251
|
Hagiwara S, McClelland A, Kantharidis P. MicroRNA in diabetic nephropathy: renin angiotensin, aGE/RAGE, and oxidative stress pathway. J Diabetes Res 2013; 2013:173783. [PMID: 24575418 PMCID: PMC3875101 DOI: 10.1155/2013/173783] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/14/2013] [Indexed: 01/13/2023] Open
Abstract
MicroRNAs (miRNA) are a novel class of small, noncoding RNA molecules that have gained the attention of many researchers in recent years due to their ability to posttranscriptionally regulate the expression of families of genes simultaneously. Their role in normal physiology and pathobiology is intriguing and their regulation in normal and disease states is fascinating. That the cells can return to a state of homeostasis when these small molecules are perturbed is truly remarkable given the multiple cellular targets of each miRNA and that many mRNAs are targeted by multiple miRNAs. Several reviews have covered aspects of miRNA function in biology and disease. Here, we review the role of miRNA in regulating the renin-angiotensin system, AGE/RAGE signalling, and under conditions of oxidative stress in the context of diabetic nephropathy.
Collapse
Affiliation(s)
- Shinji Hagiwara
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Aaron McClelland
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Phillip Kantharidis
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- *Phillip Kantharidis:
| |
Collapse
|
252
|
Eken SM, Jin H, Chernogubova E, Maegdefessel L. Making sense in antisense: therapeutic potential of noncoding RNAs in diabetes-induced vascular dysfunction. J Diabetes Res 2013; 2013:834727. [PMID: 24369540 PMCID: PMC3863503 DOI: 10.1155/2013/834727] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/26/2013] [Indexed: 12/13/2022] Open
Abstract
The rapid rise of type II diabetes mellitus and its accompanying vascular complications call for novel approaches in unravelling its pathophysiological mechanisms and designing new treatment modalities. Noncoding RNAs represent a class of previously unknown molecular modulators of this disease. The most important features of diabetes-induced vascular disease, which include metabolic deregulation, increased oxidative stress, release of inflammatory mediators like adipokines, and pathologic changes in vascular cells, all are depicted and governed by a certain set of noncoding RNAs. While these mechanisms are being unravelled, new diagnostic and therapeutic opportunities to treat diabetes-induced vascular disease emerge.
Collapse
Affiliation(s)
- Suzanne M. Eken
- Atherosclerosis Research Unit, Department of Medicine, Center for Molecular Medicine (CMM L8), Karolinska Institute, 17176 Stockholm, Sweden
| | - Hong Jin
- Atherosclerosis Research Unit, Department of Medicine, Center for Molecular Medicine (CMM L8), Karolinska Institute, 17176 Stockholm, Sweden
| | - Ekaterina Chernogubova
- Atherosclerosis Research Unit, Department of Medicine, Center for Molecular Medicine (CMM L8), Karolinska Institute, 17176 Stockholm, Sweden
| | - Lars Maegdefessel
- Atherosclerosis Research Unit, Department of Medicine, Center for Molecular Medicine (CMM L8), Karolinska Institute, 17176 Stockholm, Sweden
- *Lars Maegdefessel:
| |
Collapse
|
253
|
Chen KC, Juo SHH. MicroRNAs in atherosclerosis. Kaohsiung J Med Sci 2012; 28:631-40. [DOI: 10.1016/j.kjms.2012.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 02/20/2012] [Indexed: 02/04/2023] Open
|
254
|
Lin YM, Hsu CJ, Liao YY, Chou MC, Tang CH. The CCL2/CCR2 axis enhances vascular cell adhesion molecule-1 expression in human synovial fibroblasts. PLoS One 2012. [PMID: 23185512 PMCID: PMC3503714 DOI: 10.1371/journal.pone.0049999] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1 (MCP-1), belongs to the CC chemokine family that is associated with the disease status and outcomes of osteoarthritis (OA). Here, we investigated the intracellular signaling pathways involved in CCL2-induced vascular cell adhesion molecule-1 (VCAM-1) expression in human OA synovial fibroblasts (OASFs). Methodology/Principal Findings Stimulation of OASFs with CCL2 induced VCAM-1 expression. CCL2-mediated VCAM-1 expression was attenuated by CCR2 inhibitor (RS102895), PKCδ inhibitor (rottlerin), p38MAPK inhibitor (SB203580), and AP-1 inhibitors (curcumin and tanshinone IIA). Stimulation of cells with CCL2 increased PKCδ and p38MAPK activation. Treatment of OASFs with CCL2 also increased the c-Jun phosphorylation and c-Jun binding to the AP-1 element on the VCAM-1 promoter. Moreover, CCL2-mediated CCR2, PKCδ, p38MAPK, and AP-1 pathway promoted the adhesion of monocytes to the OASFs monolayer. Conclusions/Significance Our results suggest that CCL2 increases VCAM-1 expression in human OASFs via the CCR2, PKCδ, p38MAPK, c-Jun, and AP-1 signaling pathway. The CCL2-induced VCAM-1 expression promoted monocytes adhesion to human OASFs.
