651
|
Subedi A, Park PH. Autocrine and paracrine modulation of microRNA-155 expression by globular adiponectin in RAW 264.7 macrophages: involvement of MAPK/NF-κB pathway. Cytokine 2013; 64:638-41. [PMID: 24084329 DOI: 10.1016/j.cyto.2013.09.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/31/2013] [Accepted: 09/06/2013] [Indexed: 12/11/2022]
Abstract
Adiponectin, a hormone produced from adipose tissue, regulates various biological responses, including inflammation and many metabolic processes. MicroRNAs control expression of diverse target genes and various physiological responses. Many of these responses are commonly regulated by adiponectin. However, effects of adiponectin on microRNAs regulation are largely unknown. Herein we demonstrated that globular adiponectin induces increase in miR-155 expression, which plays an important role in inflammatory response, in RAW 264.7 macrophages. We further showed that this effect was modulated by and MAPK/NF-κB dependent mechanisms. These results suggest that miR-155 would be a novel promising target mediating adiponectin-induced various biological responses.
Collapse
Affiliation(s)
- Amit Subedi
- College of Pharmacy, Yeungnam University, Gyeongsanbuk-do 712-749, Republic of Korea
| | | |
Collapse
|
652
|
Vigorito E, Kohlhaas S, Lu D, Leyland R. miR-155: an ancient regulator of the immune system. Immunol Rev 2013; 253:146-57. [PMID: 23550644 DOI: 10.1111/imr.12057] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are a newly recognized class of regulatory genes which repress the expression of protein-coding genes. Numerous studies have uncovered a complex role for miRNAs regulating many aspects of a variety of cellular processes including cell growth, differentiation, and lineage commitment. In the immune system, miR-155 is unique in its ability to shape the transcriptome of activated myeloid and lymphoid cells controlling diverse biological functions ranging from inflammation to immunological memory. Not surprisingly, a tight control of miR-155 expression is required to avoid malignant transformation, as evidenced by miR-155 overexpression in many cancers of B-cell origin. In this review, we discuss the potential of miR-155 as a molecular target for therapeutic intervention and discuss the function of miR-155 in the context of protective immunity. We first look back into the emergence of miR-155 in evolution, which is coincidental with the emergence of the ancestors of the antigen receptors. We then summarize what we have learned about the role of miR-155 in the regulation of lymphoid subsets at the cellular and molecular level in the context of recent progress in this field.
Collapse
Affiliation(s)
- Elena Vigorito
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | | | | | | |
Collapse
|
653
|
Dooley J, Linterman MA, Liston A. MicroRNA regulation of T-cell development. Immunol Rev 2013; 253:53-64. [PMID: 23550638 DOI: 10.1111/imr.12049] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
MicroRNAs are short, 19-24 nucleotide long, RNA molecules capable of regulating the longevity and, to a lesser extent, translation of messenger RNA (mRNA) species. The function of the microRNA network, and indeed, even that of individual microRNA species, can have profoundly different roles in even a single cell type as the microRNA/mRNA composition evolves. As the role of microRNA within T cells has come under increasing scrutiny, several distinct checkpoints have been demonstrated to have a particular reliance on microRNA regulation. MicroRNAs are arguably most important in T cells during the earliest and last stages in T-cell biology. The first stages of early thymic differentiation have a crucial reliance on the microRNA network, while later stages and peripheral homeostasis are largely, although not completely, microRNA-independent. The most profound effects on T cells are in the activation of effector and regulatory functions of conventional and regulatory T cells, where microRNA deficiency results in a near-complete loss of function. In this review, we focus on integrating the research on individual microRNA into a more global understanding of the function of the microRNA regulatory network in T cells.
Collapse
Affiliation(s)
- James Dooley
- Autoimmune Genetics Laboratory, VIB, Leuven, Belgium
| | | | | |
Collapse
|
654
|
Du L, Rong H, Cheng Y, Guo S, Shi Q, Jia X, Zhu H, Hao Y, Xu K, Zhang J, Jiao H, Zhao T, Zhang H, Chen C, Wang F. Identification of MicroRNAs Dysregulated in CD14 Gene Silencing RAW264.7 Macrophage Cells. Inflammation 2013; 37:287-94. [DOI: 10.1007/s10753-013-9739-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
655
|
Naqvi AR, Fordham JB, Khan A, Nares S. MicroRNAs responsive to Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis LPS modulate expression of genes regulating innate immunity in human macrophages. Innate Immun 2013; 20:540-51. [PMID: 24062196 DOI: 10.1177/1753425913501914] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 07/26/2013] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small, noncoding RNAs that regulate post-transcriptional expression of their respective target genes and are responsive to various stimuli, including LPS. Here we examined the early (4 h) miRNA responses of THP1-differentiated macrophages challenged with LPS derived from the periodontal pathogens, Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis or environmentally-modified LPS obtained from P. gingivalis grown in cigarette smoke extract. Predicted miRNA-gene target interactions for LPS-responsive miR-29b and let-7f were confirmed using dual-luciferase assays and by transfection experiments using miRNA mimics and inhibitors. Convergent and divergent miRNA profiles were observed in treated samples where differences in miRNA levels related to the type, concentration and incubation times of LPS challenge. Dual-luciferase experiments revealed miR-29b targeting of interleukin-6 receptorα (IL-6Rα) and IFN-γ inducible protein 30 and let-7f targeting of suppressor of cytokine signaling 4 and thrombospondin-1. Transfection experiments confirmed miR-29b and let-7f modulation of IL-6Rα and SOCS4 protein expression levels, respectively. Thus, we have demonstrated convergent/divergent miRNA responses to wild type LPS and its environmentally-modified LPS, and demonstrate miRNA targeting of key genes linked to inflammation and immunity. Our data indicate that these LPS-responsive miRNAs may play a key role in fine-tuning the host response to periodontal pathogens.
Collapse
Affiliation(s)
- Afsar R Naqvi
- Department of Periodontology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA Currently at Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jezrom B Fordham
- Department of Periodontology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA Currently at Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Asma Khan
- Department of Endodontics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Salvador Nares
- Department of Periodontology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA Currently at Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
656
|
Upregulated microRNA-155 expression in peripheral blood mononuclear cells and fibroblast-like synoviocytes in rheumatoid arthritis. Clin Dev Immunol 2013; 2013:296139. [PMID: 24151514 PMCID: PMC3789322 DOI: 10.1155/2013/296139] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/19/2013] [Indexed: 11/17/2022]
Abstract
OBJECTIVE This study was to screen for the miRNAs differently expressed in peripheral blood mononuclear cells (PBMC) of RA, to further identify the expression of miR-155 in RA PBMC and fibroblast-like synoviocytes (FLS), and to evaluate the function of miR-155 in RA-FLS. METHODS Microarray was used to screen for differentially expressed miRNAs in RA PBMC. miR-155 expression in PBMC and FLS of RA were identified by real-time PCR. Enforced overexpression and downexpression of miR-155 were used to investigate the function of miR-155 in RA-FLS. Expression of IKBKE which was previously identified as the actual target of miR-155 was examined by Western blot and real-time PCR in RA-FLS. RESULTS miR-155 levels were increased in both PBMC and FLS of RA and could be induced by TNF- α . Upregulation of miR-155 decreased MMP-3 levels and suppressed proliferation and invasion of RA-FLS. Inverse relationship between the expressions of miR-155 and the MMPs production-related protein IKBKE was found. CONCLUSION An inflammatory milieu may alter miRNA expression profiles in rheumatoid arthritis. miR-155 is upregulated in RA-FLS, and it may be a protective factor against the inflammatory effect in part by attenuating expression of IKBKE.
Collapse
|
657
|
Yu Q, Zhu S, Zhou R, Yi F, Bing Y, Huang S, Wang Z, Wang C, Xia B. Effects of sinomenine on the expression of microRNA-155 in 2,4,6-trinitrobenzenesulfonic acid-induced colitis in mice. PLoS One 2013; 8:e73757. [PMID: 24066068 PMCID: PMC3774766 DOI: 10.1371/journal.pone.0073757] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/22/2013] [Indexed: 01/31/2023] Open
Abstract
Background Sinomenine, a pure alkaloid isolated in Chinese medicine from the root of Sinomenium acutum, has been demonstrated to have anti-inflammatory and immunosuppressive effects. MicroRNAs (miRNAs) are gradually being recognized as critical mediators of disease pathogenesis via coordinated regulation of molecular effector pathways. Methodology/Findings After colitis was induced in mice by instillation of 5% (w/v) 2,4,6-trinitrobenzenesulfonic acid (TNBS), sinomenine at a dose of 100 or 200 mg/kg was orally administered once daily for 7 days. We evaluated body weight, survival rate, diarrhea score, histological score and myeloperoxidase (MPO) activity. The mRNA and protein expression levels of miR-155, c-Maf, TNF-α and IFN-γ were determined by quantitative RT-PCR and immunohistochemistry, respectively. Sinomenine (100 or 200 mg/kg)-treated mice with TNBS-induced colitis were significantly improved in terms of body weight, survival rate, diarrhea score, histological score and MPO activity compared with untreated mice. Both dosages of sinomenine significantly decreased the mRNA and protein expression levels of c-Maf, TNF-α and IFN-γ, which elevated in TNBS-induced colitis. Furthermore, sinomenine at a dose of 200 mg/kg significantly decreased the level of miR-155 expression by 71% (p = 0.025) compared with untreated TNBS-induced colitis in mice. Conclusions/Significance Our study evaluated the effects and potential mechanisms of sinomenine in the anti-inflammatory response via miRNA-155 in mice with TNBS-induced colitis. Our findings suggest that sinomenine has anti-inflammatory effects on TNBS-induced colitis by down-regulating the levels of miR-155 and several related inflammatory cytokines.
Collapse
Affiliation(s)
- Qiao Yu
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, P.R. of China
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Siying Zhu
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, P.R. of China
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Rui Zhou
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, P.R. of China
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Fengming Yi
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, P.R. of China
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Yuntao Bing
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Sha Huang
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, P.R. of China
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Zixi Wang
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Chunyu Wang
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
| | - Bing Xia
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, P.R. of China
- The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, P.R. of China
- * E-mail:
| |
Collapse
|
658
|
Wujcicka W, Wilczyński J, Nowakowska D. Alterations in TLRs as new molecular markers of congenital infections with Human cytomegalovirus? Pathog Dis 2013; 70:3-16. [PMID: 23929630 DOI: 10.1111/2049-632x.12083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 07/28/2013] [Accepted: 07/31/2013] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptors (TLRs) play a crucial role in non-specific immunity against various infections. The most common intrauterine infection, caused by Human cytomegalovirus (HCMV), results in perinatal morbidity and mortality of primary infected fetuses. The induction of immune response by TLRs was observed in HCMV infections in murine models and cell lines cultured in vitro. Studies reported an immunological response in pregnant women with primary HCMV infection and TLR2 activity in collecting of HCMV particles in placental syncytiotrophoblasts (STs) in vivo and cultured ST, and in stimulation of tumor necrosis factor (TNF)-α expression and damage of villous trophoblast. Expression levels of TLRs are associated with cell type, stage of pregnancy and response to microorganisms. We show the effect of HCMV infection on the development of pregnancy as well as the effect of TLR single-nucleotide polymorphisms on the occurrence and course of infectious diseases, immune response and diseases of pregnancy. We report the impact of TLRs on the function of miRNAs and the altered expression levels of these molecules, as observed in HCMV infections. We suggest that the methylation status of TLR gene promoter regions as epigenetic modifications may be significant in the immune response to HCMV infections. We conclude that it is important to study in detail the molecular mechanisms of TLR function in the immune response to HCMV infections in pregnancy.
Collapse
Affiliation(s)
- Wioletta Wujcicka
- Department of Fetal-Maternal Medicine and Gynecology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | | | | |
Collapse
|
659
|
Zhang Y, Li YK. MicroRNAs in the regulation of immune response against infections. J Zhejiang Univ Sci B 2013; 14:1-7. [PMID: 23303626 DOI: 10.1631/jzus.b1200292] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Innate immunity is considered to provide the initial defense against infections by viruses, bacteria, fungi, and protozoa. Detection of the signature molecules of invading pathogens by front-line defense cells via various germline-encoded pattern recognition receptors (PRRs) is needed to activate intracellular signaling cascades that lead to transcriptional expression of inflammatory mediators to coordinate the elimination of pathogens and infected cells. To maintain a fine balance between protective immunity and inflammatory pathology upon infection, the innate signaling pathways in the host need to be tightly regulated. MicroRNAs (miRNAs), a new class of small non-coding RNAs, have been recently shown to be potent modulators that function at post-transcriptional levels. Accumulating evidence demonstrates that the involvement of microorganism-encoded and host miRNAs might play instructive roles in the immune response upon infection. Here, we discuss the current knowledge of miRNAs in the regulation of immune response against infections.
Collapse
Affiliation(s)
- Yue Zhang
- Department of General Surgery, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | | |
Collapse
|
660
|
Zampetaki A, Dudek K, Mayr M. Oxidative stress in atherosclerosis: the role of microRNAs in arterial remodeling. Free Radic Biol Med 2013; 64:69-77. [PMID: 23797034 DOI: 10.1016/j.freeradbiomed.2013.06.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is the underlying condition in most cardiovascular diseases. Among the highly specific cellular and molecular responses, endothelial dysfunction plays a key role in disease initiation and progression. These events coincide with the occurrence of oxidative stress. Increased reactive oxygen species production and oxidization of low-density lipoprotein are detected throughout atherosclerosis progression. MicroRNAs (miRNAs) have emerged as important regulators of gene expression that posttranscriptionally modify cellular responses and function. Accumulating studies indicate an integrated miRNA network in the molecular mechanisms that control cellular homeostasis, vascular inflammation, and metabolism. Experimental models of atherosclerosis highlight a direct link between altered miRNA expression profiles and the pathophysiology of the disease and identify putative miRNA candidates for the development of novel therapeutic strategies. In this review, we provide an overview of the role of miRNA regulatory networks in oxidative stress in atherosclerosis and arterial remodeling and discuss their potential therapeutic implications.
Collapse
Affiliation(s)
- Anna Zampetaki
- King's British Heart Foundation Centre, King's College London, London SE5 9NU, UK.
| | | | | |
Collapse
|
661
|
Li S, Zhu S, Li C, Zhang Z, Zhou L, Wang S, Wang S, Zhang Y, Wen X. Characterization of microRNAs in mud crab Scylla paramamosain under Vibrio parahaemolyticus infection. PLoS One 2013; 8:e73392. [PMID: 24023678 PMCID: PMC3758354 DOI: 10.1371/journal.pone.0073392] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 07/23/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Infection of bacterial Vibrio parahaemolyticus is common in mud crab farms. However, the mechanisms of the crab's response to pathogenic V. parahaemolyticus infection are not fully understood. MicroRNAs (miRNAs) are a class of small noncoding RNAs that function as regulators of gene expression and play essential roles in various biological processes. To understand the underlying mechanisms of the molecular immune response of the crab to the pathogens, high-throughput Illumina/Solexa deep sequencing technology was used to investigate the expression profiles of miRNAs in S. paramamosain under V. parahaemolyticus infection. METHODOLOGY/PRINCIPAL FINDINGS Two mixed RNA pools of 7 tissues (intestine, heart, liver, gill, brain, muscle and blood) were obtained from V. parahaemolyticus infected crabs and the control groups, respectively. By aligning the sequencing data with known miRNAs, we characterized 421 miRNA families, and 133 conserved miRNA families in mud crab S. paramamosain were either identical or very similar to existing miRNAs in miRBase. Stem-loop qRT-PCRs were used to scan the expression levels of four randomly chosen differentially expressed miRNAs and tissue distribution. Eight novel potential miRNAs were confirmed by qRT-PCR analysis and the precursors of these novel miRNAs were verified by PCR amplification, cloning and sequencing in S. paramamosain. 161 miRNAs (106 of which up-regulated and 55 down-regulated) were significantly differentially expressed during the challenge and the potential targets of these differentially expressed miRNAs were predicted. Furthermore, we demonstrated evolutionary conservation of mud crab miRNAs in the animal evolution process. CONCLUSIONS/SIGNIFICANCE In this study, a large number of miRNAs were identified in S. paramamosain when challenged with V. parahaemolyticus, some of which were differentially expressed. The results show that miRNAs might play some important roles in regulating gene expression in mud crab under V. parahaemolyticus infection, providing a basis for further investigation of miRNA-modulating networks in innate immunity of mud crab.
Collapse
Affiliation(s)
- Shengkang Li
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
| | - Shuo Zhu
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
- Department of Biology, College of Science, Shantou University, Shantou, China
| | - Chuanbiao Li
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
| | - Zhao Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
| | - Lizhen Zhou
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
| | - Shijia Wang
- Department of Biology, College of Science, Shantou University, Shantou, China
| | - Shuqi Wang
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
- Department of Biology, College of Science, Shantou University, Shantou, China
| | - Xiaobo Wen
- Guangdong Provincial Key Laboratory of Marine Biology, Marine Biology Institute, Shantou University, Shantou, China
- * E-mail:
| |
Collapse
|
662
|
Wang Z, Han J, Cui Y, Zhou X, Fan K. miRNA-21 inhibition enhances RANTES and IP-10 release in MCF-7 via PIAS3 and STAT3 signalling and causes increased lymphocyte migration. Biochem Biophys Res Commun 2013; 439:384-9. [PMID: 23998932 DOI: 10.1016/j.bbrc.2013.08.072] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are a class of small endogenous gene regulators that have been implicated in various developmental and pathological processes. However, the precise identities and functions of miRNAs involved in antitumor immunity are not yet well understood. miRNA-21 is an oncogenic miRNA that can be detected in various tumours. In this study, we report that a miRNA-21 inhibitor enhances the release of chemoattractants RANTES and IP-10 in the MCF-7 breast cancer cell line and results in increased lymphocyte migration. Thus, miRNA-21 is a potential therapeutic target for cancer immunotherapy. We further demonstrated that PIAS3, a protein inhibitor of activated STAT3, is a target of miRNA-21 in MCF-7. Thus, miRNA-21 is a novel miRNA regulating immune cell recruitment, which acts at least in part via its inhibition of PIAS3 expression and oncogenic STAT3 signalling in tumour cells.
Collapse
Affiliation(s)
- Zhiyu Wang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, Shandong, PR China; Shandong Medicinal Biotechnology Center, Key Laboratory for Biotech-Drugs, Ministry of Health, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, PR China
| | | | | | | | | |
Collapse
|
663
|
MicroRNA-124 mediates the cholinergic anti-inflammatory action through inhibiting the production of pro-inflammatory cytokines. Cell Res 2013; 23:1270-83. [PMID: 23979021 PMCID: PMC3817544 DOI: 10.1038/cr.2013.116] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 07/04/2013] [Accepted: 07/15/2013] [Indexed: 12/22/2022] Open
Abstract
The vagus nerve can control inflammatory response through a 'cholinergic anti-inflammatory pathway', which is mediated by the α7-nicotinic acetylcholine receptor (α7nAChR) on macrophages. However, the intracellular mechanisms that link α7nAChR activation and pro-inflammatory cytokine production remain not well understood. In this study, we found that miR-124 is upregulated by cholinergic agonists in LPS-exposed cells and mice. Utilizing miR-124 mimic and siRNA knockdown, we demonstrated that miR-124 is a critical mediator for the cholinergic anti-inflammatory action. Furthermore, our data indicated that miR-124 modulates LPS-induced cytokine production by targeting signal transducer and activator of transcription 3 (STAT3) to decrease IL-6 production and TNF-α converting enzyme (TACE) to reduce TNF-α release. These results also indicate that miR-124 is a potential therapeutic target for the treatment of inflammatory diseases.
Collapse
|
664
|
Plank M, Maltby S, Mattes J, Foster PS. Targeting translational control as a novel way to treat inflammatory disease: the emerging role of MicroRNAs. Clin Exp Allergy 2013. [DOI: 10.1111/cea.12135] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- 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
| | - S. 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
| | | | - 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
| |
Collapse
|
665
|
Hocès de la Guardia A, Staedel C, Kaafarany I, Clément A, Roubaud Baudron C, Mégraud F, Lehours P. Inflammatory cytokine and microRNA responses of primary human dendritic cells cultured with Helicobacter pylori strains. Front Microbiol 2013; 4:236. [PMID: 23970881 PMCID: PMC3747313 DOI: 10.3389/fmicb.2013.00236] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/30/2013] [Indexed: 12/31/2022] Open
Abstract
Primary human dendritic cells (DC) were used to explore the inflammatory effectors, including cytokines and microRNAs, regulated by Helicobacter pylori. In a 48 h ex-vivo co-culture system, both H. pylori B38 and B45 strains activated human DCs and promoted a strong inflammatory response characterized by the early production of pro-inflammatory TNFα and IL-6 cytokines, followed by IL-10, IL-1β, and IL-23 secretion. IL-23 was the only cytokine dependent on the cag pathogenicity island status of the bacterial strains. DC activation and cytokine production were accompanied by an early miR-146a upregulation followed by a strong miR-155 induction, which mainly controlled TNFα production. These results pave the way for further investigations into the nature of H. pylori antigens and the subsequently activated signaling pathways involved in the inflammatory response to H. pylori infection, the deregulation of which may likely contribute to gastric lymphomagenesis.
Collapse
Affiliation(s)
- Anaïs Hocès de la Guardia
- Bacteriology Laboratory, Université Bordeaux Bordeaux, France ; Institut National de la Santé et de la Recherche Médicale, U853 Bordeaux, France
| | | | | | | | | | | | | |
Collapse
|
666
|
Harris JF, Micheva-Viteva S, Li N, Hong-Geller E. Small RNA-mediated regulation of host-pathogen interactions. Virulence 2013; 4:785-95. [PMID: 23958954 PMCID: PMC3925712 DOI: 10.4161/viru.26119] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The rise in antimicrobial drug resistance, alongside the failure of conventional research to discover new antibiotics, will inevitably lead to a public health crisis that can drastically curtail our ability to combat infectious disease. Thus, there is a great global health need for development of antimicrobial countermeasures that target novel cell molecules or processes. RNA represents a largely unexploited category of potential targets for antimicrobial design. For decades, control of cellular behavior was thought to be the exclusive purview of protein-based regulators. The recent discovery of small RNAs (sRNAs) as a universal class of powerful RNA-based regulatory biomolecules has the potential to revolutionize our understanding of gene regulation in practically all biological functions. In general, sRNAs regulate gene expression by base-pairing with multiple downstream target mRNAs to prevent translation of mRNA into protein. In this review, we will discuss recent studies that document discovery of bacterial, viral, and human sRNAs and their molecular mechanisms in regulation of pathogen virulence and host immunity. Illuminating the functional roles of sRNAs in virulence and host immunity can provide the fundamental knowledge for development of next-generation antibiotics using sRNAs as novel targets.
Collapse
Affiliation(s)
- Jennifer F Harris
- Bioscience Division; Los Alamos National Laboratory; Los Alamos, NM USA
| | | | - Nan Li
- Bioscience Division; Los Alamos National Laboratory; Los Alamos, NM USA
| | | |
Collapse
|
667
|
Heymans S, Corsten MF, Verhesen W, Carai P, van Leeuwen REW, Custers K, Peters T, Hazebroek M, Stöger L, Wijnands E, Janssen BJ, Creemers EE, Pinto YM, Grimm D, Schürmann N, Vigorito E, Thum T, Stassen F, Yin X, Mayr M, de Windt LJ, Lutgens E, Wouters K, de Winther MPJ, Zacchigna S, Giacca M, van Bilsen M, Papageorgiou AP, Schroen B. Macrophage microRNA-155 promotes cardiac hypertrophy and failure. Circulation 2013; 128:1420-32. [PMID: 23956210 DOI: 10.1161/circulationaha.112.001357] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiac hypertrophy and subsequent heart failure triggered by chronic hypertension represent major challenges for cardiovascular research. Beyond neurohormonal and myocyte signaling pathways, growing evidence suggests inflammatory signaling pathways as therapeutically targetable contributors to this process. We recently reported that microRNA-155 is a key mediator of cardiac inflammation and injury in infectious myocarditis. Here, we investigated the impact of microRNA-155 manipulation in hypertensive heart disease. METHODS AND RESULTS Genetic loss or pharmacological inhibition of the leukocyte-expressed microRNA-155 in mice markedly reduced cardiac inflammation, hypertrophy, and dysfunction on pressure overload. These alterations were macrophage dependent because in vivo cardiomyocyte-specific microRNA-155 manipulation did not affect cardiac hypertrophy or dysfunction, whereas bone marrow transplantation from wild-type mice into microRNA-155 knockout animals rescued the hypertrophic response of the cardiomyocytes and vice versa. In vitro, media from microRNA-155 knockout macrophages blocked the hypertrophic growth of stimulated cardiomyocytes, confirming that macrophages influence myocyte growth in a microRNA-155-dependent paracrine manner. These effects were at least partly mediated by the direct microRNA-155 target suppressor of cytokine signaling 1 (Socs1) because Socs1 knockdown in microRNA-155 knockout macrophages largely restored their hypertrophy-stimulating potency. CONCLUSIONS Our findings reveal that microRNA-155 expression in macrophages promotes cardiac inflammation, hypertrophy, and failure in response to pressure overload. These data support the causative significance of inflammatory signaling in hypertrophic heart disease and demonstrate the feasibility of therapeutic microRNA targeting of inflammation in heart failure.
Collapse
Affiliation(s)
- Stephane Heymans
- Center for Heart Failure Research, Department of Cardiology (S.H., M.F.C., W.V., P.C., R.E.W.v.L., K.C., T.P., M.H., M.v.B., A.-P.P., B.S.), Department of Molecular Genetics (L.S., M.P.J.d.W.), Department of Pathology (E.W., E.L.), Department of Pharmacology (B.J.J.), Department of Cardiology (L.J.d.W.), and Department of Internal Medicine (K.W.), Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, the Netherlands (S.H.); Center for Molecular and Cardiovascular Biology, Department of Cardiovascular Sciences, Leuven, Belgium (S.H., P.C., A.-P.P.); Department of Medical Biochemistry (L.S., E.L., M.P.J.d.W.) and Heart Failure Research Center (E.E.C., Y.M.P.), Academic Medical Center, Amsterdam, the Netherlands; Cluster of Excellence Cell Networks, Department of Infectious Diseases/Virology, Virus Host Interactions, Heidelberg University, Heidelberg, Germany (D.G., N.S.); Laboratory of Lymphocyte Signaling and Development, Babraham Institute, Cambridge, UK (E.V.); Institute for Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany (T.T.); Medical Microbiology, Maastricht University, Maastricht, the Netherlands (F.S.); King's BHF Centre, King's College London, London UK (X.Y., M.M.); Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany (E.L.); and Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., M.G.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
668
|
Cheung ST, So EY, Chang D, Ming-Lum A, Mui ALF. Interleukin-10 inhibits lipopolysaccharide induced miR-155 precursor stability and maturation. PLoS One 2013; 8:e71336. [PMID: 23951138 PMCID: PMC3741136 DOI: 10.1371/journal.pone.0071336] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 06/25/2013] [Indexed: 12/16/2022] Open
Abstract
The anti-inflammatory cytokine interleukin-10 (IL-10) is essential for attenuating the inflammatory response, which includes reducing the expression of pro-inflammatory microRNA-155 (miR-155) in lipopolysaccharide (LPS) activated macrophages. miR-155 enhances the expression of pro-inflammatory cytokines such as TNFα and suppresses expression of anti-inflammatory molecules such as SOCS1. Therefore, we examined the mechanism by which IL-10 inhibits miR-155. We found that IL-10 treatment did not affect the transcription of the miR-155 host gene nor the nuclear export of pre-miR-155, but rather destabilized both pri-miR-155 and pre-miR-155 transcripts, as well as interfered with the final maturation of miR-155. This inhibitory effect of IL-10 on miR-155 expression involved the contribution of both the STAT3 transcription factor and the phosphoinositol phosphatase SHIP1. This is the first report showing evidence that IL-10 regulates miRNA expression post-transcriptionally.
Collapse
Affiliation(s)
- Sylvia T. Cheung
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eva Y. So
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Chang
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew Ming-Lum
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alice L-F. Mui
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
669
|
Lippai D, Bala S, Csak T, Kurt-Jones EA, Szabo G. Chronic alcohol-induced microRNA-155 contributes to neuroinflammation in a TLR4-dependent manner in mice. PLoS One 2013; 8:e70945. [PMID: 23951048 PMCID: PMC3739772 DOI: 10.1371/journal.pone.0070945] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 06/25/2013] [Indexed: 01/28/2023] Open
Abstract
Introduction Alcohol-induced neuroinflammation is mediated by pro-inflammatory cytokines and chemokines including tumor necrosis factor-α (TNFα), monocyte chemotactic protein-1 (MCP1) and interleukin-1-beta (IL-1β). Toll-like receptor-4 (TLR4) pathway induced nuclear factor-κB (NF-κB) activation is involved in the pathogenesis of alcohol-induced neuroinflammation. Inflammation is a highly regulated process. Recent studies suggest that microRNAs (miRNAs) play crucial role in fine tuning gene expression and miR-155 is a major regulator of inflammation in immune cells after TLR stimulation. Aim To evaluate the role of miR-155 in the pathogenesis of alcohol-induced neuroinflammation. Methods Wild type (WT), miR-155- and TLR4-knockout (KO) mice received 5% ethanol-containing or isocaloric control diet for 5 weeks. Microglia markers were measured by q-RTPCR; inflammasome activation was measured by enzyme activity; TNFα, MCP1, IL-1β mRNA and protein were measured by q-RTPCR and ELISA; phospho-p65 protein and NF-κB were measured by Western-blotting and EMSA; miRNAs were measured by q-PCR in the cerebellum. MiR-155 was measured in immortalized and primary mouse microglia after lipopolysaccharide and ethanol stimulation. Results Chronic ethanol feeding up-regulated miR-155 and miR-132 expression in mouse cerebellum. Deficiency in miR-155 protected mice from alcohol-induced increase in inflammatory cytokines; TNFα, MCP1 protein and TNFα, MCP1, pro-IL-1β and pro-caspase-1 mRNA levels were reduced in miR-155 KO alcohol-fed mice. NF-κB was activated in WT but not in miR-155 KO alcohol-fed mice. However increases in cerebellar caspase-1 activity and IL-1β levels were similar in alcohol-fed miR-155-KO and WT mice. Alcohol-fed TLR4-KO mice were protected from the induction of miR-155. NF-κB activation measured by phosphorylation of p65 and neuroinflammation were reduced in alcohol-fed TLR4-KO compared to control mice. TLR4 stimulation with lipopolysaccharide in primary or immortalized mouse microglia resulted in increased miR-155. Conclusion Chronic alcohol induces miR-155 in the cerebellum in a TLR4-dependent manner. Alcohol-induced miR-155 regulates TNFα and MCP1 expression but not caspase-dependent IL-1β increase in neuroinflammation.
Collapse
Affiliation(s)
- Dora Lippai
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Shashi Bala
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Timea Csak
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Evelyn A. Kurt-Jones
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
670
|
Li C, Liu T, Qi F, Li F, Zhu L, Wang P, Wang H. Analysis of intragraft MicroRNA expression in a mouse-to-rat cardiac xenotransplantation model. Microsurgery 2013; 34:44-50. [PMID: 23913343 DOI: 10.1002/micr.22139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 04/29/2013] [Accepted: 05/03/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Chuan Li
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - Tong Liu
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - Feng Qi
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - Fuxin Li
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - Liwei Zhu
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - Pengzhi Wang
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| | - Hao Wang
- Department of General Surgery, Tianjin General Surgery Institute; Tianjin Medical University General Hospital; Tianjin China
| |
Collapse
|
671
|
Ding J, Huang S, Wang Y, Tian Q, Zha R, Shi H, Wang Q, Ge C, Chen T, Zhao Y, Liang L, Li J, He X. Genome-wide screening reveals that miR-195 targets the TNF-α/NF-κB pathway by down-regulating IκB kinase alpha and TAB3 in hepatocellular carcinoma. Hepatology 2013; 58:654-66. [PMID: 23487264 DOI: 10.1002/hep.26378] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/02/2013] [Indexed: 12/16/2022]
Abstract
UNLABELLED Nuclear factor kappa B (NF-κB) is an important factor linking inflammation and tumorigenesis. In this study we experimentally demonstrated through a high-throughput luciferase reporter screen that NF-κB signaling can be directly targeted by nearly 29 microRNAs (miRNAs). Many of these miRNAs can directly target NF-κB signaling nodes by binding to their 3' untranslated region (UTR). miR-195, a member of the miR-15 family, is frequently down-regulated in gastrointestinal cancers, especially in hepatocellular carcinoma (HCC). The expression level of miR-195 is inversely correlated with HCC tumor size. We further show that miR-195 suppresses cancer cell proliferation and migration in vitro and reduces tumorigenicity and metastasis in vivo. Additionally, miR-195 may exert its tumor suppressive function by decreasing the expression of multiple NF-κB downstream effectors by way of the direct targeting of IKKα and TAB3. CONCLUSION Multiple miRNAs are involved in the NF-κB signaling pathway and miR-195 plays important inhibitory roles in cancer progression and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Jie Ding
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
672
|
Increased micro-RNA 29b in the aged brain correlates with the reduction of insulin-like growth factor-1 and fractalkine ligand. Neurobiol Aging 2013; 34:2748-58. [PMID: 23880139 DOI: 10.1016/j.neurobiolaging.2013.06.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/28/2013] [Accepted: 06/14/2013] [Indexed: 12/13/2022]
Abstract
Microglia develop an inflammatory phenotype during normal aging. The mechanism by which this occurs is not well understood, but might be related to impairments in several key immunoregulatory systems. Here we show that micro-RNA (miR)-29a and miR-29b, 2 immunoregulatory micro-RNAs, were increased in the brain of aged BALB/c mice compared with adults. Insulin-like growth factor-1 (IGF-1) and fractalkine ligand (CX3CL1) are negative modulators of microglial activation and were identified as targets of miR-29a and miR-29b using luciferase assay and primary microglia transfection. Indeed, higher expression of miR-29b in the brain of aged mice was associated with reduced messenger RNA (mRNA) levels of IGF-1 and CX3CL1. Parallel to these results in mice, miR-29a and miR-29b were also markedly increased in cortical brain tissue of older individuals (mean, 77 years) compared with middle-aged adults (mean, 45 years). Moreover, increased expression of miR-29b in human cortical tissue was negatively correlated with IGF-1 and CX3CL1 expression. Collectively, these data indicate that an age-associated increase in miR-29 corresponded with the reduction of 2 important regulators of microglia, IGF-1 and CX3CL1.
Collapse
|
673
|
Mor E, Shomron N. Species-specific microRNA regulation influences phenotypic variability: perspectives on species-specific microRNA regulation. Bioessays 2013; 35:881-8. [PMID: 23864354 DOI: 10.1002/bies.201200157] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Phenotypic divergence among animal species may be due in part to species-specific (SS) regulation of gene expression by small, non-coding regulatory RNAs termed "microRNAs". This phenomenon can be modulated by several variables. First, microRNA genes vary by their level of conservation, many of them being SS, or unique to a particular evolutionary lineage. Second, microRNA expression levels vary spatially and temporally in different species. Lastly, while microRNAs bind the 3'UTR of target genes in order to silence their expression, the binding sites themselves are often non-conserved. The variability of the miRNA-target paradigm between different species is thus multifactorial, and this paradigm has only just started to gain attention from researchers in various fields. Here we present and discuss recent findings regarding the characteristics and implications of SS microRNA regulation.
Collapse
Affiliation(s)
- Eyal Mor
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | |
Collapse
|
674
|
Zeng Z, Gong H, Li Y, Jie K, Ding C, Shao Q, Liu F, Zhan Y, Nie C, Zhu W, Qian K. Upregulation of miR-146a contributes to the suppression of inflammatory responses in LPS-induced acute lung injury. Exp Lung Res 2013; 39:275-82. [DOI: 10.3109/01902148.2013.808285] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
675
|
Aalaei-andabili SH, Rezaei N. Toll like receptor (TLR)-induced differential expression of microRNAs (MiRs) promotes proper immune response against infections: a systematic review. J Infect 2013; 67:251-64. [PMID: 23850616 DOI: 10.1016/j.jinf.2013.07.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/25/2013] [Accepted: 07/06/2013] [Indexed: 12/19/2022]
Abstract
Toll like receptors (TLRs) are one of the major families of pattern recognition receptors (PRRs). MicroRNAs (MiRs) are small noncoding RNAs with regulatory effects on biological process, and it has been recently shown that they can control inflammatory process and the response to an infection by modulating the function of TLRs. In this study, we designed a systematic review to clarify the reciprocal interaction between TLRs and MiRs, in order to identify possible future therapeutic targets and strategies. On the one hand, TLRs stimulation can change expression level of miRs in various ways, which can lead to modulating their effects. On the other hand, MiRs also influence the expression of TLRs and the intensity of the inflammatory reaction. We therefore conclude that the interaction between MiRs and TLRs is a key regulator of innate immune system. Investigations discovering therapeutic approaches by manipulation of miRs expression level may open a new approach for the treatment of inflammatory diseases.
Collapse
|
676
|
Staedel C, Darfeuille F. MicroRNAs and bacterial infection. Cell Microbiol 2013; 15:1496-507. [PMID: 23795564 DOI: 10.1111/cmi.12159] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
Abstract
MicroRNAs, small non-coding RNAs expressed by eukaryotic cells, play pivotal roles in shaping cell differentiation and organism development. Deregulated microRNA expression is associated with several types of diseases including cancers, immune disorders and infection. Acting at the post-transcriptional level, miRNAs have expanded our understanding of the control of gene expression in regulatory networks involved in the adaptation to environmental situations such as biotic stress. It is increasingly clear that miRNAs are an important part of the host response to microbes. This review presents the current state of knowledge about the role of miRNAs in the response to both bacterial pathogens and commensal bacteria in human cells or animal experimental models. Some microRNAs, including miR-146, miR-155, miR-125, let-7 and miR-21, are commonly affected during bacterial infection and contribute to immune responses protecting the organism against overwhelmed inflammation. Cell-specific relationships between miRNAs and their targets are also engaged in the alterations induced by virulent bacteria in the proliferation/differentiation/apoptosis pathways of their host cells. In a separate role, miRNA modulation also represents a mechanism through which commensal bacteria impact the regulation of the barrier function and intestinal homeostasis.
Collapse
Affiliation(s)
- Cathy Staedel
- Univ. Bordeaux, ARNA Laboratory, F-33000, Bordeaux, France.
| | | |
Collapse
|
677
|
MicroRNAs implicated in the immunopathogenesis of lupus nephritis. Clin Dev Immunol 2013; 2013:430239. [PMID: 23983769 PMCID: PMC3741610 DOI: 10.1155/2013/430239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 05/20/2013] [Accepted: 06/12/2013] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the deposition of immune complexes due to widespread loss of immune tolerance to nuclear self-antigens. Deposition in the renal glomeruli results in the development of lupus nephritis (LN), the leading cause of morbidity and mortality in SLE. In addition to the well-recognized genetic susceptibility to SLE, disease pathogenesis is influenced by epigenetic regulators such as microRNAs (miRNAs). miRNAs are small, noncoding RNAs that bind to the 3′ untranslated region of target mRNAs resulting in posttranscriptional gene modulation. miRNAs play an important and dynamic role in the activation of innate immune cells and are critical in regulating the adaptive immune response. Immune stimulation and the resulting cytokine milieu alter miRNA expression while miRNAs themselves modify cellular responses to stimulation. Here we examine dysregulated miRNAs implicated in LN pathogenesis from human SLE patients and murine lupus models. The effects of LN-associated miRNAs in the kidney, peripheral blood mononuclear cells, macrophages, mesangial cells, dendritic cells, and splenocytes are discussed. As the role of miRNAs in immunopathogenesis becomes delineated, it is likely that specific miRNAs may serve as targets for therapeutic intervention in the treatment of LN and other pathologies.
Collapse
|
678
|
TLR3-induced placental miR-210 down-regulates the STAT6/interleukin-4 pathway. PLoS One 2013; 8:e67760. [PMID: 23844087 PMCID: PMC3699491 DOI: 10.1371/journal.pone.0067760] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/21/2013] [Indexed: 11/19/2022] Open
Abstract
Several clinical studies have reported increased placental miR-210 expression in women with PE compared to normotensive women, but whether miR-210 plays a role in the etiology of PE is unknown. We reported that activation of TLR3 produces the PE-like symptoms of hypertension, endothelial dysfunction, and proteinuria in mice only when pregnant, but whether TLR3 activation in pregnant mice and human cytotrophoblasts (CTBs) increases miR-210 and modulates its targets related to inflammation are unknown. Placental miR-210 levels were increased significantly in pregnant mice treated with the TLR3 agonist poly I:C (P-PIC). Both HIF-1α and NF-κBp50, known to bind the miR-210 promoter and induce its expression, were also increased significantly in placentas of P-PIC mice. Target identification algorithms and gene ontology predicted STAT6 as an inflammation-related target of miR-210 and STAT6 was decreased significantly in placentas of P-PIC mice. IL-4, which is regulated by STAT6 and increases during normotensive pregnancy, failed to increase in serum of P-PIC mice. P-PIC TLR3 KO mice did not develop hypertension and placental HIF-1α, NF-κBp50, miR-210, STAT6, and IL-4 levels were unchanged. To determine the placental etiology, treatment of human CTBs with poly I:C significantly increased HIF-1α, NF-κBp50, and miR-210 levels and decreased STAT6 and IL-4 levels. Overexpression of miR-210 in CTBs decreased STAT6 and IL-4 while inhibition of miR-210 increased STAT6 and IL-4. These findings demonstrate that TLR3 activation induces placental miR-210 via HIF-1α and NF-κBp50 leading to decreased STAT6 and IL-4 levels and this may contribute to the development of PE.
Collapse
|
679
|
miRNA signature of mouse helper T cell hyper-proliferation. PLoS One 2013; 8:e66709. [PMID: 23825558 PMCID: PMC3692518 DOI: 10.1371/journal.pone.0066709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 05/09/2013] [Indexed: 11/29/2022] Open
Abstract
Helper T cells from a mutant mouse model, LAT Y136F, hyper-proliferate and cause a severe lymphoproliferative disease that kills the mice by six months of age. LAT Y136F mice carry a tyrosine to phenylalanine mutation in the Linker for Activation of T cells (LAT) gene. This mutation leads to a number of changes in T cells that result in altered cytokine production including increased IL-4 production, increased proliferation, and decreased apoptosis. Hyper-proliferation of the mutant T cells contributes to lymphadenopathy, splenomegaly, and multi-organ T cell infiltration. miRNAs are short non-coding RNAs that regulate expression of cohorts of genes. This study investigates which miRNAs are expressed in LAT Y136F T cells and compares these to miRNAs expressed in wild type T cells that are undergoing proliferation in two other settings. The first setting is homeostatic proliferation, which was modeled by adoptive transfer of wild type T cells into T cell-deficient mice. The second setting is proliferation in response to infection, which was modeled by infection of wild type mice with the nematode H. polygyrus. By comparing miRNA expression in these three proliferative states (LAT Y136F hyper-proliferation, homeostatic proliferation and proliferation in response to H. polygyrus infection) to expression in wild type naïve CD4+ T cells, we found miRNAs that were highly regulated in all three proliferative states (miR-21 and miR-146a) and some that were more specific to individual settings of proliferation such as those more specific for LAT Y136F lymphoproliferative disease (miR-669f, miR-155 and miR-466a/b). Future experiments that modulate levels of the miRNAs identified in this study may reveal the roles of these miRNAs in T cell proliferation and/or lymphoproliferative disease.
Collapse
|
680
|
Sittka A, Schmeck B. MicroRNAs in the lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 774:121-34. [PMID: 23377971 DOI: 10.1007/978-94-007-5590-1_7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The lung constitutes one of the most delicate tissue structures in mammalian organisms to accomplish the vital function of gas exchange. On the other hand, its immense surface area, necessary in this context, exhibits the first line of defense against a variety of pro-inflammatory stimuli.MicroRNAs (miRNAs) are a class of post-transcriptional regulators that revolutionized our view of gene expression regulation. By now, it is well established that miRNAs impair all known cellular and developmental processes. Extensive research over the last years revealed not only a fundamental role for miRNAs in lung development and homeostasis, but also in the process of lung inflammation. Lung inflammation occurs in response to stimuli very different in nature (e.g., physical, radioactive, infective, pro-allergenic, or toxic), and in some cases becomes manifest in chronic diseases (e.g., chronic bronchitis/chronic obstructive pulmonary disease (COPD), asthma and allergic airway diseases) or even lung cancer.This review chapter will briefly describe the current knowledge concerning miRNA expression and their exerted target regulation in the course of lung inflammation and lung cancer.
Collapse
Affiliation(s)
- Alexandra Sittka
- Department of Molecular Pulmonology, Philipps-University Marburg, Marburg, Germany.
| | | |
Collapse
|
681
|
Galli R, Paone A, Fabbri M, Zanesi N, Calore F, Cascione L, Acunzo M, Stoppacciaro A, Tubaro A, Lovat F, Gasparini P, Fadda P, Alder H, Volinia S, Filippini A, Ziparo E, Riccioli A, Croce CM. Toll-like receptor 3 (TLR3) activation induces microRNA-dependent reexpression of functional RARβ and tumor regression. Proc Natl Acad Sci U S A 2013; 110:9812-9817. [PMID: 23716670 PMCID: PMC3683754 DOI: 10.1073/pnas.1304610110] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Toll-like receptor 3 (TLR3) is a key effector of the innate immune system against viruses. Activation of TLR3 exerts an antitumoral effect through a mechanism of action still poorly understood. Here we show that TLR3 activation by polyinosinic:polycytidylic acid induces up-regulation of microRNA-29b, -29c, -148b, and -152 in tumor-derived cell lines and primary tumors. In turn, these microRNAs induce reexpression of epigenetically silenced genes by targeting DNA methyltransferases. In DU145 and TRAMP-C1 prostate and MDA-MB-231 breast cancer cells, we demonstrated that polyinosinic:polycytidylic acid-mediated activation of TLR3 induces microRNAs targeting DNA methyltransferases, leading to demethylation and reexpression of the oncosuppressor retinoic acid receptor beta (RARβ). As a result, cancer cells become sensitive to retinoic acid and undergo apoptosis both in vitro and in vivo. This study provides evidence of an antitumoral mechanism of action upon TLR3 activation and the biological rationale for a combined TLR3 agonist/retinoic acid treatment of prostate and breast cancer.
Collapse
Affiliation(s)
- Roberta Galli
- Istituto Pasteur–Fondazione Cenci Bolognetti, Department of Anatomical, Histological, Forensic & Orthopaedic Sciences (DAHFMO) Unit of Histology and Medical Embryology, La Sapienza University of Rome, 00161 Rome, Italy
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Alessio Paone
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Muller Fabbri
- Children’s Center for Cancer and Blood Diseases and Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center and Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
- Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, CA 90027
| | - Nicola Zanesi
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Federica Calore
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Luciano Cascione
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
- Department of Clinical and Molecular Biomedicine, University of Catania, 95122 Catania, Italy
| | - Mario Acunzo
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Antonella Stoppacciaro
- Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, La Sapienza University, Italy; and
| | - Andrea Tubaro
- Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, La Sapienza University, Italy; and
| | - Francesca Lovat
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Pierluigi Gasparini
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Paolo Fadda
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Hansjuerg Alder
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| | - Stefano Volinia
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
- Department of Morphology and Embryology, University of Ferrara, 44100 Ferrara, Italy
| | - Antonio Filippini
- Istituto Pasteur–Fondazione Cenci Bolognetti, Department of Anatomical, Histological, Forensic & Orthopaedic Sciences (DAHFMO) Unit of Histology and Medical Embryology, La Sapienza University of Rome, 00161 Rome, Italy
| | - Elio Ziparo
- Istituto Pasteur–Fondazione Cenci Bolognetti, Department of Anatomical, Histological, Forensic & Orthopaedic Sciences (DAHFMO) Unit of Histology and Medical Embryology, La Sapienza University of Rome, 00161 Rome, Italy
| | - Anna Riccioli
- Istituto Pasteur–Fondazione Cenci Bolognetti, Department of Anatomical, Histological, Forensic & Orthopaedic Sciences (DAHFMO) Unit of Histology and Medical Embryology, La Sapienza University of Rome, 00161 Rome, Italy
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology, and Medical Genetics and Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210
| |
Collapse
|
682
|
El Gazzar M. microRNAs as potential regulators of myeloid-derived suppressor cell expansion. Innate Immun 2013; 20:227-38. [PMID: 23757323 DOI: 10.1177/1753425913489850] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Proper development and activation of cells of the myeloid lineage are critical for supporting innate immunity. This myelopoiesis is orchestrated by interdependent interactions between cytokine receptors, transcription factors and, as recently described, microRNAs (miRNAs). miRNAs contribute to normal and dysregulated myelopoiesis. Alterations in myelopoiesis underlie myeloid-derived suppressor cell (MDSC) expansion, a poorly understood heterogeneous population of immature and suppressive myeloid cells that expand in nearly all diseases where inflammation exists. MDSCs associated with inflammation often have immunosuppressive properties, but molecular mechanisms responsible for MDSC expansion are unclear. Emerging data implicate miRNAs in MDSC expansion. This review focuses on miRNAs that contribute to myeloid lineage differentiation and maturation under physiological conditions, and introduces the concept that altered miRNA expression my underlie expansion and accumulation of MDSCs. We divide our miRNAs into those with potential to promote MDSC expansion and two with known direct links to MDSC expansion, miR-223 and miR-494.
Collapse
Affiliation(s)
- Mohamed El Gazzar
- Department of Internal Medicine, East Tennessee State University College of Medicine, Johnson City, TN, USA
| |
Collapse
|
683
|
Wolbachia uses a host microRNA to regulate transcripts of a methyltransferase, contributing to dengue virus inhibition in Aedes aegypti. Proc Natl Acad Sci U S A 2013; 110:10276-81. [PMID: 23733960 DOI: 10.1073/pnas.1303603110] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The endosymbiont Wolbachia is common among insects and known for the reproductive manipulations it exerts on hosts as well as inhibition of virus replication in their hosts. Recently, we showed that Wolbachia uses host microRNAs to manipulate host gene expression for its efficient maintenance in the dengue mosquito vector, Aedes aegypti. Cytosine methylation is mediated by a group of proteins called DNA (cytosine-5) methyltransferases, which are structurally and functionally conserved from prokaryotes to eukaryotes. The biological functions of cytosine methylation include host defense, genome stability, gene regulation, developmental promotion of organs, and lifespan regulation. Ae. aegypti has only one DNA methyltransferase gene (AaDnmt2) belonging to the cytosine methyltransferase family 2, which is the most deeply conserved and widely distributed gene among metazoans. Here, we show that in mosquitoes the introduced endosymbiont, Wolbachia, significantly suppresses expression of AaDnmt2, but dengue virus induces expression of AaDnmt2. Interestingly, we found that aae-miR-2940 microRNA, which is exclusively expressed in Wolbachia-infected mosquitoes, down-regulates the expression of AaDnmt2. Reversely, overexpression of AaDnmt2 in mosquito cells led to inhibition of Wolbachia replication, but significantly promoted replication of dengue virus, suggesting a causal link between this Wolbachia manipulation and the blocking of dengue replication in Wolbachia-infected mosquitoes. In addition, our findings provide an explanation for hypomethylation of the genome in Wolbachia-infected Ae. aegypti.
Collapse
|
684
|
Abstract
BACKGROUND MicroRNA-132 (miR-132) targets acetylcholinesterase (AChE) and potentiates the cholinergic blockade of inflammatory reactions in cultured cells and experimental mice, but the implications of this interaction to human inflammatory disease remained unexplored. This study aimed to test whether miR-132 is causally involved in anti-inflammatory reactions of patients with inflammatory bowel disease (IBD) and modulates vagal tone and consequently inflammation in patients with IBD. METHODS We prospectively measured inflammation readouts and the cholinergic status (total capacity for hydrolyzing acetylcholine in one's circulation), and AChE activity in 2 independent cohorts of patients with IBD and quantified miR-132 levels in intestinal tissue biopsies removed at colonoscopy from inflamed and apparently quiescent tissues of tested volunteers. RESULTS MiR-132 levels are higher in inflamed compared with apparently quiescent intestinal biopsies from patients with IBD. Correspondingly, the cholinergic status and AChE activity was significantly lower in patients with IBD suffering from moderate-severe disease as compared with healthy controls or patient with IBD presenting low disease severity. Patients with IBD (n = 16) presented lower AChE activity compared with healthy controls (n = 33; 289 ± 128 AU versus 391 ± 102 AU, P = 0.001), and a negative correlation between AChE activity and C-reactive protein levels (r = -0.47, P = 0.01). Corroborating these observations in an additional cohort of participants, C-reactive protein and AChE activity were negatively correlated in patients with moderate-severe disease (n = 16; r = -0.6, P = 0.04) and positively correlated in healthy controls (n = 74, r = 0.24, P = 0.046). CONCLUSIONS Taken together, these findings support an inflammation-dependent homeostatic role for the regulation by miR-132 of AChE in IBD, opening new venues for therapeutic interference.
Collapse
|
685
|
Philippe L, Alsaleh G, Pichot A, Ostermann E, Zuber G, Frisch B, Sibilia J, Pfeffer S, Bahram S, Wachsmann D, Georgel P. MiR-20a regulates ASK1 expression and TLR4-dependent cytokine release in rheumatoid fibroblast-like synoviocytes. Ann Rheum Dis 2013; 72:1071-9. [PMID: 23087182 DOI: 10.1136/annrheumdis-2012-201654] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To evaluate whether miR-20a belonging to the cluster miR-17-92 is a negative regulator of inflammation in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS) by modulating expression of apoptosis signal-regulating kinase (ASK) 1, a key component of the toll-like receptors 4 pathway, upstream of p38 mitogen-activated protein kinase. METHODS Evaluation of miR-20a and ASK1 mRNA was performed by RT-qPCR. ASK1 protein expression was assessed by western blotting. Overexpression of miR-20a was performed by transfection of RA FLS and THP-1 cells with miR-20a mimics. Interleukin (IL)-6, CXCL-10, IL-1β and TNF-α release were measured by ELISA. The role of miR-20a in vivo was assessed by IL-6 release from macrophages obtained from mice injected intraperitoneally with vectorised miR-20a mimics. RESULTS We showed that stimulation of RA FLS with lipopolysacharide (LPS) and bacterial lipoproteins (BLP) induces a drop in expression of miR-20a and that this decrease is associated with an upregulation of ASK1 expression. Using transfection of Ask1 3'UTR reporters, we demonstrate that Ask1 is a direct target of miR-20a. Overexpression of miR-20a led to a global decrease in ASK1 protein in BLP- and LPS-activated cells indicating that miR-20a regulates the expression of ASK1 at the translational level. Transfection of miR-20a mimics decreases IL-6 and CXCL10 release by RA FLS and IL-1β and TNF-α by activated THP-1 cells but only in response to LPS. Last, injection of vectorised miR-20a mimics to mice led to a global decrease in ASK1 protein expression and IL-6 secretion in LPS-activated macrophages. CONCLUSIONS Our data point toward an important role for miR-20a in the regulation of pro-inflammatory cytokines release, by controlling ASK1 expression in RA FLS.
Collapse
Affiliation(s)
- Lucas Philippe
- ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Université de Strasbourg, Strasbourg, Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
686
|
Rebane A, Akdis CA. MicroRNAs: Essential players in the regulation of inflammation. J Allergy Clin Immunol 2013; 132:15-26. [PMID: 23726263 DOI: 10.1016/j.jaci.2013.04.011] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/02/2013] [Accepted: 04/04/2013] [Indexed: 12/16/2022]
Abstract
Regulation of inflammatory responses is ensured by coordinated control of gene expression in participating immune system and tissue cells. One group of gene expression regulators, the functions of which have recently been started to be uncovered in relation to any type of inflammatory condition, is a class of short single-stranded RNA molecules termed microRNAs (miRNAs). miRNAs function together with partner proteins and mainly cause gene silencing through degradation of target mRNAs or inhibition of translation. A particular miRNA can have hundreds of target genes, and thereby miRNAs together influence the expression of a large proportion of proteins. The role of miRNAs in the immune system has been extensively studied since the discovery of miRNAs in mammalian cells approximately 10 years ago. The purpose of the current review is to provide an overview on the functions of miRNAs in the regulation of inflammation, with a specific focus on the mechanisms of allergic inflammation. Because recent studies clearly demonstrate the presence of extracellular miRNAs in body fluids and propose the involvement of miRNAs in cell-cell communication, we will also highlight findings about functions of extracellular miRNAs. The possible use of miRNAs as biomarkers, as well as miRNA-related novel treatment modalities, might open a new future for the diagnosis and treatment of many inflammatory conditions, including allergic diseases.
Collapse
Affiliation(s)
- Ana Rebane
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland.
| | | |
Collapse
|
687
|
Trenkmann M, Brock M, Gay RE, Michel BA, Gay S, Huber LC. Tumor necrosis factor α-induced microRNA-18a activates rheumatoid arthritis synovial fibroblasts through a feedback loop in NF-κB signaling. ACTA ACUST UNITED AC 2013; 65:916-27. [PMID: 23280137 DOI: 10.1002/art.37834] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 12/13/2012] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To elucidate whether the microRNA (miRNA) cluster miR-17-92 contributes to the activated phenotype of rheumatoid arthritis synovial fibroblasts (RASFs). METHODS RASFs were stimulated with tumor necrosis factor α (TNFα), and the expression and regulation of the miR-17-92 cluster were studied using real-time quantitative PCR (PCR) and promoter activity assays. RASFs were transfected with single precursor molecules of miRNAs from miR-17-92 and the expression of matrix-degrading enzymes and cytokines was measured by quantitative PCR and enzyme-linked immunosorbent assay. Potential miRNA targets were identified by computational prediction and were validated using reporter gene assays and Western blotting. The activity of NF-κB signaling was determined by reporter gene assays. RESULTS We found that TNFα induces the expression of miR-17-92 in RASFs in an NF-κB-dependent manner. Transfection of RASFs with precursor molecules of single members of miR-17-92 revealed significantly increased expression levels of matrix-degrading enzymes, proinflammatory cytokines, and chemokines in precursor miR-18a (pre-miR-18a)-transfected RASFs. Using reporter gene assays, we identified the NF-κB pathway inhibitor TNFα-induced protein 3 as a new target of miR-18a. In addition, pre-miR-18a-transfected RASFs showed stronger activation of NF-κB signaling, both constitutively and in response to TNFα stimulation. CONCLUSION Our data suggest that the miR-17-92-derived miR-18a contributes to cartilage destruction and chronic inflammation in the joint through a positive feedback loop in NF-κB signaling, with concomitant up-regulation of matrix-degrading enzymes and mediators of inflammation in RASFs.
Collapse
Affiliation(s)
- Michelle Trenkmann
- Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | |
Collapse
|
688
|
Xue P, Zheng M, Diao Z, Shen L, Liu M, Gong P, Sun H, Hu Y. miR-155* mediates suppressive effect of PTEN 3'-untranslated region on AP-1/NF-κB pathway in HTR-8/SVneo cells. Placenta 2013; 34:650-6. [PMID: 23684381 DOI: 10.1016/j.placenta.2013.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/30/2013] [Accepted: 04/23/2013] [Indexed: 11/30/2022]
Abstract
Among miRNAs, miR-155 is a known regulator of immune system. Accumulating studies have revealed the connections between miR-155 and activator protein 1 (AP-1)/nuclear factor (NF)-κB. However, miR-155*, a miR-155 paralog, has so far been less studied. Here we demonstrated that miR-155*, induced by lipopolysaccharide (LPS) in an AP-1/NF-κB dependent manner, played a positive feedback role in AP-1/NF-κB pathway via targeting interleukin-1 receptor-associated kinase M (IRAKM) and NF-κB inhibitor interacting Ras-like 1 (NKIRAS1) in trophoblasts. Our study further proved that miR-155*-targeted PTEN 3'-untranslated region (3'UTR) increased IRAKM and NKIRAS1 expression by competing for miR-155* binding, thereby suppressing AP-1/NF-κB activation induced by LPS.
Collapse
Affiliation(s)
- P Xue
- Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | |
Collapse
|
689
|
Ashhab MU, Omran A, Kong H, Gan N, He F, Peng J, Yin F. Expressions of tumor necrosis factor alpha and microRNA-155 in immature rat model of status epilepticus and children with mesial temporal lobe epilepsy. J Mol Neurosci 2013; 51:950-8. [PMID: 23636891 DOI: 10.1007/s12031-013-0013-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/08/2013] [Indexed: 12/28/2022]
Abstract
Recently, the role of inflammation has attracted great attention in the pathogenesis of mesial temporal lobe epilepsy (MTLE), and microRNAs start to emerge as promising new players in MTLE pathogenesis. In this study, we investigated the dynamic expression patterns of tumor necrosis factor alpha (TNF-α) and microRNA-155 (miR-155) in the hippocampi of an immature rat model of status epilepticus (SE) and children with MTLE. The expressions of TNF-α and miR-155 were significantly upregulated in the seizure-related acute and chronic stages of MTLE in the immature rat model and also in children with MTLE. Modulation of TNF-α expression, either by stimulation using myeloid-related protein (MRP8) or lipopolysaccharide or inhibition using lenalidomide on astrocytes, leads to similar dynamic changes in miR-155 expression. Our study is the first to focus on the dynamic expression pattern of miR-155 in the immature rat of SE lithium-pilocarpine model and children with MTLE and to detect their relationship at the astrocyte level. TNF-α and miR-155, having similar expression patterns in the three stages of MTLE development, and their relationship at the astrocyte level may suggest a direct interactive relationship during MTLE development. Therefore, modulation of the TNF-α/miR-155 axis may be a novel therapeutic target for the treatment of MTLE.
Collapse
Affiliation(s)
- Muhammad Usman Ashhab
- Department of Pediatrics, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, China
| | | | | | | | | | | | | |
Collapse
|
690
|
Zhu S, Pan W, Qian Y. MicroRNA in immunity and autoimmunity. J Mol Med (Berl) 2013; 91:1039-50. [PMID: 23636510 DOI: 10.1007/s00109-013-1043-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 04/12/2013] [Accepted: 04/15/2013] [Indexed: 01/17/2023]
Abstract
MicroRNAs (miRNAs) are about 20-22 nucleotide conserved non-coding RNA molecules that post-transcriptionally regulate gene expression by targeting the 3'-untranslated region of specific messenger RNAs (mRNAs) for degradation or translational repression. During the last two decades, miRNAs have emerged as critical regulators of a range of biological processes including immune cell lineage commitment, differentiation, maturation, and immune signaling pathways. The endoribonucleases such as Dicer, which is required for miRNA biogenesis, has also been shown to play an important role in inflammatory response and autoimmunity. Thus, dysregulated miRNA expression patterns have been documented in a broad range of human diseases including inflammatory and autoimmune diseases. In this review, we will discuss recent advances in miRNAs mediated regulation of inflammatory responses and autoimmune pathogenesis. Specifically, we will discuss how miRNAs regulate autoimmunity through affecting the development, differentiation, and function of various cell types such as innate immune cells, adaptive immune cells and local resident cells. The identification of distinct miRNA expression patterns, and a comprehensive understanding of the roles of those dysregulated miRNAs in inflammatory autoimmune pathogenesis offers inspirations of not only potential molecular diagnostic markers but also novel therapeutic strategies for treating inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Shu Zhu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | | | | |
Collapse
|
691
|
Abstract
Yin and Yang are two complementary forces that together describe the nature of real-world elements. Yin is the dark side; Yang is the light side. We describe microRNAs having both Yin and Yang characteristics because they can contribute to normal function (Yang) but also to autoimmunity, myeloproliferation, and cancer (Yin). We have been working on a number of microRNAs that have these dual characteristics and here we focus on two, miR-125b and miR-146a. We have concentrated on these two RNAs because we have very extensive knowledge of them, much of it from our laboratory, and also because they provide a strong contrast: the effects of overexpression of miR-125b are rapid, suggesting that it acts directly, whereas the effects of miR-146a are slow to develop, suggesting that they arise from chronic alterations in cellular behavior.
Collapse
Affiliation(s)
- Alex Yick-Lun So
- Department of Biology, California Institute of Technology, Pasadena, CA, USA
| | - Jimmy Zhao
- Department of Biology, California Institute of Technology, Pasadena, CA, USA
| | - David Baltimore
- Department of Biology, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
692
|
Moelants EAV, Mortier A, Van Damme J, Proost P. Regulation of TNF-α with a focus on rheumatoid arthritis. Immunol Cell Biol 2013; 91:393-401. [PMID: 23628802 DOI: 10.1038/icb.2013.15] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 03/21/2013] [Accepted: 03/23/2013] [Indexed: 12/13/2022]
Abstract
Cytokines and chemokines represent two important groups of proteins that control the human immune system. Dysregulation of the network in which these immunomodulators function can result in uncontrolled inflammation, leading to various diseases including rheumatoid arthritis (RA), characterized by chronic inflammation and bone erosion. Potential triggers of RA include autoantibodies, cytokines and chemokines. The tight regulation of cytokine and chemokine production, and biological activity is important. Tumor necrosis factor-α (TNF-α) is abundantly present in RA patients' serum and the arthritic synovium. This review, therefore, discusses first the role and regulation of the major proinflammatory cytokine TNF-α, in particular the regulation of TNF-α production, post-translational processing and signaling of TNF-α and its receptors. Owing to the important role of TNF-α in RA, the TNF-α-producing cells and the dynamics of its expression, the direct and indirect action of this cytokine and possible biological therapy for RA are described.
Collapse
Affiliation(s)
- Eva A V Moelants
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
693
|
Halkein J, Tabruyn SP, Ricke-Hoch M, Haghikia A, Nguyen NQN, Scherr M, Castermans K, Malvaux L, Lambert V, Thiry M, Sliwa K, Noel A, Martial JA, Hilfiker-Kleiner D, Struman I. MicroRNA-146a is a therapeutic target and biomarker for peripartum cardiomyopathy. J Clin Invest 2013; 123:2143-54. [PMID: 23619365 DOI: 10.1172/jci64365] [Citation(s) in RCA: 367] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 02/07/2013] [Indexed: 12/11/2022] Open
Abstract
Peripartum cardiomyopathy (PPCM) is a life-threatening pregnancy-associated cardiomyopathy in previously healthy women. Although PPCM is driven in part by the 16-kDa N-terminal prolactin fragment (16K PRL), the underlying molecular mechanisms are poorly understood. We found that 16K PRL induced microRNA-146a (miR-146a) expression in ECs, which attenuated angiogenesis through downregulation of NRAS. 16K PRL stimulated the release of miR-146a-loaded exosomes from ECs. The exosomes were absorbed by cardiomyocytes, increasing miR-146a levels, which resulted in a subsequent decrease in metabolic activity and decreased expression of Erbb4, Notch1, and Irak1. Mice with cardiomyocyte-restricted Stat3 knockout (CKO mice) exhibited a PPCM-like phenotype and displayed increased cardiac miR-146a expression with coincident downregulation of Erbb4, Nras, Notch1, and Irak1. Blocking miR-146a with locked nucleic acids or antago-miRs attenuated PPCM in CKO mice without interrupting full-length prolactin signaling, as indicated by normal nursing activities. Finally, miR-146a was elevated in the plasma and hearts of PPCM patients, but not in patients with dilated cardiomyopathy. These results demonstrate that miR-146a is a downstream-mediator of 16K PRL that could potentially serve as a biomarker and therapeutic target for PPCM.
Collapse
Affiliation(s)
- Julie Halkein
- Unit of Molecular Biology and Genetic Engineering, GIGA, University of Liège, Liège, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
694
|
The microRNA miR-155 controls CD8(+) T cell responses by regulating interferon signaling. Nat Immunol 2013; 14:593-602. [PMID: 23603793 PMCID: PMC3664306 DOI: 10.1038/ni.2576] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 02/26/2013] [Indexed: 02/07/2023]
Abstract
We found upregulation of expression of the microRNA miR-155 in primary effector and effector memory CD8(+) T cells, but low miR-155 expression in naive and central memory cells. Antiviral CD8(+) T cell responses and viral clearance were impaired in miR-155-deficient mice, and this defect was intrinsic to CD8(+) T cells, as miR-155-deficient CD8(+) T cells mounted greatly diminished primary and memory responses. Conversely, miR-155 overexpression augmented antiviral CD8(+) T cell responses in vivo. Gene-expression profiling showed that miR-155-deficient CD8(+) T cells had enhanced type I interferon signaling and were more susceptible to interferon's antiproliferative effect. Inhibition of the type I interferon-associated transcription factors STAT1 or IRF7 resulted in enhanced responses of miR-155-deficient CD8(+) T cells in vivo. We have thus identified a previously unknown role for miR-155 in regulating responsiveness to interferon and CD8(+) T cell responses to pathogens in vivo.
Collapse
|
695
|
MicroRNAs as pharmacological targets in endothelial cell function and dysfunction. Pharmacol Res 2013; 75:15-27. [PMID: 23603154 DOI: 10.1016/j.phrs.2013.04.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/03/2013] [Accepted: 04/05/2013] [Indexed: 12/11/2022]
Abstract
Endothelial cell dysfunction is a term which implies the dysregulation of normal endothelial cell functions, including impairment of the barrier functions, control of vascular tone, disturbance of proliferative, migratory and morphogenic capacities of endothelial cells, as well as control of leukocyte trafficking. MicroRNAs are short non-coding RNAs that have emerged as critical regulators of gene expression acting predominantly at the post-transcriptional level. This review summarizes the latest insights in the identification of endothelial-specific microRNAs and their targets, as well as their roles in controlling endothelial cell functions in both autocrine and paracrine manner. In addition, we discuss the therapeutic potential for the treatment of endothelial cell dysfunction and associated vascular pathophysiological conditions.
Collapse
|
696
|
Dudda JC, Salaun B, Ji Y, Palmer DC, Monnot GC, Merck E, Boudousquie C, Utzschneider DT, Escobar TM, Perret R, Muljo SA, Hebeisen M, Rufer N, Zehn D, Donda A, Restifo NP, Held W, Gattinoni L, Romero P. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity 2013; 38:742-53. [PMID: 23601686 PMCID: PMC3788592 DOI: 10.1016/j.immuni.2012.12.006] [Citation(s) in RCA: 252] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 12/18/2012] [Indexed: 01/08/2023]
Abstract
MicroRNAs (miRNAs) regulate the function of several immune cells, but their role in promoting CD8(+) T cell immunity remains unknown. Here we report that miRNA-155 is required for CD8(+) T cell responses to both virus and cancer. In the absence of miRNA-155, accumulation of effector CD8(+) T cells was severely reduced during acute and chronic viral infections and control of virus replication was impaired. Similarly, Mir155(-/-) CD8(+) T cells were ineffective at controlling tumor growth, whereas miRNA-155 overexpression enhanced the antitumor response. miRNA-155 deficiency resulted in accumulation of suppressor of cytokine signaling-1 (SOCS-1) causing defective cytokine signaling through STAT5. Consistently, enforced expression of SOCS-1 in CD8(+) T cells phenocopied the miRNA-155 deficiency, whereas SOCS-1 silencing augmented tumor destruction. These findings identify miRNA-155 and its target SOCS-1 as key regulators of effector CD8(+) T cells that can be modulated to potentiate immunotherapies for infectious diseases and cancer.
Collapse
Affiliation(s)
- Jan C Dudda
- Ludwig Center for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
697
|
Chen KD, Goto S, Hsu LW, Lin TY, Nakano T, Lai CY, Chang YC, Weng WT, Kuo YR, Wang CC, Cheng YF, Ma YY, Lin CC, Chen CL. Identification of miR-27b as a novel signature from the mRNA profiles of adipose-derived mesenchymal stem cells involved in the tolerogenic response. PLoS One 2013; 8:e60492. [PMID: 23613728 PMCID: PMC3628792 DOI: 10.1371/journal.pone.0060492] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 02/26/2013] [Indexed: 12/31/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (adipose-derived MSCs, ASCs) possess the ability to differentiate into multiple tissue types and have immune-modulatory properties similar to those of MSCs from other origins. However, the regulation of the MSC-elicited immune-modulatory activity by specific microRNA (miRNA) mechanisms remains unexplored. Gene expression profiling with knowledge-based functional enrichment analysis is an appropriate approach for unraveling these mechanisms. This tool can be used to examine the transcripts and miRNA regulators that differentiate the rat tolerogenic orthotopic liver transplantation (OLT; DA liver into PVG) and rejection OLT (DA liver into LEW) models. In both models, the rejection reaction was observed on postoperative day 7∼14 (rejection phase) but was overcome only by the PVG recipients. Thus, the global gene expression patterns of ASCs from spontaneously tolerant (PVG) and acute rejecting (LEW) rats in response to LPS activation were compared. In this study, we performed miRNA enrichment analysis based on the analysis of pathway, gene ontology (GO) terms and transcription factor binding site (TFBS) motif annotations. We found that the top candidate, miR-27, was specifically enriched and had the highest predicted frequency. We also identified a greater than 3-fold increase of miR-27b expression in the ASCs of tolerant recipients (DA to PVG) compared to those of rejecting recipients (DA to LEW) during the rejection phase in the rat OLT model. Furthermore, our data showed that miR-27b knockdown has a positive influence on the allosuppressive activity that inhibits T-cell proliferation. We found that miR-27 knockdown significantly induced the expression of CXCL12 in cultured ASCs and the expression of CXCL12 was responsible for the miR-27b antagomir-mediated inhibition of T-cell proliferation. These results, which through a series of comprehensive miRNA enrichment analyses, might be relevant for stem cell-based therapeutic applications in immunosuppressive function using ASCs.
Collapse
Affiliation(s)
- Kuang-Den Chen
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shigeru Goto
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Iwao Hospital, Yufuin, Japan
| | - Li-Wen Hsu
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Yang Lin
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Toshiaki Nakano
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Yun Lai
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Chen Chang
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wei-Teng Weng
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yur-Ren Kuo
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Chi Wang
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Fan Cheng
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Ying Ma
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Che Lin
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chao-Long Chen
- Center for Translational Research in Biomedical Sciences, Liver Transplantation Program and Departments of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
698
|
Matejuk A, Collet G, Nadim M, Grillon C, Kieda C. MicroRNAs and tumor vasculature normalization: impact on anti-tumor immune response. Arch Immunol Ther Exp (Warsz) 2013; 61:285-99. [PMID: 23575964 DOI: 10.1007/s00005-013-0231-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 01/15/2013] [Indexed: 12/21/2022]
Abstract
Inefficient immune response is a major glitch during tumor growth and progression. Chaotic and leaky blood vessels created in the process of angiogenesis allow tumor cells to escape and extricate anti-cancer immunity. Proangiogenic characteristics of hypoxic tumor microenvironment maintained by low oxygen tension attract endothelial progenitor cells, drive expansion of cancer stem cells, and deviantly differentiate monocyte descendants. Such cellular milieu further boosts immune tolerance and eventually appoint immunity for cancer advantage. Blood vessel normalization strategies that equilibrate oxygen levels within tumor and fix abnormal vasculature bring exciting promises to future anticancer therapies especially when combined with conventional chemotherapy. Recently, a new group of microRNAs (miRs) engaged in angiogenesis, called angiomiRs and hypoxamiRs, emerged as new therapeutic targets in cancer. Some of those miRs were found to efficiently regulate cancer immunity and their dysregulation efficiently programs aberrant angiogenesis and cancer metastasis. The present review highlights new findings in the field of miRs proficiency to normalize aberrant angiogenesis and to restore anti-tumor immune responses.
Collapse
Affiliation(s)
- Agata Matejuk
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, 45071 Orléans, France.
| | | | | | | | | |
Collapse
|
699
|
Abstract
miRNAs have been shown to play essential regulatory roles in the innate immune system. They function at multiple levels to shape the innate immune response and maintain homeostasis by direct suppression of the expression of their target proteins, preferentially crucial signaling components and transcription factors. Studies in humans and in disease models have revealed that dysregulation of several miRNAs such as miR-146a and miR-155 in rheumatic diseases leads to aberrant production of and/or signaling by inflammatory cytokines and, thus, critically contributes to disease pathogenesis. In addition, the recent description of the role of certain extracellular miRNAs as innate immune agonist to induce inflammatory response would have direct relevance to rheumatic diseases.
Collapse
|
700
|
Chen L, Li C, Peng Z, Zhao J, Gong G, Tan D. miR-197 Expression in Peripheral Blood Mononuclear Cells from Hepatitis B Virus-Infected Patients. Gut Liver 2013; 7:335-42. [PMID: 23710316 PMCID: PMC3661967 DOI: 10.5009/gnl.2013.7.3.335] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/15/2012] [Indexed: 01/08/2023] Open
Abstract
Background/Aims This study aimed to investigate the microRNA (miRNA) expression profiles in peripheral blood mononuclear cell (PBMC) of hepatitis B virus (HBV)-infected patients with different clinical manifestations and to analyze the function of miR-197. Methods PBMC miRNA expression profiles in 51 healthy controls, 70 chronic asymptomatic carriers,
107 chronic hepatitis B patients, and 76 HBV-related acute on chronic liver failure patients were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). miR-197 mimic and inhibitor were transfected in THP-1 cells. qRT-PCR and ELISA for interleukin (IL)-18 mRNA and protein levels were performed, respectively. Results The microarray analysis revealed that 17 PBMC miRNA expression profiles (12 miRNAs downregulated and five miRNAs upregulated) differed significantly in HBV-induced liver disease patients presenting with various symptoms. The qRT-PCR results suggested that the PBMC miR-197 levels regularly decreased as the severity of liver disease symptoms became aggravated. IL-18, a key regulator in inflammation and immunity, was inversely correlated with miR-197 levels. Bioinformatic analysis indicated that IL-18 was a target of miR-197. Exogenous expression of miR-197 could significantly repress IL-18 expression at both the mRNA and protein levels in THP-1 cells. Conclusions We concluded that multiple PBMC miRNAs had differential expression profiles during HBV infection and that miR-197 may play an important role in the reactivation of liver inflammation by targeting IL-18.
Collapse
Affiliation(s)
- Li Chen
- Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | | | |
Collapse
|