cigarette smoking decreases global microRNA expression in human alveolar macrophages. PLoS ONE 2012; 7: e44066.
- 185Ezzie ME, Crawford M, Cho JH et al. Gene expression networks in COPD: microRNA and mRNA regulation. Thorax 2011; 67: 122–31.
- 186Mizuno S, Bogaard HJ, Gomez-Arroyo J et al. MicroRNA-199a-5p is associated with hypoxia-inducible factor-1alpha expression in lungs from patients with COPD. Chest 2012; 142: 663–72.
- 187Liu G, Friggeri A, Yang Y et al. miR-21 mediates fibrogenic activation of pulmonary fibroblasts and lung fibrosis. J Exp Med 2010; 142: 663–72.
- 188Kuhn AR, Schlauch K, Lao R, Halayko AJ, Gerthoffer WT, Singer CA. MicroRNA expression in human airway smooth muscle cells: role of miR-25 in regulation of airway smooth muscle phenotype. Am J Respir Cell Mol Biol 2009; 42: 506–13.
- 189Mohamed JS, Lopez MA, Boriek AM. Mechanical stretch up-regulates microRNA-26a and induces human airway smooth muscle hypertrophy by suppressing glycogen synthase kinase-3beta. J Biol Chem 2010; 285: 29336–47.
- 190Chiba Y, Tanabe M, Goto K, Sakai H, Misawa M. Down-regulation of miR-133a contributes to up-regulation of Rhoa in bronchial smooth muscle cells. Am J Respir Crit Care Med 2009; 180: 713–9.
- 191Iliopoulos D, Hirsch HA, Struhl K. An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell 2009; 139: 693–706.
- 192Witwer KW, Sisk JM, Gama L, Clements JE. MicroRNA regulation of IFN-beta protein expression: rapid and sensitive modulation of the innate immune response. J Immunol 2010; 184: 2369–76.
- 193Zhao H, Kalota A, Jin S, Gewirtz AM. The c-myb proto-oncogene and microRNA-15a comprise an active autoregulatory feedback loop in human hematopoietic cells. Blood 2009; 113: 505–16.
- 194Gantier MP, Stunden HJ, McCoy CE et al. A miR-19 regulon that controls NF-kappaB signaling. Nucleic Acids Res 2012; 40: 8048–58.
- 195Lu Z, Liu M, Stribinskis V et al. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene 2008; 27: 4373–9.
- 196Kim YJ, Hwang SJ, Bae YC, Jung JS. MiR-21 regulates adipogenic differentiation through the modulation of TGF-beta signaling in mesenchymal stem cells derived from human adipose tissue. Stem Cells 2009; 27: 3093–102.
- 197Rogler CE, Levoci L, Ader T et al. MicroRNA-23b cluster microRNAs regulate transforming growth factor-beta/bone morphogenetic protein signaling and liver stem cell differentiation by targeting Smads. Hepatology 2009; 50: 575–84.
- 198Petrocca F, Vecchione A, Croce CM. Emerging role of miR-106b-25/miR-17-92 clusters in the control of transforming growth factor beta signaling. Cancer Res 2008; 68: 8191–4.
- 199Leeper NJ, Raiesdana A, Kojima Y et al. MicroRNA-26a is a novel regulator of vascular smooth muscle cell function. J Cell Physiol 2011; 226: 1035–43.
- 200Li Z, Hassan MQ, Jafferji M et al. Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J Biol Chem 2009; 284: 15676–84.
- 201Rouas R, Fayyad-Kazan H, El Zein N et al. Human natural Treg microRNA signature: role of microRNA-31 and microRNA-21 in FOXP3 expression. Eur J Immunol 2009; 39: 1608–18.
- 202Suarez Y, Wang C, Manes TD, Pober JS. Cutting edge: TNF-induced microRNAs regulate TNF-induced expression of E-selectin and intercellular adhesion molecule-1 on human endothelial cells: feedback control of inflammation. J Immunol 2010; 184: 21–5.
- 203Hatziapostolou M, Polytarchou C, Aggelidou E et al. An HNF4alpha-miRNA inflammatory feedback circuit regulates hepatocellular oncogenesis. Cell 2011; 147: 1233–47.
- 204Kim SW, Ramasamy K, Bouamar H, Lin AP, Jiang D, Aguiar RC. MicroRNAs miR-125a and miR-125b constitutively activate the NF-kappaB pathway by targeting the tumor necrosis factor alpha-induced protein 3 (TNFAIP3, A20). Proc Natl Acad Sci U S A 2012; 109: 7865–70.
- 205Qin A, Wen Z, Zhou Y et al. MicroRNA-126 regulates the induction and function of CD4(+) Foxp3(+) regulatory T cells through PI3K/AKT pathway. J Cell Mol Med 2013; 17: 252–64.
- 206Yang K, He YS, Wang XQ et al. MiR-146a inhibits oxidized low-density lipoprotein-induced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett 2011; 585: 854–60.
- 207Banerjee A, Schambach F, DeJong CS, Hammond SM, Reiner SL. Micro-RNA-155 inhibits IFN-gamma signaling in CD4 + T cells. Eur J Immunol 2010; 40: 225–31.
- 208Rai D, Kim SW, McKeller MR, Dahia PL, Aguiar RC. Targeting of SMAD5 links microRNA-155 to the TGF-beta pathway and lymphomagenesis. Proc Natl Acad Sci U S A 2010; 107: 3111–6.
- 209He M, Xu Z, Ding T, Kuang DM, Zheng L. MicroRNA-155 regulates inflammatory cytokine production in tumor-associated macrophages via targeting C/EBPbeta. Cell Mol Immunol 2009; 6: 343–52.
- 210Ceppi M, Pereira PM, Dunand-Sauthier I et al. MicroRNA-155 modulates the interleukin-1 signaling pathway in activated human monocyte-derived dendritic cells. Proc Natl Acad Sci U S A 2009; 106: 2735–40.
- 211Di Leva G, Gasparini P, Piovan C et al. MicroRNA cluster 221-222 and estrogen receptor alpha interactions in breast cancer. J Natl Cancer Inst 2010; 102: 706–21.
- 212He H, Jazdzewski K, Li W et al. The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci U S A 2005; 102: 19075–80.