Collapse
Affiliation(s)
- Yu-Min Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chin-Jung Hsu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopaedics, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Ya Liao
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- * E-mail: (CHT); (MCC)
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- * E-mail: (CHT); (MCC)
| |
Collapse
|
255
|
Song JT, Hu B, Qu HY, Bi CL, Huang XZ, Zhang M. Mechanical stretch modulates microRNA 21 expression, participating in proliferation and apoptosis in cultured human aortic smooth muscle cells. PLoS One 2012; 7:e47657. [PMID: 23082189 PMCID: PMC3474731 DOI: 10.1371/journal.pone.0047657] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/14/2012] [Indexed: 12/31/2022] Open
Abstract
Objectives Stretch affects vascular smooth muscle cell proliferation and apoptosis, and several responsible genes have been proposed. We tested whether the expression of microRNA 21 (miR-21) is modulated by stretch and is involved in stretch-induced proliferation and apoptosis of human aortic smooth muscle cells (HASMCs). Methods and Results RT-PCR revealed that elevated stretch (16% elongation, 1 Hz) increased miR-21 expression in cultured HASMCs, and moderate stretch (10% elongation, 1 Hz) decreased the expression. BrdU incorporation assay and cell counting showed miR-21 involved in the proliferation of HASMCs mediated by stretch, likely by regulating the expression of p27 and phosphorylated retinoblastoma protein (p-Rb). FACS analysis revealed that the complex of miR-21 and programmed cell death protein 4 (PDCD4) participated in regulating apoptosis with stretch. Stretch increased the expression of primary miR-21 and pre-miR-21 in HASMCs. Electrophoretic mobility shift assay (EMSA) demonstrated that stretch increased NF-κB and AP-1 activities in HASMCs, and blockade of AP-1 activity by c-jun siRNA significantly suppressed stretch-induced miR-21 expression. Conclusions Cyclic stretch modulates miR-21 expression in cultured HASMCs, and miR-21 plays important roles in regulating proliferation and apoptosis mediated by stretch. Stretch upregulates miR-21 expression at least in part at the transcription level and AP-1 is essential for stretch-induced miR-21 expression.
Collapse
Affiliation(s)
- Jian tao Song
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
256
|
Olivieri F, Spazzafumo L, Santini G, Lazzarini R, Albertini MC, Rippo MR, Galeazzi R, Abbatecola AM, Marcheselli F, Monti D, Ostan R, Cevenini E, Antonicelli R, Franceschi C, Procopio AD. Age-related differences in the expression of circulating microRNAs: miR-21 as a new circulating marker of inflammaging. Mech Ageing Dev 2012; 133:675-85. [PMID: 23041385 DOI: 10.1016/j.mad.2012.09.004] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Revised: 09/13/2012] [Accepted: 09/24/2012] [Indexed: 12/20/2022]
Abstract
Circulating microRNAs (miRs) have been investigated as diagnostic/prognostic biomarkers in human diseases. However, little is known about their expression throughout the aging process. Eleven healthy individuals aged 20, 80 and 100 years underwent miR plasma profiling. The validation cohort consisted of 111 healthy adults (CTR) aged 20-105 years and included 30 centenarians. In addition, 34 patients with cardiovascular disease (CVD) and 15 healthy centenarian offspring (CO) were enrolled. An exploratory factorial analysis grouped the miRs into three main factors: factor 1 primarily higher in 20-year-old subjects, but these differences did not reach statistical significance, factor 2 primarily higher in octogenarians and factor 3 primarily higher in centenarians. MiR-21, the most highly expressed miR of factors 2 and 3, was further validated, confirming the differences in the age groups. MiR-21 expression was higher in the CVD patients and lower in the CO compared to the age-matched CTR. MiR-21 was correlated with C-reactive protein and fibrinogen levels. TGF-β signaling was the predicted common pathway targeted by miRs of factors 2 and 3. TGF-βR2 mRNA, a validated miR-21 target, showed the highest expression in the leukocytes from a subset of the octogenarians. Our findings suggest that miR-21 may be a new biomarker of inflammation.
Collapse
Affiliation(s)
- Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
257
|
Peyrou M, Ramadori P, Bourgoin L, Foti M. PPARs in Liver Diseases and Cancer: Epigenetic Regulation by MicroRNAs. PPAR Res 2012; 2012:757803. [PMID: 23024649 PMCID: PMC3449131 DOI: 10.1155/2012/757803] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 06/27/2012] [Indexed: 12/19/2022] Open
Abstract
Peroxisome-proliferator-activated receptors (PPARs) are ligand-activated nuclear receptors that exert in the liver a transcriptional activity regulating a whole spectrum of physiological functions, including cholesterol and bile acid homeostasis, lipid/glucose metabolism, inflammatory responses, regenerative mechanisms, and cell differentiation/proliferation. Dysregulations of the expression, or activity, of specific PPAR isoforms in the liver are therefore believed to represent critical mechanisms contributing to the development of hepatic metabolic diseases, disorders induced by hepatic viral infections, and hepatocellular adenoma and carcinoma. In this regard, specific PPAR agonists have proven to be useful to treat these metabolic diseases, but for cancer therapies, the use of PPAR agonists is still debated. Interestingly, in addition to previously described mechanisms regulating PPARs expression and activity, microRNAs are emerging as new important regulators of PPAR expression and activity in pathophysiological conditions and therefore may represent future therapeutic targets to treat hepatic metabolic disorders and cancers. Here, we reviewed the current knowledge about the general roles of the different PPAR isoforms in common chronic metabolic and infectious liver diseases, as well as in the development of hepatic cancers. Recent works highlighting the regulation of PPARs by microRNAs in both physiological and pathological situations with a focus on the liver are also discussed.
