Immune Netw.  2014 Jun;14(3):138-148. 10.4110/in.2014.14.3.138.

MicroRNA Regulation in Systemic Lupus Erythematosus Pathogenesis

Affiliations
  • 1Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong. sfvchan@hku.hk
  • 2Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.

Abstract

MicroRNAs (miRNAs) are endogenous small RNA molecules best known for their function in post-transcriptional gene regulation. Immunologically, miRNA regulates the differentiation and function of immune cells and its malfunction contributes to the development of various autoimmune diseases including systemic lupus erythematosus (SLE). Over the last decade, accumulating researches provide evidence for the connection between dysregulated miRNA network and autoimmunity. Interruption of miRNA biogenesis machinery contributes to the abnormal T and B cell development and particularly a reduced suppressive function of regulatory T cells, leading to systemic autoimmune diseases. Additionally, multiple factors under autoimmune conditions interfere with miRNA generation via key miRNA processing enzymes, thus further skewing the miRNA expression profile. Indeed, several independent miRNA profiling studies reported significant differences between SLE patients and healthy controls. Despite the lack of a consistent expression pattern on individual dysregulated miRNAs in SLE among these studies, the aberrant expression of distinct groups of miRNAs causes overlapping functional outcomes including perturbed type I interferon signalling cascade, DNA hypomethylation and hyperactivation of T and B cells. The impact of specific miRNA-mediated regulation on function of major immune cells in lupus is also discussed. Although research on the clinical application of miRNAs is still immature, through an integrated approach with advances in next generation sequencing, novel tools in bioinformatics database analysis and new in vitro and in vivo models for functional evaluation, the diagnostic and therapeutic potentials of miRNAs may bring to fruition in the future.

Keyword

MicroRNAs; Systemic Lupus Erythematosus; Autoimmunity

MeSH Terms

Autoimmune Diseases
Autoimmunity
B-Lymphocytes
Computational Biology
DNA
Humans
Interferon Type I
Lupus Erythematosus, Systemic*
MicroRNAs*
Organelle Biogenesis
RNA
T-Lymphocytes, Regulatory
DNA
Interferon Type I
MicroRNAs
RNA

Figure

  • Figure 1 Biogenesis of miRNA. MiRNA coding genes or the regions within introns of protein-coding genes are transcribed in the nucleus by RNA polymerase II. The transcription products are the hairpin-structured primary miRNAs (pri-miRNA) which will be further processed into precursor miRNA (pre-miRNA) by the microprocessor complex containing the Dorsha and DiGeorge Syndrome critical region 8 (DGCR8). These pre-miRNA will be transported into the cytoplasm by exportin 5 (XPO5), which the terminal loop will be excised and processed into a 18- to 24-nucleotide long duplex by Dicer. The RNA interfering silencing complex (RISC) will then unwind the miRNA duplex into single-stranded miRNA. While the single-stranded star form miRNA will be degraded rapidly, the other mature single-stranded miRNA will be directed to its complementary target mRNA. The target mRNA will be degraded if the complementarity is sufficient, while mRNA translation will be inhibited if complementarity is insufficient.


Cited by  2 articles

Wolves Trapped in the NETs–The Pathogenesis of Lupus Nephritis
Young Kim, Seung Cheol Shim
J Rheum Dis. 2018;25(2):81-99.    doi: 10.4078/jrd.2018.25.2.81.

Folate Receptor-Targeted Diagnostics and Therapeutics for Inflammatory Diseases
Young-Su Yi
Immune Netw. 2016;16(6):337-343.    doi: 10.4110/in.2016.16.6.337.