Collapse
Affiliation(s)
- Marion Peyrou
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universiatire (CMU), 1206 Geneva, Switzerland
| | - Pierluigi Ramadori
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universiatire (CMU), 1206 Geneva, Switzerland
| | - Lucie Bourgoin
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universiatire (CMU), 1206 Geneva, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universiatire (CMU), 1206 Geneva, Switzerland
| |
Collapse
|
258
|
Fucci A, Colangelo T, Votino C, Pancione M, Sabatino L, Colantuoni V. The role of peroxisome proliferator-activated receptors in the esophageal, gastric, and colorectal cancer. PPAR Res 2012; 2012:242498. [PMID: 22991505 PMCID: PMC3444044 DOI: 10.1155/2012/242498] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 07/24/2012] [Indexed: 12/21/2022] Open
Abstract
Tumors of the gastrointestinal tract are among the most frequent human malignancies and account for approximately 30% of cancer-related deaths worldwide. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that control diverse cellular functions such as proliferation, differentiation, and cell death. Owing to their involvement in so many processes, they play crucial roles also in the development and physiology of the gastrointestinal tract. Consistently, PPARs deregulation has been implicated in several pathophysiological conditions, including chronic inflammation and cancer development. This paper summarizes the current knowledge on the role that the various PPAR isoforms play in the pathogenesis of the esophageal, gastric, and intestinal cancer. Elucidation of the molecular mechanisms underlying PPARs' signaling pathways will provide insights into their possible use as predictive biomarkers in the initial stages of the process. In addition, this understanding will provide the basis for new molecular targets in cancer therapy and chemoprevention.
Collapse
Affiliation(s)
- Alessandra Fucci
- Department of Biological, Geological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| | - Tommaso Colangelo
- Department of Biological, Geological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| | - Carolina Votino
- Department of Biological, Geological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| | - Massimo Pancione
- Department of Biological, Geological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| | - Lina Sabatino
- Department of Biological, Geological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| | - Vittorio Colantuoni
- Department of Biological, Geological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| |
Collapse
|
259
|
Wu C, Gong Y, Yuan J, Zhang W, Zhao G, Li H, Sun A, Zou Y, Ge J. microRNA-181a represses ox-LDL-stimulated inflammatory response in dendritic cell by targeting c-Fos. J Lipid Res 2012; 53:2355-63. [PMID: 22956783 PMCID: PMC3466004 DOI: 10.1194/jlr.m028878] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Oxidized LDL (ox-LDL) activates dendritic cells (DCs), thereby initiating
inflammation responses in atherosclerosis, yet the modulatory mechanisms remain
unclear. MicroRNAs (miRNAs) are important regulators for DC functions. This study
evaluated the regulation by miRNAs of the ox-LDL-induced DC immune response. In
CD11c+ DCs from ApoE-deficient mice with hyperlipidemia, microRNA
miR-181a was significantly up-regulated. In cultured bone marrow-derived DCs (BMDCs),
ox-LDL promoted DC maturation and up-regulated miR-181a expression. Abundance of
miR-181a attenuated ox-LDL-induced CD83 and CD40 expression, inhibited the secretion
of interleukin (IL)-6 and TNF-α, and up-regulated IL-10, an important
anti-inflammatory cytokine that was inhibited by ox-LDL. Inhibition of the endogenous
miR-181a reversed the effects on CD83 and CD40 as well as the effects on IL-6 and
TNF-α. The putative target genes of miR-181a were evaluated by gene ontology
assessment, and the c-Fos-mediated inflammation pathway was
identified. miR-181a targeted the 3′ untranslated region of
c-Fos mRNA by luciferase experiments. Thus, abundance of miR-181a
reduced c-Fos protein, whereas inhibition of miR-181a increased
c-Fos protein in BMDCs. We therefore suggest that miR-181a
attenuates ox-LDL-stimulated immune inflammation responses by targeting
c-Fos in DCs.
Collapse
Affiliation(s)
- Chaoneng Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
260
|
Shyu KG, Wang BW, Wu GJ, Lin CM, Chang H. Mechanical stretch via transforming growth factor-β1 activates microRNA208a to regulate endoglin expression in cultured rat cardiac myoblasts. Eur J Heart Fail 2012; 15:36-45. [PMID: 22941949 DOI: 10.1093/eurjhf/hfs143] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AIMS MicroRNAs (miRNAs) play a role in cardiac remodelling. MiR208a is essential for the expression of the genes involved in cardiac hypertrophy and fibrosis. The mechanism of regulation of miR208a involved in cardiac hypertrophy by mechanical stress is still unclear. We sought to investigate the mechanism of regulation of miR208a and the target gene of miR208a in cardiac cells by mechanical stretch. METHODS AND RESULTS Rat H9c2 cells (cardiac myoblasts) grown on a flexible membrane base were stretched via vacuum to 20% of maximum elongation at 60 cycles/min. Mechanical stretch significantly enhanced miR208a expression after 4 h of stretch. Exogenous addition of transforming growth factor-β1 (TGF-β1) increased miR208a expression, and pre-treatment with TGF-β1 antibody attenuated the miR208a expression induced by stretch. Mechanical stretch significantly increased endoglin and collagen I expression for 6-24 h. Exogenous addition of TGF-β1 and overexpression of miR208a up-regulated endoglin and collagen I expression, while antagomir208a and Smad3/4 inhibitor attenuated endoglin and collagen I expression induced by stretch. Mechanical stretch and TGF-β1 increased Smad3/4-DNA binding activity and miR208a promoter activity, and TGF-β1 antibody and Smad3/4 inhibitor decreased the Smad3/4-DNA binding activity and miR208a promoter activity induced by stretch. CONCLUSION Cyclic mechanical stretch enhances miR208a expression in cultured rat cardiac myoblasts. The stretch-induced miR208a is mediated by TGF-β1. Mir208a activates endoglin expression and may result in cardiac fibrosis.
Collapse
Affiliation(s)
- Kou-Gi Shyu
- Division of Cardiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
261
|
Chen LJ, Lim SH, Yeh YT, Lien SC, Chiu JJ. Roles of microRNAs in atherosclerosis and restenosis. J Biomed Sci 2012; 19:79. [PMID: 22931291 PMCID: PMC3438039 DOI: 10.1186/1423-0127-19-79] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/03/2012] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is commonly appreciated to represent a chronic inflammatory response of the vascular wall, and its complications cause high mortality in patients. Angioplasty with stent replacement is commonly performed in patients with atherosclerotic disease. However, the restenosis usually has a high incidence rate in angioplasty patients. Although the pathophysiological mechanisms underlying atherosclerosis and restenosis have been well established, new signaling molecules that control the progress of these pathologies have continuously been discovered. MicroRNAs (miRs) have recently emerged as a novel class of gene regulators that work via transcriptional degradation and translational inhibition or activation. Over 30% of genes in the cell can be directly regulated by miRs. Thus, miRs are recognized as crucial regulators in normal development, physiology and pathogenesis. Alterations of miR expression profiles have been revealed in diverse vascular diseases. A variety of functions of vascular cells, such as cell differentiation, contraction, migration, proliferation and inflammation that are involved in angiogenesis, neointimal formation and lipid metabolism underlying various vascular diseases, have been found to be regulated by miRs. This review summarizes current research progress and knowledge on the roles of miRs in regulating vascular cell function in atherosclerosis and restenosis. These discoveries are expected to present opportunities for clinical diagnostic and therapeutic approaches in vascular diseases resulting from atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Li-Jing Chen
- Division of Medical Engineering Research, National Health Research Institutes, Miaoli 350, Taiwan
| | | | | | | | | |
Collapse
|
262
|
|
263
|
Chau BN, Xin C, Hartner J, Ren S, Castano AP, Linn G, Li J, Tran PT, Kaimal V, Huang X, Chang AN, Li S, Kalra A, Grafals M, Portilla D, MacKenna DA, Orkin SH, Duffield JS. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci Transl Med 2012; 4:121ra18. [PMID: 22344686 DOI: 10.1126/scitranslmed.3003205] [Citation(s) in RCA: 427] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Scarring of the kidney is a major public health concern, directly promoting loss of kidney function. To understand the role of microRNA (miRNA) in the progression of kidney scarring in response to injury, we investigated changes in miRNA expression in two kidney fibrosis models and identified 24 commonly up-regulated miRNAs. Among them, miR-21 was highly elevated in both animal models and in human transplanted kidneys with nephropathy. Deletion of miR-21 in mice resulted in no overt abnormality. However, miR-21(-/-) mice suffered far less interstitial fibrosis in response to kidney injury, a phenotype duplicated in wild-type mice treated with anti-miR-21 oligonucleotides. Global derepression of miR-21 target mRNAs was readily detectable in miR-21(-/-) kidneys after injury. Analysis of gene expression profiles up-regulated in the absence of miR-21 identified groups of genes involved in metabolic pathways, including the lipid metabolism pathway regulated by peroxisome proliferator-activated receptor-α (Pparα), a direct miR-21 target. Overexpression of Pparα prevented ureteral obstruction-induced injury and fibrosis. Pparα deficiency abrogated the antifibrotic effect of anti-miR-21 oligonucleotides. miR-21 also regulated the redox metabolic pathway. The mitochondrial inhibitor of reactive oxygen species generation Mpv17l was repressed by miR-21, correlating closely with enhanced oxidative kidney damage. These studies demonstrate that miR-21 contributes to fibrogenesis and epithelial injury in the kidney in two mouse models and is a candidate target for antifibrotic therapies.
Collapse
|
264
|
Zhou X, Yuan P, He Y. Role of microRNAs in peripheral artery disease (review). Mol Med Rep 2012; 6:695-700. [PMID: 22767222 DOI: 10.3892/mmr.2012.978] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 07/02/2012] [Indexed: 11/06/2022] Open
Abstract
Peripheral arterial disease (PAD) involves a general vascular problem of diffuse atherosclerosis. The key pathological process is characterized by the aberrant proliferation of vascular smooth muscle cells and the formation of neointimal lesions. The molecular mechanisms involved in the regulation of the occurrence and development of PAD remain unclear. microRNAs (miRNAs) are highly conserved 20-25 nt-long non-coding RNAs that negatively regulate gene expression. Recent evidence has demonstrated that specific miRNAs are involved in the pathological processes of PAD, and these miRNAs are found to be critical modulators of vascular cell functions, including cell differentiation, contraction, migration, proliferation and apoptosis. This review summarizes findings of studies regarding the roles of specific miRNAs in PAD.
Collapse
Affiliation(s)
- Xiangyu Zhou
- Department of Vascular Surgery, Affiliated Hospital of Luzhou Medical College, Luzhou, Sichun, PR China.
| | | | | |
Collapse
|
265
|
Wahli W, Michalik L. PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol Metab 2012; 23:351-63. [PMID: 22704720 DOI: 10.1016/j.tem.2012.05.001] [Citation(s) in RCA: 514] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 04/27/2012] [Accepted: 05/01/2012] [Indexed: 12/12/2022]
Abstract
Nuclear receptors (NRs) are ligand-dependent transcription factors whose activation affects genes controlling vital processes. Among them, the peroxisome proliferator-activated receptors (PPARs) have emerged as links between lipids, metabolic diseases, and innate immunity. PPARs are activated by fatty acids and their derivatives, many of which also signal through membrane receptors, thereby creating a lipid signaling network between the cell surface and the nucleus. Tissues that play a role in whole-body metabolic homeostasis, such as adipose tissue, liver, skeletal muscle, intestines, and blood vessel walls, are prone to inflammation when metabolism is disturbed, a complication that promotes type 2 diabetes and cardiovascular disease. This review discusses the protective roles of PPARs in inflammatory conditions and the therapeutic anti-inflammatory potential of PPAR ligands.
Collapse
Affiliation(s)
- Walter Wahli
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland.
| | | |
Collapse
|
266
|
Mechanotransduction in embryonic vascular development. Biomech Model Mechanobiol 2012; 11:1149-68. [PMID: 22744845 DOI: 10.1007/s10237-012-0412-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/09/2012] [Indexed: 12/25/2022]
Abstract
A plethora of biochemical signals provides spatial and temporal cues that carefully orchestrate the complex process of vertebrate embryonic development. The embryonic vasculature develops not only in the context of these biochemical cues, but also in the context of the biomechanical forces imparted by blood flow. In the mature vasculature, different blood flow regimes induce distinct genetic programs, and significant progress has been made toward understanding how these forces are perceived by endothelial cells and transduced into biochemical signals. However, it cannot be assumed that paradigms that govern the mature vasculature are pertinent to the developing embryonic vasculature. The embryonic vasculature can respond to the mechanical forces of blood flow, and these responses are critical in vascular remodeling, certain aspects of sprouting angiogenesis, and maintenance of arterial-venous identity. Here, we review data regarding mechanistic aspects of endothelial cell mechanotransduction, with a focus on the response to shear stress, and elaborate upon the multifarious effects of shear stress on the embryonic vasculature. In addition, we discuss emerging predictive vascular growth models and highlight the prospect of combining signaling pathway information with computational modeling. We assert that correlation of precise measurements of hemodynamic parameters with effects on endothelial cell gene expression and cell behavior is required for fully understanding how blood flow-induced loading governs normal vascular development and shapes congenital cardiovascular abnormalities.
Collapse
|
267
|
|
268
|
Huang Y, Crawford M, Higuita-Castro N, Nana-Sinkam P, Ghadiali SN. miR-146a regulates mechanotransduction and pressure-induced inflammation in small airway epithelium. FASEB J 2012; 26:3351-64. [PMID: 22593544 DOI: 10.1096/fj.11-199240] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mechanical ventilation generates biophysical forces, including high transmural pressures, which exacerbate lung inflammation. This study sought to determine whether microRNAs (miRNAs) respond to this mechanical force and play a role in regulating mechanically induced inflammation. Primary human small airway epithelial cells (HSAEpCs) were exposed to 12 h of oscillatory pressure and/or the proinflammatory cytokine TNF-α. Experiments were also conducted after manipulating miRNA expression and silencing the transcription factor NF-κB or toll-like receptor proteins IRAK1 and TRAF6. NF-κB activation, IL-6/IL-8/IL-1β cytokine secretion, miRNA expression, and IRAK1/TRAF6 protein levels were monitored. A total of 12 h of oscillatory pressure and TNF-α resulted in a 5- to 7-fold increase in IL-6/IL-8 cytokine secretion, and oscillatory pressure also resulted in a time-dependent increase in IL-6/IL-8/IL-1β cytokine secretion. Pressure and TNF-α also resulted in distinct patterns of miRNA expression, with miR-146a being the most deregulated miRNA. Manipulating miR-146a expression altered pressure-induced cytokine secretion. Silencing of IRAK1 or TRAF6, confirmed targets of miR-146a, resulted in a 3-fold decrease in pressure-induced cytokine secretion. Cotransfection experiments demonstrate that miR-146a's regulation of pressure-induced cytokine secretion depends on its targeting of both IRAK1 and TRAF6. MiR-146a is a mechanosensitive miRNA that is rapidly up-regulated by oscillatory pressure and plays an important role in regulating mechanically induced inflammation in lung epithelia.
Collapse
Affiliation(s)
- Yan Huang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
269
|
Abstract
Recent findings demonstrated the importance of microRNAs (miRNAs) in the vasculature and the orchestration of lipid metabolism and glucose homeostasis. MiRNA networks represent an additional layer of regulation for gene expression that absorbs perturbations and ensures the robustness of biological systems. This function is very elegantly demonstrated in cholesterol metabolism where miRNAs reducing cellular cholesterol export are embedded in the very same genes that increase cholesterol synthesis. Often their alteration does not affect normal development but changes under stress conditions and in disease. A detailed understanding of the molecular and cellular mechanisms of miRNA-mediated effects on metabolism and vascular pathophysiology could pave the way for the development of novel diagnostic markers and therapeutic approaches. In the first part of this review, we summarize the role of miRNAs in vascular and metabolic diseases and explore potential confounding effects by platelet miRNAs in preclinical models of cardiovascular disease. In the second part, we discuss experimental strategies for miRNA target identification and the challenges in attributing miRNA effects to specific cell types and single targets.
Collapse
Affiliation(s)
- Anna Zampetaki
- King's British Heart Foundation Centre, King’s College London, United Kingdom
| | | |
Collapse
|
270
|
Schaefer T, Zajonz A, Lorentz P, Bohnacker T, Wymann MP, Schweighoffer T. Luminal decoration of blood vessels by activated perivasal mast cells in allergic rhinitis. Allergy 2012; 67:510-20. [PMID: 22313335 DOI: 10.1111/j.1398-9995.2012.02790.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND In allergic diseases, like in rhinitis, antigen challenge induces rapid degranulation of tissue resident mast cells and subsequent recruitment of leukocytes in response to soluble immunmodulators. The fate of mast cell-derived, membrane associated factors in inflamed tissue remained however unresolved. METHODS Components of the mast cell granular membrane, including the unique marker CD63var, were examined by FACS and by confocal laser scanning microscopy in cell culture and in diseased human tissue. RESULTS We discovered that selected mast cell membrane components appeared on the surface of distinct bystander cells. Acceptor cells did not acquire these molecules simply by uptake of soluble material or in the form of exosomes. Instead, physically stable cell-to-cell contact was required for transfer, in which a Notch2-Jagged1 interaction played a decisive role. This process is activation-dependent, unidirectional, and involves a unique membrane topology. Endothelial cells were particularly efficient acceptors. In organotypic 3D in vitro cultures we found that transferred mast cell molecules traversed an endothelial monolayer, and reappeared focally compacted on its distal surface, away from the actual contact zone. Moreover, we observed that such mast cell-derived membrane patches decorate microcapillaries in the nasal mucosa of allergic rhinitis patients. CONCLUSION Direct membrane transfer from perivasal mast cells into nearby blood vessels constitutes a novel mechanism to modulate endothelial surface features with apparent significance in allergic diseases.