Reference

1. Carthew RW, Sontheimer EJ. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009; 136:642–655.
Article
2. Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009; 10:126–139.
Article
3. Inui M, Martello G, Piccolo S. MicroRNA control of signal transduction. Nat Rev Mol Cell Biol. 2010; 11:252–263.
Article
4. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009; 136:215–233.
Article
5. Chen CZ, Li I, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004; 303:83–86.
Article
6. Carrington JC, Ambros V. Role of microRNAs in plant and animal development. Science. 2003; 301:336–338.
Article
7. Baltimore D, Boldin MP, O'Connell RM, Rao DS, Taganov KD. MicroRNAs: new regulators of immune cell development and function. Nat Immunol. 2008; 9:839–845.
Article
8. O'Connell RM, Rao DS, Chaudhuri AA, Baltimore D. Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol. 2010; 10:111–122.
9. Asirvatham AJ, Gregorie CJ, Hu Z, Magner WJ, Tomasi TB. MicroRNA targets in immune genes and the Dicer/Argonaute and ARE machinery components. Mol Immunol. 2008; 45:1995–2006.
Article
10. Mak A, Isenberg DA, Lau CS. Global trends, potential mechanisms and early detection of organ damage in SLE. Nat Rev Rheumatol. 2013; 9:301–310.
Article
11. Rahman A, Isenberg DA. Systemic lupus erythematosus. N Engl J Med. 2008; 358:929–939.
Article
12. Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011; 365:2110–2121.
Article
13. Luo X, Tsai LM, Shen N, Yu D. Evidence for microRNA-mediated regulation in rheumatic diseases. Ann Rheum Dis. 2010; 69:Suppl 1. i30–i36.
Article
14. Zhu S, Pan W, Qian Y. MicroRNA in immunity and autoimmunity. J Mol Med (Berl). 2013; 91:1039–1050.
Article
15. Miao CG, Yang YY, He X, Huang C, Huang Y, Zhang L, Lv XW, Jin Y, Li J. The emerging role of microRNAs in the pathogenesis of systemic lupus erythematosus. Cell Signal. 2013; 25:1828–1836.
Article
16. Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar R. The Microprocessor complex mediates the genesis of microRNAs. Nature. 2004; 432:235–240.
Article
17. Lund E, Güttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science. 2004; 303:95–98.
Article
18. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004; 116:281–297.
19. Muljo SA, Ansel KM, Kanellopoulou C, Livingston DM, Rao A, Rajewsky K. Aberrant T cell differentiation in the absence of Dicer. J Exp Med. 2005; 202:261–269.
Article
20. Chong MM, Rasmussen JP, Rundensky AY, Littman DR. The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease. J Exp Med. 2008; 205:2005–2017.
Article
21. Cobb BS, Nesterova TB, Thompson E, Hertweck A, O'Connor E, Godwin J, Wilson CB, Brockdorff N, Fisher AG, Smale ST, Merkenschlager M. T cell lineage choice and differentiation in the absence of the RNase III enzyme Dicer. J Exp Med. 2005; 201:1367–1373.
Article
22. Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, McManus MT, Bluestone JA. Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J Exp Med. 2008; 205:1983–1991.
Article
23. Liston A, Lu LF, O'Carroll D, Tarakhovsky A, Rudensky AY. Dicer-dependent microRNA pathway safeguards regulatory T cell function. J Exp Med. 2008; 205:1993–2004.
Article
24. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+ CD25+ regulatory T cells. Nat Immunol. 2003; 4:330–336.
Article
25. Divekar AA, Dubey S, Gangalum PR, Singh RR. Dicer insufficiency and microRNA-155 overexpression in lupus regulatory T cells: an apparent paradox in the setting of an inflammatory milieu. J Immunol. 2011; 186:924–930.