Collapse
Affiliation(s)
- T. Schaefer
- Novartis Institutes for Biomedical Research (NIBR); Basel; Switzerland
| | - A. Zajonz
- Novartis Institutes for Biomedical Research (NIBR); Basel; Switzerland
| | - P. Lorentz
- Institute of Biochemistry and Genetics; Department of Biomedicine; University of Basel; Basel; Switzerland
| | - T. Bohnacker
- Institute of Biochemistry and Genetics; Department of Biomedicine; University of Basel; Basel; Switzerland
| | - M. P. Wymann
- Institute of Biochemistry and Genetics; Department of Biomedicine; University of Basel; Basel; Switzerland
| | - T. Schweighoffer
- Novartis Institutes for Biomedical Research (NIBR); Basel; Switzerland
| |
Collapse
|
271
|
Abstract
There are considerable interindividual variations in drug absorption, distribution, metabolism and excretion (ADME) in humans, which may lead to undesired drug effects in pharmacotherapy. Some of the mechanistic causes are known, e.g., genetic polymorphism, inhibition and induction of ADME enzymes and transporters, while others such as posttranscriptional regulation of ADME genes are under active study. MicroRNAs (miRNAs) are a large group of small, noncoding RNAs that control posttranscriptional expression of target genes. More than 1000 miRNAs have been identified in the human genome, which may regulate thousands of protein-coding genes. Some miRNAs directly or indirectly control the expression of xenobiotic-metabolizing cytochrome P450 enzymes, ATP-binding cassette or solute carrier transporters and/or nuclear receptors. Consequently, intervention of miRNA epigenetic signaling may alter ADME gene expression, change the capacity of drug metabolism and transport, and influence the sensitivity of cells to xenobiotics. In addition, the expression of some ADME regulatory miRNAs is significantly changed in cells following the exposure to a given drug, and the consequent changes in ADME gene expression might result in distinct ADME properties and drug response. In this review, we summarized recent findings on the role of noncoding miRNAs in epigenetic regulation of ADME genes and discussed the potential impact on pharmacokinetics and pharmacodynamics.
Collapse
Affiliation(s)
- Ai-Ming Yu
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260-1200, USA
| | - Yu-Zhuo Pan
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260-1200, USA
| |
Collapse
|
272
|
Koturbash I, Beland FA, Pogribny IP. Role of microRNAs in the regulation of drug metabolizing and transporting genes and the response to environmental toxicants. Expert Opin Drug Metab Toxicol 2012; 8:597-606. [PMID: 22435483 DOI: 10.1517/17425255.2012.673587] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION MicroRNAs (miRNAs) comprise a family of short non-coding RNAs that negatively regulate gene expression at the post-transcriptional level. Rapidly growing evidence indicates that miRNAs play a key role in drug (xenobiotic) metabolism. AREAS COVERED This review summarizes the current knowledge of the role of miRNAs in the regulation of drug (xenobiotic) metabolizing genes and the cellular responses to exposure to exogenous toxicants. The literature search was performed using the PubMed database (up to March 2012). EXPERT OPINION miRNAs play a major role in control of the proper functioning of the cellular drug metabolizing system. Emerging evidence indicates that exposure to environmental and occupational toxicants and therapeutic drugs may alter the expression of miRNAs, including those that regulate the expression of drug metabolizing genes. This suggests that drug-induced (xenobiotic-induced) miRNA abnormalities may be one of the underlying mechanisms in the pathogenesis of exposure-related pathologies, and that miRNAs may be potential non-invasive biomarkers of exposure and indicators of the severity of tissue injury induced by toxicants. Also, the evaluation of the expression of miRNAs may be applied for the chemical and drug safety assessment. Additionally, differences in the expression of miRNAs that target drug metabolizing genes may be important determinants for inter-individual differences in sensitivity to toxicants and can serve as critical biomarkers for identifying subpopulations sensitive to exposure.
Collapse
Affiliation(s)
- Igor Koturbash
- National Center for Toxicological Research, Division of Biochemical Toxicology, Jefferson, AR 72079, USA.
| | | | | |
Collapse
|
273
|
Fu H, Yang G, Wei M, Liu L, Jin L, Lu X, Wang L, Shen L, Zhang J, Lu H, Yao L, Lu Z. The RNA-binding protein QKI5 is a direct target of C/EBPα and delays macrophage differentiation. Mol Biol Cell 2012; 23:1628-35. [PMID: 22398723 PMCID: PMC3338430 DOI: 10.1091/mbc.e11-05-0412] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
During monocyte–macrophage differentiation, C/EBPα transcriptionally activates QKI, which in turn represses CSF1R and thus provides negative feedback to C/EBPα-induced macrophage differentiation. This feedback loop should be important in keeping the balance between cell proliferation and differentiation. Differentiated macrophages are essential for the innate immune system; however, the molecular mechanisms underlying the generation of macrophages remain largely unknown. Here we show that the RNA-binding protein QKI, mainly QKI-5, is transcriptionally activated in the early differentiated monocytic progenitors when CCAAT/enhancer-binding protein (C/EBP) α is expressed. The forced expression of C/EBPα increases the endogenous expression of QKI. Chromatin immunoprecipitation analysis and reporter assays further confirm that C/EBPα activates the transcription of QKI, primarily by binding to the distal C/EBPα-binding site. Blocking the induction of QKI using RNA interference enhances the expression of endogenous CSF1R and facilitates macrophage differentiation. Further study of the mechanism reveals that QKI-5 facilitates the degradation of CSF1R mRNA by interacting with the distal QRE in the 3′ untranslated region. In summary, we show that in committed macrophage progenitors, C/EBPα-activated QKI-5 negatively regulates macrophage differentiation by down-regulating CSF1R expression, forming a negative feedback loop during macrophage differentiation.
Collapse
Affiliation(s)
- Haiyan Fu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Mgilitary Medical University, 710032 Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Nazari-Jahantigh M, Wei Y, Schober A. The role of microRNAs in arterial remodelling. Thromb Haemost 2012; 107:611-8. [PMID: 22371089 DOI: 10.1160/th11-12-0826] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 02/13/2012] [Indexed: 01/13/2023]
Abstract
Adaptive alterations of the vessel wall architecture, called vascular remodelling, can be found in arterial hypertension, during the formation of aneurysms, in restenosis after vascular interventions, and in atherosclerosis. MicroRNAs (miR) critically affect the main cellular players in arterial remodelling and may either promote or inhibit the structural changes in the vessel wall. They regulate the phenotype of smooth muscle cells (SMCs) and control the inflammatory response in endothelial cells and macrophages. In SMCs, different sets of miRs induce either a synthetic or contractile phenotype, respectively. The conversion into a synthetic SMC phenotype is a crucial event in arterial remodelling. Therefore, reprogramming of the SMC phenotype by miR targeting can modulate the remodelling process. Furthermore, the effects of stimuli that induce remodelling, such as shear stress, angiotensin II, oxidised low-density lipoprotein, or apoptosis, on endothelial cells are mediated by miRs. The endothelial cell-specific miR-126, for example, is transferred in microvesicles from apoptotic endothelial cells and plays a protective role in atherogenesis. The inflammatory response of the innate immune system, especially through macrophages, promotes arterial remodelling. miR-155 induces the expression of inflammatory cytokines, whereas miR-146a and miR-147 are involved in the resolution phase of inflammation. However, in vivo data on the role of miRs in vascular remodelling are still scarce, which are required to test the therapeutic potential of the available, highly effective miR inhibitors.
Collapse
Affiliation(s)
- M Nazari-Jahantigh
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | | | | |
Collapse
|
275
|
|
276
|
Duan Q, Wang X, Gong W, Ni L, Chen C, He X, Chen F, Yang L, Wang P, Wang DW. ER stress negatively modulates the expression of the miR-199a/214 cluster to regulates tumor survival and progression in human hepatocellular cancer. PLoS One 2012; 7:e31518. [PMID: 22359598 PMCID: PMC3281082 DOI: 10.1371/journal.pone.0031518] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 01/09/2012] [Indexed: 12/21/2022] Open
Abstract
Background Recent studies have emphasized causative links between microRNAs (miRNAs) deregulation and tumor development. In hepatocellular carcinoma (HCC), more and more miRNAs were identified as diagnostic and prognostic cancer biomarkers, as well as additional therapeutic tools. This study aimed to investigate the functional significance and regulatory mechanism of the miR-199a2/214 cluster in HCC progression. Methods and Findings In this study, we showed that miR-214, as well as miR-199a-3p and miR-199a-5p levels were significantly reduced in the majority of examined 23 HCC tissues and HepG2 and SMMC-7721 cell lines, compared with their nontumor counterparts. To further explore the role of miR-214 in hepatocarcinogenesis, we disclosed that the ER stress-induced pro-survival factor XBP-1 is a target of miR-214 by using western blot assay and luciferase reporter assay. Re-expression of miR-214 in HCC cell lines (HepG2 and SMMC-7721) inhibited proliferation and induced apoptosis. Furthermore, ectopic expression of miR-214 dramatically suppressed the ability of HCC cells to form colonies in vitro and to develop tumors in a subcutaneous xenotransplantation model of the BALB/c athymic nude mice. Moreover, reintroduction of XBP-1s attenuated miR-214-mediated suppression of HCC cells proliferation, colony and tumor formation. To further understand the mechanism of the miR-199a/214 cluster down-expression in HCC, we found that thapsigargin (TG) and tunicamycin (TM) or hypoxia-induced unfolded protein response (UPR) suppresses the expression of the miR-199a/214 cluster in HCC cells. By promoter analysis of the miR-199a2/214 gene, we conjectured NFκB as a potential negative regulator. We further found that UPR and LPS-induced NFκB activation suppressed miR-199a2/214 transcription, and this suppression was reversed by NFκB inhibition in HCC cells. Conclusions Our study suggest that modulation of miR-214 levels may provide a new therapeutic approach for cancer treatment and revealed that UPR may offer a new explanation for why the miR-199a/214 cluster were down-regulated in the progression in HCC.
Collapse
Affiliation(s)
- Quanlu Duan
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xingxu Wang
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wei Gong
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li Ni
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chen Chen
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- * E-mail: (DW); (CC)
| | - Xingxing He
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fuqiong Chen
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lei Yang
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Peihua Wang
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Dao Wen Wang
- Department of Internal Medicine and Gene Therapy Center, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- * E-mail: (DW); (CC)
| |
Collapse
|
277
|
Fang Y, Davies PF. Site-specific microRNA-92a regulation of Kruppel-like factors 4 and 2 in atherosusceptible endothelium. Arterioscler Thromb Vasc Biol 2012; 32:979-87. [PMID: 22267480 DOI: 10.1161/atvbaha.111.244053] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Endothelial transcription factors Krüppel-like factor 4 (KLF4) and KLF2 are implicated in protection against atherogenesis. Steady-state microRNA (miR) regulation of KLFs in vivo is accessible by screening region-specific endothelial miRs and their targets. METHODS AND RESULTS A subset of differentially expressed endothelial miRs was identified in atherosusceptible versus protected regions of normal swine aorta. In silico analyses predicted highly conserved binding sites in the 3'-untranslated region (3'UTR) of KLF4 for 5 miRs of the subset (miR-26a, -26b, -29a, -92a, and -103) and a single binding site for a miR-92a complex in the 3'UTR of KLF2. Of these, only miR-92a knockdown and knock-in resulted in responses of KLF4 and KLF2 expression in human arterial endothelial cells. Dual luciferase reporter assays demonstrated functional interactions of miR-92a with full-length 3'UTR sequences of both KLFs and with the specific binding elements therein. Two evolutionarily conserved miR-92a sites in KLF4 3'UTR and 1 site in KLF2 3'UTR were functionally validated. Knockdown of miR-92a in vitro resulted in partial rescue from cytokine-induced proinflammatory marker expression (monocyte chemotactic protein 1, vascular cell adhesion molecule-1, E-selectin, and endothelial nitric oxide synthase) that was attributable to enhanced KLF4 expression. Leukocyte-human arterial endothelial cell adhesion experiments supported this conclusion. In swine aortic arch endothelium, a site of atherosusceptibility where miR-92a expression was elevated, both KLFs were expressed at low levels relative to protected thoracic aorta. CONCLUSIONS miR-92a coregulates KLF4 and KLF2 expression in arterial endothelium and contributes to phenotype heterogeneity associated with regional atherosusceptibility and protection in vivo.
Collapse
Affiliation(s)
- Yun Fang
- Institute for Medicine and Engineering, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | | |
Collapse
|
278
|
Diehl P, Fricke A, Sander L, Stamm J, Bassler N, Htun N, Ziemann M, Helbing T, El-Osta A, Jowett JBM, Peter K. Microparticles: major transport vehicles for distinct microRNAs in circulation. Cardiovasc Res 2012; 93:633-44. [PMID: 22258631 PMCID: PMC3291092 DOI: 10.1093/cvr/cvs007] [Citation(s) in RCA: 394] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aims Circulating microRNAs (miRNAs) have attracted major interest as biomarkers for cardiovascular diseases. Since RNases are abundant in circulating blood, there needs to be a mechanism protecting miRNAs from degradation. We hypothesized that microparticles (MP) represent protective transport vehicles for miRNAs and that these are specifically packaged by their maternal cells. Methods and results Conventional plasma preparations, such as the ones used for biomarker detection, are shown to contain substantial numbers of platelet-, leucocyte-, and endothelial cell-derived MP. To analyse the widest spectrum of miRNAs, Next Generation Sequencing was used to assess miRNA profiles of MP and their corresponding stimulated and non-stimulated cells of origin. THP-1 (monocytic origin) and human umbilical vein endothelial cell (HUVEC) MP were used for representing circulating MP at a high purity. miRNA profiles of MP differed significantly from those of stimulated and non-stimulated maternal THP-1 cells and HUVECs, respectively. Quantitative reverse transcription–polymerase chain reaction of miRNAs which have been associated with cardiovascular diseases also demonstrated significant differences in miRNA profiles between platelets and their MP. Notably, the main fraction of miRNA in plasma was localized in MP. Furthermore, miRNA profiles of MP differed significantly between patients with stable and unstable coronary artery disease. Conclusion Circulating MP represent transport vehicles for large numbers of specific miRNAs, which have been associated with cardiovascular diseases. miRNA profiles of MP are significantly different from their maternal cells, indicating an active mechanism of selective ‘packaging’ from cells into MP. These findings describe an interesting mechanism for transferring gene-regulatory function from MP-releasing cells to target cells via MP circulating in blood.
Collapse
Affiliation(s)
- Philipp Diehl
- Atherothrombosis and Vascular Biology, BakerIDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Schroen B, Heymans S. Small but smart--microRNAs in the centre of inflammatory processes during cardiovascular diseases, the metabolic syndrome, and ageing. Cardiovasc Res 2011; 93:605-13. [PMID: 21994347 DOI: 10.1093/cvr/cvr268] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
With a progressively growing elderly population, ageing-associated pathologies such as cardiovascular diseases are becoming an increasing economic, social, and personal burden for Western societies. Interestingly, all ageing-associated diseases share a common denominator: inflammation. Recently, microRNAs were shown to be implicated in the full range of processes of ageing, inflammation, and cardiovascular diseases. This review focuses on their role in cardiovascular diseases with emphasis on their implication in the inflammatory processes that accompany heart failure, atherosclerosis, coronary artery disease, and finally obesity and diabetes as components of the ageing-associated metabolic syndrome.
Collapse
Affiliation(s)
- Blanche Schroen
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | | |
Collapse
|
280
|
Zhou J, Lim SH, Chiu JJ. Epigenetic Regulation of Vascular Endothelial Biology/Pathobiology and Response to Fluid Shear Stress. Cell Mol Bioeng 2011. [DOI: 10.1007/s12195-011-0199-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
281
|
|