Article
26. Belver L, de Yébenes VG, Ramiro AR. MicroRNAs prevent the generation of autoreactive antibodies. Immunity. 2010; 33:713–722.
Article
27. Xu S, Guo K, Zeng Q, Huo J, Lam KP. The RNase III enzyme Dicer is essential for germinal center B-cell formation. Blood. 2012; 119:767–776.
Article
28. Ademokun A, Turner M. Regulation of B-cell differentiation by microRNAs and RNA-binding proteins. Biochem Soc Trans. 2008; 36:1191–1193.
Article
29. Koralov SB, Muljo SA, Galler GR, Krek A, Chakraborty T, Kanellopoulou C, Jensen K, Cobb BS, Merkenschlager M, Rajewsky N, Rajewsky K. Dicer ablation affects antibody diversity and cell survival in the B lymphocyte lineage. Cell. 2008; 132:860–874.
Article
30. Wiesen JL, Tomasi TB. Dicer is regulated by cellular stresses and interferons. Mol Immunol. 2009; 46:1222–1228.
Article
31. Satoh M, Chan JYF, Ceribelli A, Vazquez del-Mercado M, Chan EK. Autoantibodies to Argonaute 2 (Su antigen). Adv Exp Med Biol. 2013; 768:45–59.
Article
32. Jakymiw A, Ikeda K, Fritzler MJ, Reeves WH, Satoh M, Chan EK. Autoimmune targeting of key components of RNA interference. Arthritis Res Ther. 2006; 8:R87.
33. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR. MicroRNA expression profiles classify human cancers. Nature. 2005; 435:834–838.
Article
34. Wang H, Peng W, Ouyang X, Li W, Dai Y. Circulating microRNAs as candidate biomarkers in patients with systemic lupus erythematosus. Transl Res. 2012; 160:198–206.
Article
35. Carlsen AL, Schetter AJ, Nielsen CT, Lood C, Knudsen S, Voss A, Harris CC, Hellmark T, Segelmark M, Jacobsen S, Bengtsson AA, Heegaard NH. Circulating microRNA expression profiles associated with systemic lupus erythematosus. Arthritis Rheum. 2013; 65:1324–1334.
Article
36. Tang Y, Luo X, Cui H, Ni X, Yuan M, Guo Y, Huang X, Zhou H, de Vries N, Tak PP, Chen S, Shen N. MicroRNA-146A contributes to abnormal activation of the type I interferon pathway in human lupus by targeting the key signaling proteins. Arthritis Rheum. 2009; 60:1065–1075.
Article
37. Te JL, Dozmorov IM, Guthridge JM, Nguyen KL, Cavett JW, Kelly JA, Bruner GR, Harley JB, Ojwang JO. Identification of unique microRNA signature associated with lupus nephritis. PLoS One. 2010; 5:e10344.
Article
38. Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A. 2003; 100:2610–2615.
Article
39. Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003; 197:711–723.
Article
40. Obermoser G, Pascual V. The interferon-alpha signature of systemic lupus erythematosus. Lupus. 2010; 19:1012–1019.
Article
41. Luo X, Zhang L, Li M, Zhang W, Leng X, Zhang F, Zhao Y, Zeng X. The role of miR-125b in T lymphocytes in the pathogenesis of systemic lupus erythematosus. Clin Exp Rheumatol. 2013; 31:263–271.
42. Lu MC, Lai NS, Chen HC, Yu HC, Huang KY, Tung CH, Huang HB, Yu CL. Decreased microRNA(miR)-145 and increased miR-224 expression in T cells from patients with systemic lupus erythematosus involved in lupus immunopathogenesis. Clin Exp Immunol. 2013; 171:91–99.
Article
43. Zhao S, Wang Y, Liang Y, Zhao M, Long H, Ding S, Yin H, Lu Q. MicroRNA-126 regulates DNA methylation in CD4+ T cells and contributes to systemic lupus erythematosus by targeting DNA methyltransferase 1. Arthritis Rheum. 2011; 63:1376–1386.
Article
44. Stagakis E, Bertsias G, Verginis P, Nakou M, Hatziapostolou M, Kritikos H, Iliopoulos D, Boumpas DT. Identification of novel microRNA signatures linked to human lupus disease activity and pathogenesis: miR-21 regulates aberrant T cell responses through regulation of PDCD4 expression. Ann Rheum Dis. 2011; 70:1496–1506.
Article
45. Pan W, Zhu S, Yuan M, Cui H, Wang L, Luo X, Li J, Zhou H, Tang Y, Shen N. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J Immunol. 2010; 184:6773–6781.
Article
46. Zhu X, Liang J, Li F, Yang Y, Xiang L, Xu J. Analysis of associations between the patterns of global DNA hypomethylation and expression of DNA methyltransferase in patients with systemic lupus erythematosus. Int J Dermatol. 2011; 50:697–704.
Article
47. Richardson B, Scheinbart L, Strahler J, Gross L, Hanash S, Johnson M. Evidence for impaired T cell DNA methylation in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum. 1990; 33:1665–1673.
Article
48. Zhang Y, Zhao M, Sawalha AH, Richardson B, Lu Q. Impaired DNA methylation and its mechanisms in CD4+ T cells of systemic lupus erythematosus. J Autoimmun. 2013; 41:92–99.
Article
49. Qin H, Zhu X, Liang J, Wu J, Yang Y, Wang S, Shi W, Xu J. MicroRNA-29b contributes to DNA hypomethylation of CD4+ T cells in systemic lupus erythematosus by indirectly targeting DNA methyltransferase 1. J Dermatol Sci. 2013; 69:61–67.
Article
50. Dai Y, Huang YS, Tang M, Lv TY, Hu CX, Tan YH, Xu ZM, Yin YB. Microarray analysis of microRNA expression in peripheral blood cells of systemic lupus erythematosus patients. Lupus. 2007; 16:939–946.
Article
51. Sheedy FJ, Palsson-McDermott E, Hennessy EJ, Martin C, O'Leary JJ, Ruan Q, Johnson DS, Chen Y, O'Neill LA. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat Immunol. 2010; 11:141–147.
Article
52. Ding S, Liang Y, Zhao M, Liang G, Long H, Zhao S, Wang Y, Yin H, Zhang P, Zhang Q, Lu Q. Decreased microRNA-142-3p/5p expression causes CD4+ T cell activation and B cell hyperstimulation in systemic lupus erythematosus. Arthritis Rheum. 2012; 64:2953–2963.
Article
53. Cannons JL, Qi H, Lu KT, Dutta M, Gomez-Rodriguez J, Cheng J, Wakeland EK, Germain RN, Schwartzberg PL. Optimal germinal center responses require a multistage T cell: B cell adhesion process involving integrins, SLAM-associated protein, and CD84. Immunity. 2010; 32:253–265.
Article
54. Komori H, Furukawa H, Mori S, Ito MR, Terada M, Zhang MC, Ishii N, Sakuma N, Nose M, Ono M. A signal adaptor SLAM-associated protein regulates spontaneous autoimmunity and Fas-dependent lymphoproliferation in MRL-Faslpr lupus mice. J Immunol. 2006; 176:395–400.
Article
55. Alcocer-Varela J, Alarcón-Segovia D. Decreased production of and response to interleukin-2 by cultured lymphocytes from patients with systemic lupus erythematosus. J Clin Invest. 1982; 69:1388–1392.
Article
56. Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012; 12:180–190.
Article
57. Fan W, Liang D, Tang Y, Qu B, Cui H, Luo X, Huang X, Chen S, Higgs BW, Jallal B, Yao Y, Harley JB, Shen N. Identification of microRNA-31 as a novel regulator contributing to impaired interleukin-2 production in T cells from patients with systemic lupus erythematosus. Arthritis Rheum. 2012; 64:3715–3725.
Article
58. Xue F, Li H, Zhang J, Lu J, Xia Y, Xia Q. miR-31 regulates interleukin 2 and kinase suppressor of ras 2 during T cell activation. Genes Immun. 2013; 14:127–131.
Article
59. Helms WS, Jeffrey JL, Holmes DA, Townsend MB, Clipstone NA, Su L. Modulation of NFAT-dependent gene expression by the RhoA signaling pathway in T cells. J Leukoc Biol. 2007; 82:361–369.
Article
60. Rouas R, Fayyad-Kazan H, El Zein N, Lewalle P, Rothé F, Simion A, Akl H, Mourtada M, El Rifai M, Burny A, Romero P, Martiat P, Badran B. Human natural Treg microRNA signature: role of microRNA-31 and microRNA-21 in FOXP3 expression. Eur J Immunol. 2009; 39:1608–1618.
Article
61. Zhao X, Tang Y, Qu B, Cui H, Wang S, Wang L, Luo X, Huang X, Li J, Chen S, Shen N. MicroRNA-125a contributes to elevated inflammatory chemokine RANTES levels via targeting KLF13 in systemic lupus erythematosus. Arthritis Rheum. 2010; 62:3425–3435.
Article
62. Lu MM, Wang J, Pan HF, Chen GM, Li J, Cen H, Feng CC, Ye DQ. Increased serum RANTES in patients with systemic lupus erythematosus. Rheumatol Int. 2012; 32:1231–1233.
Article
63. Song A, Chen YF, Thamatrakoln K, Storm TA, Krensky AM. RFLAT-1: a new zinc finger transcription factor that activates RANTES gene expression in T lymphocytes. Immunity. 1999; 10:93–103.
64. Liu Y, Dong J, Mu R, Gao Y, Tan X, Li Y, Li Z, Yang G. MicroRNA-30a promotes B cell hyperactivity in patients with systemic lupus erythematosus by direct interaction with Lyn. Arthritis Rheum. 2013; 65:1603–1611.
Article
65. Liossis SN, Solomou EE, Dimopoulos MA, Panayiotidis P, Mavrikakis MM, Sfikakis PP. B-cell kinase lyn deficiency in patients with systemic lupus erythematosus. J Investig Med. 2001; 49:157–165.
Article
66. Flores-Borja F, Kabouridis PS, Jury EC, Isenberg DA, Mageed RA. Decreased Lyn expression and translocation to lipid raft signaling domains in B lymphocytes from patients with systemic lupus erythematosus. Arthritis Rheum. 2005; 52:3955–3965.
Article
67. Zan H, Casali P. Regulation of Aicda expression and AID activity. Autoimmunity. 2013; 46:83–101.
68. Diaz M. The role of activation-induced deaminase in lupus nephritis. Autoimmunity. 2013; 46:115–120.
Article
69. Jiang C, Foley J, Clayton N, Kissling G, Jokinen M, Herbert R, Diaz M. Abrogation of lupus nephritis in activation-induced deaminase-deficient MRL/lpr mice. J Immunol. 2007; 178:7422–7431.
Article
70. Dorsett Y, McBride KM, Jankovic M, Gazumyan A, Thai TH, Robbiani DF, Di Virgilio M, Reina San-Martin B, Heidkamp G, Schwickert TA, Eisenreich T, Rajewsky K, Nussenzweig MC. MicroRNA-155 suppresses activation-induced cytidine deaminase-mediated Myc-Igh translocation. Immunity. 2008; 28:630–638.
Article
71. Teng G, Hakimpour P, Landgraf P, Rice A, Tuschl T, Casellas R, Papavasiliou FN. MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity. 2008; 28:621–629.
Article
72. De Yébenes VG, Belver L, Pisano DG, González S, Villasante A, Croce C, He L, Ramiro AR. miR-181b negatively regulates activation-induced cytidine deaminase in B cells. J Exp Med. 2008; 205:2199–2206.
Article
73. Dai R, Zhang Y, Khan D, Heid B, Caudell D, Crasta O, Ahmed SA. Identification of a common lupus disease-associated microRNA expression pattern in three different murine models of lupus. PLoS One. 2010; 5:e14302.
Article
74. Thai TH, Patterson HC, Pham DH, Kis-Toth K, Kaminski DA, Tsokos GC. Deletion of microRNA-155 reduces autoantibody responses and alleviates lupus-like disease in the Fas(lpr) mouse. Proc Natl Acad Sci U S A. 2013; 110:20194–20199.
Article
75. Yuan Y, Kasar S, Underbayev C, Vollenweider D, Salerno E, Kotenko SV, Raveche E. Role of microRNA-15a in autoantibody production in interferon- augmented murine model of lupus. Mol Immunol. 2012; 52:61–70.
Article
76. Boldin MP, Taganov KD, Rao DS, Yang L, Zhao JL, Kalwani M, Garcia-Flores Y, Luong M, Devrekanli A, Xu J, Sun G, Tay J, Linsley PS, Baltimore D. miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J Exp Med. 2011; 208:1189–1201.
Article
77. Hou J, Wang P, Lin L, Liu X, Ma F, An H, Wang Z, Cao X. MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J Immunol. 2009; 183:2150–2158.
Article
78. Luo X, Yang W, Ye DQ, Cui H, Zhang Y, Hirankarn N, Qian X, Tang Y, Lau YL, de Vries N, Tak PP, Tsao BP, Shen N. A functional variant in microRNA-146a promoter modulates its expression and confers disease risk for systemic lupus erythematosus. PLoS Genet. 2011; 7:e1002128.
Article
79. Karrich JJ, Jachimowski LC, Libouban M, Iyer A, Brandwijk K, Taanman-Kueter EW, Nagasawa M, de Jong EC, Uittenbogaart CH, Blom B. MicroRNA-146a regulates survival and maturation of human plasmacytoid dendritic cells. Blood. 2013; 122:3001–3009.
Article
80. Chan VS, Nie YJ, Shen N, Yan S, Mok MY, Lau CS. Distinct roles of myeloid and plasmacytoid dendritic cells in systemic lupus erythematosus. Autoimmun Rev. 2012; 11:890–897.
Article
81. Charrier E, Cordeiro P, Cordeau M, Dardari R, Michaud A, Harnois M, Merindol N, Herblot S, Duval M. Post-transcriptional down-regulation of Toll-like receptor signaling pathway in umbilical cord blood plasmacytoid dendritic cells. Cell Immunol. 2012; 276:114–121.
Article
82. Jin O, Kavikondala S, Sun L, Fu R, Mok MY, Chan A, Yeung J, Lau CS. Systemic lupus erythematosus patients have increased number of circulating plasmacytoid dendritic cells, but decreased myeloid dendritic cells with deficient CD83 expression. Lupus. 2008; 17:654–662.
Article
83. Jin O, Kavikondala S, Mok MY, Sun L, Gu J, Fu R, Chan A, Yeung J, Nie Y, Lau CS. Abnormalities in circulating plasmacytoid dendritic cells in patients with systemic lupus erythematosus. Arthritis Res Ther. 2010; 12:R137.
Article
84. Pan Y, Jia T, Zhang Y, Zhang K, Zhang R, Li J, Wang L. MS2 VLP-based delivery of microRNA-146a inhibits autoantibody production in lupus-prone mice. Int J Nanomedicine. 2012; 7:5957–5967.
Article
85. Garchow BG, Bartulos Encinas O, Leung YT, Tsao PY, Eisenberg RA, Caricchio R, Obad S, Petri A, Kauppinen S, Kiriakidou M. Silencing of microRNA-21 in vivo ameliorates autoimmune splenomegaly in lupus mice. EMBO Mol Med. 2011; 3:605–615.
Article
86. Keller A, Leidinger P, Bauer A, Elsharawy A, Haas J, Backes C, Wendschlag A, Giese N, Tjaden C, Ott K, Werner J, Hackert T, Ruprecht K, Huwer H, Huebers J, Jacobs G, Rosenstiel P, Dommisch H, Schaefer A, Müller-Quernheim J, Wullich B, Keck B, Graf N, Reichrath J, Vogel B, Nebel A, Jager SU, Staehler P, Amarantos I, Boisguerin V, Staehler C, Beier M, Scheffler M, Büchler MW, Wischhusen J, Haeusler SFM, Dietl J, Hofmann S, Lenhof HP, Schreiber S, Katus HA, Rottbauer W, Meder B, Hoheisel JD, Franke A, Meese E. Toward the blood-borne miRNome of human diseases. Nat Methods. 2011; 8:841–843.
Article
87. Su X, Qian C, Zhang Q, Hou J, Gu Y, Han Y, Chen Y, Jiang M, Cao X. miRNomes of haematopoietic stem cells and dendritic cells identify miR-30b as a regulator of Notch1. Nat Commun. 2013; 4:2903.
Article
Full Text Links
  • IN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr