51
|
Schurgers LJ, Akbulut AC, Kaczor DM, Halder M, Koenen RR, Kramann R. Initiation and Propagation of Vascular Calcification Is Regulated by a Concert of Platelet- and Smooth Muscle Cell-Derived Extracellular Vesicles. Front Cardiovasc Med 2018; 5:36. [PMID: 29682509 PMCID: PMC5897433 DOI: 10.3389/fcvm.2018.00036] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
The ageing population continues to suffer from its primary killer, cardiovascular disease (CVD). Despite recent advances in interventional medicinal and surgical therapies towards the end of the 20th century, the epidemic of cardiovascular disease has not been halted. Yet, rather than receding globally, the burden of CVD has risen to become a top cause of morbidity and mortality worldwide. Most CVD arises from thrombotic rupture of an atherosclerotic plaque, the pathologic thickening of coronary and carotid artery segments and subsequent distal ischemia in heart or brain. In fact, one-fifth of deaths are directly attributable to thrombotic rupture of a vulnerable plaque. Atherosclerotic lesion formation is caused by a concert of interactions between circulating leukocytes and platelets, interacting with the endothelial barrier, signalling into the arterial wall by the release of cytokines and extracellular vesicles (EVs). Both platelet- and cell-derived EVs represent a novel mechanism of cellular communication, particularly by the transport and transfer of cargo and by reprogramming of the recipient cell. These interactions result in phenotypic switching of vascular smooth muscle cells (VSMCs) causing migration and proliferation, and subsequent secretion of EVs. Loss of VSMCs attracts perivascular Mesenchymal Stem Cells (MSCs) from the adventitia, which are a source of VSMCs and contribute to repair after vascular injury. However, continuous stress stimuli eventually switch phenotype of cells into osteochondrogenic VSMCs facilitating vascular calcification. Although Virchow’s triad is over 100 years old, it is a reality that is accurate today. It can be briefly summarised as changes in the composition of blood (platelet EVs), alterations in the vessel wall (VSMC phenotypic switching, MSC infiltration and EV release) and disruption of blood flow (atherothrombosis). In this paper, we review the latest relevant advances in the identification of extracellular vesicle pathways as well as VSMCs and pericyte/MSC phenotypic switching, underlying vascular calcification.
Collapse
Affiliation(s)
- Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Asim C Akbulut
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Dawid M Kaczor
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Maurice Halder
- Division of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Rafael Kramann
- Division of Nephrology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
52
|
Sun WL, Wang N, Xu Y. Impact of miR-302b on Calcium-phosphorus Metabolism and Vascular Calcification of Rats with Chronic Renal Failure by Regulating BMP-2/Runx2/Osterix Signaling Pathway. Arch Med Res 2018; 49:164-171. [DOI: 10.1016/j.arcmed.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 07/30/2018] [Indexed: 12/22/2022]
|
53
|
Ghosh N, Katare R. Molecular mechanism of diabetic cardiomyopathy and modulation of microRNA function by synthetic oligonucleotides. Cardiovasc Diabetol 2018; 17:43. [PMID: 29566757 PMCID: PMC5863891 DOI: 10.1186/s12933-018-0684-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/10/2018] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a chronic complication in individuals with diabetes and is characterized by ventricular dilation and hypertrophy, diastolic dysfunction, decreased or preserved systolic function and reduced ejection fraction eventually resulting in heart failure. Despite being well characterized, the fundamental mechanisms leading to DCM are still elusive. Recent studies identified the involvement of small non-coding small RNA molecules such as microRNAs (miRs) playing a key role in the etiology of DCM. Therefore, miRs associated with DCM represents a new class of targets for the development of mechanistic therapeutics, which may yield marked benefits compared to other therapeutic approaches. Indeed, few miRs currently under active clinical investigation, with many expressing cautious optimism that miRs based therapies will succeed in the coming years. The major caution in using miRs based therapy is the need to improve the stability and specificity following systemic injection, which can be achieved through chemical and structural modification. In this review, we first discuss the established role of miRs in DCM and the advances in miRs based therapeutic strategies for the prevention/treatment of DCM. We next discuss the currently employed chemical modification of miR oligonucleotides and their utility in therapies specifically focusing on the DCM. Finally, we summarize the commonly used delivery system and approaches for assessment of miRNA modulation and potential off-target effects.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Department of Physiology-HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
| |
Collapse
|
54
|
Buyukterzi Z, Can U, Alpaydin S, Guzelant A, Karaarslan S, Mustu M, Kocyigit D, Gurses KM. Enhanced serum levels of matrix Gla protein and bone morphogenetic protein in acute coronary syndrome patients. J Clin Lab Anal 2018; 32:e22278. [PMID: 28605143 PMCID: PMC6816878 DOI: 10.1002/jcla.22278] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/15/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Vascular calcification has been found to be associated with increased risk of cardiovascular (CV) morbidity and mortality. Various bone-associated proteins have been suggested to be related with this process. In this study, we aimed to evaluate whether serum levels of bone morphogenic protein-4 (BMP-4) and matrix Gla protein (MGP) differed in patients who were found to have normal epicardial coronary arteries or a culprit lesion in the coronary angiography leading to acute coronary syndrome (ACS). METHODS Patients admitted to emergency department with the diagnosis of ACS who underwent primary percutaneous coronary intervention (PCI) between October 2015 and April 2016 were consecutively recruited as the patient group. Age and gender-matched subjects who underwent coronary angiography following non-invasive ischemia assessment made the control group. RESULTS A total of 90 subjects (63.00±14.02 years, 70% male) were included in this study. MGP (<0.001) and BMP-4 (<0.001) levels were significantly elevated when compared to subjects with normal coronary arteries. Fasting blood glucose (P=.024), HDL-cholesterol (P=.002), C-reactive protein (CRP) (P=.001) levels, and left ventricular ejection fraction (LVEF) (P=.021) were significantly correlated with serum MGP levels. HDL-cholesterol (P=.001) and CRP (P=.030) levels were also significantly correlated with serum BMP-4 levels. In the model including HDL-cholesterol, CRP, MGP, and BMP-4 levels, only MGP (odds ratio[OR]: 1.018, P=.019) and BMP-4 (OR: 1.313, P=.023) were found to be independently associated with ACS. CONCLUSION This study shows that serum BMP-4 and MGP are independently associated with ACS occurrence when adjusted for other CV risk factors. These biomarkers may have a diagnostic potential in ACS patients.
Collapse
Affiliation(s)
- Zafer Buyukterzi
- Department of CardiologyKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| | - Ummugulsum Can
- Department of BiochemistryKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| | - Sertac Alpaydin
- Department of CardiologyKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| | - Asuman Guzelant
- Department of MicrobiologyKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| | - Sukru Karaarslan
- Department of CardiologyKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| | - Mehmet Mustu
- Department of CardiologyKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| | - Duygu Kocyigit
- Department of CardiologyHacettepe University Faculty of MedicineAnkaraTurkey
| | - Kadri Murat Gurses
- Department of CardiologyKonya Training and Research HospitalUniversity of Health SciencesMeramKonyaTurkey
| |
Collapse
|
55
|
Alkagiet S, Tziomalos K. Vascular calcification: the role of microRNAs. Biomol Concepts 2018; 8:119-123. [PMID: 28426428 DOI: 10.1515/bmc-2017-0001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/21/2017] [Indexed: 01/22/2023] Open
Abstract
Vascular calcification represents the deposition of calcium phosphate salts in the tunica media of the vascular wall. It occurs during aging but is accelerated and pronounced in patients with diabetes mellitus, chronic kidney disease (CKD) and established cardiovascular disease. Due to the loss of elasticity of the vessel wall, vascular calcification might result in left ventricular hypertrophy and compromise coronary perfusion. Accordingly, several studies showed that vascular calcification is associated with increased risk for cardiovascular morbidity and mortality. Accumulating data suggest that microRNAs (miRs) play an important role in vascular calcification. A variety of miRs have been implicated in the development of vascular calcification, whereas others appear to play a protective role. Accordingly, miRs might represent promising targets for the prevention of vascular calcification and its adverse cardiovascular sequelae. However, given the complexity of regulation of this process and the multitude of miRs involved, more research is needed to identify the optimal candidate miRs for targeting.
Collapse
|
56
|
Cardoso L, Weinbaum S. Microcalcifications, Their Genesis, Growth, and Biomechanical Stability in Fibrous Cap Rupture. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1097:129-155. [PMID: 30315543 DOI: 10.1007/978-3-319-96445-4_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
For many decades, cardiovascular calcification has been considered as a passive process, accompanying atheroma progression, correlated with plaque burden, and apparently without a major role on plaque vulnerability. Clinical and pathological analyses have previously focused on the total amount of calcification (calcified area in a whole atheroma cross section) and whether more calcification means higher risk of plaque rupture or not. However, this paradigm has been changing in the last decade or so. Recent research has focused on the presence of microcalcifications (μCalcs) in the atheroma and more importantly on whether clusters of μCalcs are located in the cap of the atheroma. While the vast majority of μCalcs are found in the lipid pool or necrotic core, they are inconsequential to vulnerable plaque. Nevertheless, it has been shown that μCalcs located within the fibrous cap could be numerous and that they behave as an intensifier of the background circumferential stress in the cap. It is now known that such intensifying effect depends on the size and shape of the μCalc as well as the proximity between two or more μCalcs. If μCalcs are located in caps with very low background stress, the increase in stress concentration may not be sufficient to reach the rupture threshold. However, the presence of μCalc(s) in the cap with a background stress of about one fifth to one half the rupture threshold (a stable plaque) will produce a significant increase in local stress, which may exceed the cap rupture threshold and thus transform a non-vulnerable plaque into a vulnerable one. Also, the classic view that treats cardiovascular calcification as a passive process has been challenged, and emerging data suggest that cardiovascular calcification may encompass both passive and active processes. The passive calcification process comprises biochemical factors, specifically circulating nucleating complexes, which would lead to calcification of the atheroma. The active mechanism of atherosclerotic calcification is a cell-mediated process via cell death of macrophages and smooth muscle cells (SMCs) and/or the release of matrix vesicles by SMCs.
Collapse
Affiliation(s)
- Luis Cardoso
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| |
Collapse
|
57
|
|
58
|
Nanoudis S, Pikilidou M, Yavropoulou M, Zebekakis P. The Role of MicroRNAs in Arterial Stiffness and Arterial Calcification. An Update and Review of the Literature. Front Genet 2017; 8:209. [PMID: 29312437 PMCID: PMC5733083 DOI: 10.3389/fgene.2017.00209] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022] Open
Abstract
Arterial stiffness is an independent risk factor for fatal and non-fatal cardiovascular events, such as systolic hypertension, coronary artery disease, stroke, and heart failure. Moreover it reflects arterial aging which in many cases does not coincide with chronological aging, a fact that is in large attributed to genetic factors. In addition to genetic factors, microRNAs (miRNAs) seem to largely affect arterial aging either by advancing or by regressing arterial stiffness. MiRNAs are small RNA molecules, ~22 nucleotides long that can negatively control their target gene expression posttranscriptionally. Pathways that affect main components of stiffness such as fibrosis and calcification seem to be influenced by up or downregulation of specific miRNAs. Identification of this aberrant production of miRNAs can help identify epigenetic changes that can be therapeutic targets for prevention and treatment of vascular diseases. The present review summarizes the specific role of the so far discovered miRNAs that are involved in pathways of arterial stiffness.
Collapse
Affiliation(s)
- Sideris Nanoudis
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Maria Pikilidou
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Maria Yavropoulou
- Division of Endocrinology and Metabolism, AHEPA University Hospital, Thessaloniki, Greece
| | - Pantelis Zebekakis
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
59
|
Gošev I, Zeljko M, Đurić Ž, Nikolić I, Gošev M, Ivčević S, Bešić D, Legčević Z, Paić F. Epigenome alterations in aortic valve stenosis and its related left ventricular hypertrophy. Clin Epigenetics 2017; 9:106. [PMID: 29026447 PMCID: PMC5627415 DOI: 10.1186/s13148-017-0406-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022] Open
Abstract
Aortic valve stenosis is the most common cardiac valve disease, and with current trends in the population demographics, its prevalence is likely to rise, thus posing a major health and economic burden facing the worldwide societies. Over the past decade, it has become more than clear that our traditional genetic views do not sufficiently explain the well-known link between AS, proatherogenic risk factors, flow-induced mechanical forces, and disease-prone environmental influences. Recent breakthroughs in the field of epigenetics offer us a new perspective on gene regulation, which has broadened our perspective on etiology of aortic stenosis and other aortic valve diseases. Since all known epigenetic marks are potentially reversible this perspective is especially exciting given the potential for development of successful and non-invasive therapeutic intervention and reprogramming of cells at the epigenetic level even in the early stages of disease progression. This review will examine the known relationships between four major epigenetic mechanisms: DNA methylation, posttranslational histone modification, ATP-dependent chromatin remodeling, and non-coding regulatory RNAs, and initiation and progression of AS. Numerous profiling and functional studies indicate that they could contribute to endothelial dysfunctions, disease-prone activation of monocyte-macrophage and circulatory osteoprogenitor cells and activation and osteogenic transdifferentiation of aortic valve interstitial cells, thus leading to valvular inflammation, fibrosis, and calcification, and to pressure overload-induced maladaptive myocardial remodeling and left ventricular hypertrophy. This is especcialy the case for small non-coding microRNAs but was also, although in a smaller scale, convincingly demonstrated for other members of cellular epigenome landscape. Equally important, and clinically most relevant, the reported data indicate that epigenetic marks, particularly certain microRNA signatures, could represent useful non-invasive biomarkers that reflect the disease progression and patients prognosis for recovery after the valve replacement surgery.
Collapse
Affiliation(s)
- Igor Gošev
- Department of Surgery, University of Rochester Medical center, Rochester, NY USA
| | - Martina Zeljko
- Department of Cardiology, Clinical Unit of Internal Medicine, Clinical Hospital Merkur, Zajćeva 19, 10 000 Zagreb, Croatia
| | - Željko Đurić
- Department of Cardiac Surgery, University Hospital Center Zagreb, Kišpatićeva 12, 10 000 Zagreb, Croatia
| | - Ivana Nikolić
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115 USA
| | - Milorad Gošev
- School of Medicine, University of Josip Juraj Strossmayer, Trg Svetog trojstva 3, 31 000 Osijek, Croatia
| | - Sanja Ivčević
- Department of Physiology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Dino Bešić
- Laboratory for Epigenetics and Molecular Medicine, Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Zoran Legčević
- Laboratory for Epigenetics and Molecular Medicine, Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Frane Paić
- Laboratory for Epigenetics and Molecular Medicine, Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| |
Collapse
|
60
|
Murabito JM, Rong J, Lunetta KL, Huan T, Lin H, Zhao Q, Freedman JE, Tanriverdi K, Levy D, Larson MG. Cross-sectional relations of whole-blood miRNA expression levels and hand grip strength in a community sample. Aging Cell 2017; 16:888-894. [PMID: 28597569 PMCID: PMC5506437 DOI: 10.1111/acel.12622] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2017] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression with emerging data suggesting miRNAs play a role in skeletal muscle biology. We sought to examine the association of miRNAs with grip strength in a community-based sample. Framingham Heart Study Offspring and Generation 3 participants (n = 5668 54% women, mean age 55 years, range 24, 90 years) underwent grip strength measurement and miRNA profiling using whole blood from fasting morning samples. Linear mixed-effects regression modeling of grip strength (kg) versus continuous miRNA 'Cq' values and versus binary miRNA expression was performed. We conducted an integrative miRNA-mRNA coexpression analysis and examined the enrichment of biologic pathways for the top miRNAs associated with grip strength. Grip strength was lower in women than in men and declined with age with a mean 44.7 (10.0) kg in men and 26.5 (6.3) kg in women. Among 299 miRNAs interrogated for association with grip strength, 93 (31%) had FDR q value < 0.05, 54 (18%) had an FDR q value < 0.01, and 15 (5%) had FDR q value < 0.001. For almost all miRNA-grip strength associations, increasing miRNA concentration is associated with increasing grip strength. miR-20a-5p (FDR q 1.8 × 10-6 ) had the most significant association and several among the top 15 miRNAs had links to skeletal muscle including miR-126-3p, miR-30a-5p, and miR-30d-5p. The top associated biologic pathways included metabolism, chemokine signaling, and ubiquitin-mediated proteolysis. Our comprehensive assessment in a community-based sample of miRNAs in blood associated with grip strength provides a framework to further our understanding of the biology of muscle strength.
Collapse
Affiliation(s)
- Joanne M. Murabito
- The Framingham Heart StudyFraminghamMAUSA
- Department of Medicine, Section of General Internal MedicineBoston University School of MedicineBostonMAUSA
| | - Jian Rong
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
| | - Kathryn L. Lunetta
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
| | - Tianxiao Huan
- The Framingham Heart StudyFraminghamMAUSA
- The Population Sciences BranchDivision of Intramural Research, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Honghuang Lin
- Section of Computational BiomedicineDepartment of MedicineBoston University School of MedicineBostonMAUSA
| | - Qiang Zhao
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
| | - Jane E. Freedman
- Cardiology DivisionDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Kahraman Tanriverdi
- Cardiology DivisionDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Daniel Levy
- The Framingham Heart StudyFraminghamMAUSA
- The Population Sciences BranchDivision of Intramural Research, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Martin G. Larson
- The Framingham Heart StudyFraminghamMAUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
| |
Collapse
|
61
|
Abstract
Despite rapid advances in cardiovascular research and therapeutic strategies, ischemic heart disease (IHD) remains the leading cause of mortality worldwide. MicroRNAs (miRNAs) are small, noncoding RNAs which post transcriptionally regulate gene expression. In the past few years, miRNAs have emerged as key tools for the understanding of the pathophysiology of IHD, with potential uses as new biomarkers and therapeutic targets. Several studies report a regulatory role of miRNAs, with regard to fundamental components of IHD pathogenesis and progression, such as lipoprotein metabolism, atherogenesis, vascular calcification, platelet function, and angiogenesis. Due to their high stability in biofluids, circulating miRNAs have attracted attention as promising biomarkers of IHD, especially in cardiovascular risk prediction and the diagnosis of myocardial infarction. Furthermore, experimental studies have demonstrated the potential of miRNA-targeted therapy in improving hyperlipidemia, atherosclerosis, and angiogenesis. In this review, the current knowledge on the role of miRNAs in IHD and translational perspectives of their use is discussed.
Collapse
|
62
|
Ouyang L, Zhang K, Chen J, Wang J, Huang H. Roles of platelet-derived growth factor in vascular calcification. J Cell Physiol 2017; 233:2804-2814. [PMID: 28467642 DOI: 10.1002/jcp.25985] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is prevalent in aging, and patients with hypertension, chronic kidney disease (CKD), or diabetes. VC is regarded as an active and complex process that involves multiple mechanisms responsible for calcium deposition in vessel wall. In light of the complicated pathogenesis of VC, effective therapy for ameliorating VC is limited. Thus, it is urgent to explore the potential mechanisms and find new targets for the therapy of VC. Platelet-derived growth factor (PDGF), a potent mitogen, and chemoattractant have been found to disturb the vascular homeostasis by inducing inflammation, oxidative stress, and phenotype transition, all of which accelerate the process of VC. The aim of current review is to present a review about the roles of PDGF in affecting VC and to establish a potential target for treating VC.
Collapse
Affiliation(s)
- Liu Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Kun Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Jie Chen
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingfeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Hui Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| |
Collapse
|
63
|
MicroRNA-32 promotes calcification in vascular smooth muscle cells: Implications as a novel marker for coronary artery calcification. PLoS One 2017; 12:e0174138. [PMID: 28319142 PMCID: PMC5358880 DOI: 10.1371/journal.pone.0174138] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/03/2017] [Indexed: 11/19/2022] Open
Abstract
Cardiovascular calcification is one of the most severe outcomes associated with cardiovascular disease and often results in significant morbidity and mortality. Previous reports indicated that epigenomic regulation of microRNAs (miRNAs) might play important roles in vascular smooth muscle cell (VSMC) calcification. Here, we identified potential key miRNAs involved in vascular calcification in vivo and investigated the role of miR-32-5p (miR-32). According to microarray analysis, we observed increased expression of miR-125b, miR-30a, and miR-32 and decreased expression of miR-29a, miR-210, and miR-320 during the progression of vascularcalcification. Additionally, gain- and loss-of-function studies of miR-32 confirmed promotion of VSMC calcification in mice through the enhanced expression of bonemorphogenetic protein-2, runt-related transcription factor-2(RUNX2), osteopontin, and the bone-specific phosphoprotein matrix GLA protein in vitro. Moreover, miR-32 modulated vascularcalcification progression by activating phosphoinositide 3-kinase (PI3K)signaling and increasing RUNX2 expression and phosphorylation by targeting the 3'-untranslated region of phosphatase and tensin homolog Mrna (PTEN) in mouse VSMCs. Furthermore, we detected higher miR-32 levels in plasmafrom patients with coronary artery disease with coronary artery calcification (CAC) as compared with levels observed in non-CAC patients (P = 0.016), further confirming miR-32 as a critical modulator and potential diagnostic marker for CAC.
Collapse
|
64
|
Wang S, He W, Wang C. MiR-23a Regulates the Vasculogenesis of Coronary Artery Disease by Targeting Epidermal Growth Factor Receptor. Cardiovasc Ther 2017; 34:199-208. [PMID: 27085964 DOI: 10.1111/1755-5922.12187] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Circulating microRNAs (miRNAs) in patient body fluids have recently been considered to hold the potential of being novel disease biomarkers and drug targets. We aimed to investigate the correlation between the levels of circulating miR-23a and the expression of epidermal growth factor receptor (EGFR) in the pathogenesis of patients with coronary heart disease to further explore the mechanism involved in its vasculogenesis. METHOD Three different cohorts, including 13 acute myocardial infarction (AMI) patients, 176 angina pectoris patients, and 127 control subjects, were enrolled to investigate the expression levels of circulating miR-23a in patients with myocardial ischemia and also the relationship between plasma miR-23a and severity of coronary stenosis. Plasma miR-23a levels of participants were examined by real-time quantitative PCR. Simultaneously, plasma cardiac troponin I (cTnI) concentrations were measured by ELISAs. We further detected the correlation of miR-23a and EGFR by molecular and animal assays. RESULT MiR-23a was enriched in not only diseased endothelial progenitor cells (EPCs) but also in the plasma of patients with coronary artery disease (CAD). Besides, we found out miR-23a was able to suppress EGFR expression and EPC activities. Reporter assays confirmed the direct binding and repression of miR-23a to the 3'-UTR of EGFR mRNA. Knockdown of miR-23a not only restored EGFR levels and angiogenic activities of diseased EPCs in vitro, but further promoted blood flow recovery in ischemic limbs of mice. CONCLUSION Circulating miR-23a may be a new biomarker for CAD and as a potential diagnostic tool. And increased miR-23a level may be used to predict the presence and severity of coronary lesions in patients with CAD.
Collapse
Affiliation(s)
- Shixi Wang
- Department of Cardiology, Central Hospital of Zaozhuang Mining Group, Zaozhuang, Shandong, China
| | - Weidong He
- Department of Nutritional, Affiliated Hospital, Qingdao University Medical College, Qingdao, Shandong, China
| | - Caijin Wang
- Department of Cardiology, Institute of Traditional Chinese Medicine, Liaocheng, Shandong, China
| |
Collapse
|
65
|
The Involvement of miR-29b-3p in Arterial Calcification by Targeting Matrix Metalloproteinase-2. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6713606. [PMID: 28164126 PMCID: PMC5253168 DOI: 10.1155/2017/6713606] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/05/2016] [Accepted: 11/15/2016] [Indexed: 02/06/2023]
Abstract
Vascular calcification is a risk predictor and common pathological change in cardiovascular diseases that are associated with elastin degradation and phenotypic transformation of vascular smooth muscle cells via gelatinase matrix metalloproteinase-2 (MMP2). However, the mechanisms involved in this process remain unclear. In this study, we investigated the relationships between miR-29b-3p and MMP2, to confirm miR-29b-3p-mediated MMP2 expression at the posttranscriptional level in arterial calcification. In male Sprague Dawley rats, arterial calcification was induced by subcutaneous injection of a toxic dose of cholecalciferol. In vivo, the quantitative real-time polymerase chain reaction (qRT-PCR) showed that MMP2 expression was upregulated in calcified arterial tissues, and miR-29b-3p expression was downregulated. There was a negative correlation between MMP2 mRNA expression and miR-29b-3p levels (P = 0.0014, R2 = 0.481). Western blotting showed that MMP2 expression was significantly increased in rats treated with cholecalciferol. In vitro, overexpression of miR-29b-3p led to decreased MMP2 expression in rat vascular smooth muscle cells, while downregulation of miR-29b-3p expression led to increased MMP2 expression. Moreover, the luciferase reporter assay confirmed that MMP2 is the direct target of miR-29b-3p. Together, our results demonstrated that a role of miR-29b-3p in vascular calcification involves targeting MMP2.
Collapse
|
66
|
Function, Role, and Clinical Application of MicroRNAs in Vascular Aging. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6021394. [PMID: 28097140 PMCID: PMC5209603 DOI: 10.1155/2016/6021394] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/07/2016] [Accepted: 11/23/2016] [Indexed: 01/31/2023]
Abstract
Vascular aging, a specific type of organic aging, is related to age-dependent changes in the vasculature, including atherosclerotic plaques, arterial stiffness, fibrosis, and increased intimal thickening. Vascular aging could influence the threshold, process, and severity of various cardiovascular diseases, thus making it one of the most important risk factors in the high mortality of cardiovascular diseases. As endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are the main cell biological basis of these pathology changes of the vasculature, the structure and function of ECs and VSMCs play a key role in vascular aging. MicroRNAs (miRNAs), small noncoding RNAs, have been shown to regulate the expression of multiple messenger RNAs (mRNAs) posttranscriptionally, contributing to many crucial aspects of cell biology. Recently, miRNAs with functions associated with aging or aging-related diseases have been studied. In this review, we will summarize the reported role of miRNAs in the process of vascular aging with special emphasis on EC and VSMC functions. In addition, the potential application of miRNAs to clinical practice for the diagnosis and treatment of cardiovascular diseases will also be discussed.
Collapse
|
67
|
Identification of suitable reference gene and biomarkers of serum miRNAs for osteoporosis. Sci Rep 2016; 6:36347. [PMID: 27821865 PMCID: PMC5099566 DOI: 10.1038/srep36347] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/14/2016] [Indexed: 12/30/2022] Open
Abstract
Our objective was to identify suitable reference genes in serum miRNA for normalization and screen potential new biomarkers for osteoporosis diagnosis by a systematic study. Two types of osteoporosis models were used like as mechanical unloading and estrogen deficiency. Through a large-scale screening using microarray, qPCR validation and statistical algorithms, we first identified miR-25-3p as a suitable reference gene for both type of osteoporosis, which also showed stability during the differentiation processes of osteoblast and osteoclast. Then 15 serum miRNAs with differential expression in OVX rats were identified by microarray and qPCR validation. We further detected these 15 miRNAs in postmenopausal women and bedrest rhesus monkeys and evaluated their diagnostic value by ROC analysis. Among these miRNAs, miR-30b-5p was significantly down-regulated in postmenopausal women with osteopenia or osteoporosis; miR-103-3p, miR-142-3p, miR-328-3p were only significantly decreased in osteoporosis. They all showed positive correlations with BMD. Except miR328-3p, the other three miRNAs were also declined in the rhesus monkeys after long-duration bedrest. Their AUC values (all >0.75) proved the diagnostic potential. Our results provided a reliable normalization reference gene and verified a group of circulating miRNAs as non-invasive biomarkers in the detection of postmenopausal- and mechanical unloading- osteoporosis.
Collapse
|
68
|
Ferraz EP, Oliveira FS, de Oliveira PT, Crovace MC, Peitl-Filho O, Beloti MM, Rosa AL. Bioactive glass-based surfaces induce differential gene expression profiling of osteoblasts. J Biomed Mater Res A 2016; 105:419-423. [PMID: 27682446 DOI: 10.1002/jbm.a.35915] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 01/08/2023]
Abstract
The ability of Biosilicate® with two crystalline phases (BioS-2P) to drive osteoblast differentiation encourages the investigation of the cellular mechanisms involved in this process. Then, the aim of our study was to analyze the large-scale gene expression of osteoblasts grown on BioS-2P compared with Bioglass® 45S5 (45S5). Osteoblasts differentiated from rat bone marrow mesenchymal stem cells were cultured under osteogenic conditions on BioS-2P, 45S5 and polystyrene (control). After 10 days, the expression of 23,794 genes was analyzed using mRNA Sequencing and the data were validated by real-time PCR. The BioS-2P exhibited 5 genes upregulated and 3 downregulated compared with 45S5. Compared with control, BioS-2P upregulated 15 and downregulated 11 genes, while 45S5 upregulated 25 and downregulated 21 genes. Eight genes were commonly upregulated and 4 downregulated by both bioactive glasses. In conclusion, our results demonstrated that bioactive glasses affect the gene expression profiling of osteoblasts. Most of the regulated genes by both BioS-2P and 45S5 are associated with the process of mineralization highlighting their osteostimulation property that is, at least in part, derived from the ability to modulate the intracellular machinery to promote osteoblast genotype expression. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 419-423, 2017.
Collapse
Affiliation(s)
- Emanuela P Ferraz
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Fabiola S Oliveira
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Paulo T de Oliveira
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Murilo C Crovace
- Vitreous Materials Laboratory (LaMaV), Federal University of São Carlos, Rod Washington Luis, Km 235, São Carlos, SP, 13565-905, Brazil
| | - Oscar Peitl-Filho
- Vitreous Materials Laboratory (LaMaV), Federal University of São Carlos, Rod Washington Luis, Km 235, São Carlos, SP, 13565-905, Brazil
| | - Marcio M Beloti
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Adalberto L Rosa
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| |
Collapse
|
69
|
Abstract
Transforming growth factor β (TGF-β) and related growth factors are secreted pleiotropic factors that play critical roles in embryogenesis and adult tissue homeostasis by regulating cell proliferation, differentiation, death, and migration. The TGF-β family members signal via heteromeric complexes of type I and type II receptors, which activate members of the Smad family of signal transducers. The main attribute of the TGF-β signaling pathway is context-dependence. Depending on the concentration and type of ligand, target tissue, and developmental stage, TGF-β family members transmit distinct signals. Deregulation of TGF-β signaling contributes to developmental defects and human diseases. More than a decade of studies have revealed the framework by which TGF-βs encode a context-dependent signal, which includes various positive and negative modifiers of the principal elements of the signaling pathway, the receptors, and the Smad proteins. In this review, we first introduce some basic components of the TGF-β signaling pathways and their actions, and then discuss posttranslational modifications and modulatory partners that modify the outcome of the signaling and contribute to its context-dependence, including small noncoding RNAs.
Collapse
Affiliation(s)
- Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94143
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
70
|
Zhao W, Zhang S, Wang B, Huang J, Lu WW, Chen D. Runx2 and microRNA regulation in bone and cartilage diseases. Ann N Y Acad Sci 2016; 1383:80-87. [PMID: 27526290 DOI: 10.1111/nyas.13206] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/13/2016] [Accepted: 07/19/2016] [Indexed: 12/14/2022]
Abstract
The homeostasis of skeletal tissues requires tight regulation of a variety of signaling pathways, and the onset and progression of skeletal diseases are often caused by signaling abnormalities. MicroRNAs (miRNAs) are short noncoding RNA molecules that have emerged as a new dimension of gene regulation. MiRNAs have been shown to play an important role in the regulation of the differentiation of embryonic and hematopoietic stem cells. However, the role of specific miRNAs and their target genes has not been fully defined in the regulation of mesenchymal stem cells. Runx2 is a key transcription factor controlling MSC differentiation and bone and cartilage function. This article reviews work on Runx2 and miRNA regulation in bone and cartilage diseases.
Collapse
Affiliation(s)
- Weiwei Zhao
- Department of Biochemistry, Rush University Medical Center, Chicago, Illionois.,Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Shanxing Zhang
- Department of Biochemistry, Rush University Medical Center, Chicago, Illionois
| | - Baoli Wang
- Key Lab of Hormone and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jian Huang
- Department of Biochemistry, Rush University Medical Center, Chicago, Illionois
| | - William W Lu
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, Illionois
| |
Collapse
|
71
|
Liu X, Li M, Peng Y, Hu X, Xu J, Zhu S, Yu Z, Han S. miR-30c regulates proliferation, apoptosis and differentiation via the Shh signaling pathway in P19 cells. Exp Mol Med 2016; 48:e248. [PMID: 27469029 PMCID: PMC4973315 DOI: 10.1038/emm.2016.57] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/28/2016] [Accepted: 03/02/2016] [Indexed: 01/03/2023] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding single-stranded RNAs that suppress protein expression by binding to the 3′ untranslated regions of their target genes. Many studies have shown that miRNAs have important roles in congenital heart diseases (CHDs) by regulating gene expression and signaling pathways. We previously found that miR-30c was highly expressed in the heart tissues of aborted embryos with ventricular septal defects. Therefore, this study aimed to explore the effects of miR-30c in CHDs. miR-30c was overexpressed or knocked down in P19 cells, a myocardial cell model that is widely used to study cardiogenesis. We found that miR-30c overexpression not only increased cell proliferation by promoting cell entry into S phase but also suppressed cell apoptosis. In addition, we found that miR-30c inhibited dimethyl sulfoxide-induced differentiation of P19 cells. miR-30c knockdown, in contrast, inhibited cell proliferation and increased apoptosis and differentiation. The Sonic hedgehog (Shh) signaling pathway is essential for normal embryonic development. Western blotting and luciferase assays revealed that Gli2, a transcriptional factor that has essential roles in the Shh signaling pathway, was a potential target gene of miR-30c. Ptch1, another important player in the Shh signaling pathway and a transcriptional target of Gli2, was downregulated by miR-30c overexpression and upregulated by miR-30c knockdown. Collectively, our study revealed that miR-30c suppressed P19 cell differentiation by inhibiting the Shh signaling pathway and altered the balance between cell proliferation and apoptosis, which may result in embryonic cardiac malfunctions.
Collapse
Affiliation(s)
- Xuehua Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Mengmeng Li
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Yuzhu Peng
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaoshan Hu
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Jing Xu
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Shasha Zhu
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Zhangbin Yu
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Shuping Han
- State Key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University, Nanjing, China
| |
Collapse
|
72
|
Louvet L, Metzinger L, Büchel J, Steppan S, Massy ZA. Magnesium Attenuates Phosphate-Induced Deregulation of a MicroRNA Signature and Prevents Modulation of Smad1 and Osterix during the Course of Vascular Calcification. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7419524. [PMID: 27419135 PMCID: PMC4933865 DOI: 10.1155/2016/7419524] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/13/2016] [Accepted: 05/22/2016] [Indexed: 12/20/2022]
Abstract
Vascular calcification (VC) is prevalent in patients suffering from chronic kidney disease (CKD). High phosphate levels promote VC by inducing abnormalities in mineral and bone metabolism. Previously, we demonstrated that magnesium (Mg(2+)) prevents inorganic phosphate- (Pi-) induced VC in human aortic vascular smooth muscle cells (HAVSMC). As microRNAs (miR) modulate gene expression, we investigated the role of miR-29b, -30b, -125b, -133a, -143, and -204 in the protective effect of Mg(2+) on VC. HAVSMC were cultured in the presence of 3 mM Pi with or without 2 mM Mg(2+) chloride. Total RNA was extracted after 4 h, 24 h, day 3, day 7, and day 10. miR-30b, -133a, and -143 were downregulated during the time course of Pi-induced VC, whereas the addition of Mg(2+) restored (miR-30b) or improved (miR-133a, miR-143) their expression. The expression of specific targets Smad1 and Osterix was significantly increased in the presence of Pi and restored by coincubation with Mg(2+). As miR-30b, miR-133a, and miR-143 are negatively regulated by Pi and restored by Mg(2+) with a congruent modulation of their known targets Runx2, Smad1, and Osterix, our results provide a potential mechanistic explanation of the observed upregulation of these master switches of osteogenesis during the course of VC.
Collapse
Affiliation(s)
- Loïc Louvet
- INSERM U 1088, CURS, University of Picardie Jules Verne, Amiens, France
| | - Laurent Metzinger
- INSERM U 1088, CURS, University of Picardie Jules Verne, Amiens, France
| | - Janine Büchel
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | - Sonja Steppan
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | - Ziad A. Massy
- Division of Nephrology, Ambroise Paré University Hospital, APHP, University of Paris Ouest-Versailles-St-Quentin-en-Yvelines (UVSQ), Boulogne-Billancourt, Paris, France
- INSERM U 1018, Research Centre in Epidemiology and Population Health (CESP) Team 5, Villejuif, France
| |
Collapse
|
73
|
Song CL, Liu B, Wang JP, Zhang BL, Zhang JC, Zhao LY, Shi YF, Li YX, Wang G, Diao HY, Li Q, Xue X, Wu JD, Liu J, Yu YP, Cai D, Liu ZX. Anti-apoptotic effect of microRNA-30b in early phase of rat myocardial ischemia-reperfusion injury model. J Cell Biochem 2016; 116:2610-9. [PMID: 25925903 DOI: 10.1002/jcb.25208] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/21/2015] [Indexed: 01/01/2023]
Abstract
This study aimed to investigate the effect of microRNA-30b (miR-30b) in rat myocardial ischemic-reperfusion (I/R) injury model. We randomly divided Sprague-Dawley (SD) rats (n = 80) into five groups: 1) control group; 2) miR-30b group; 3) sham-operated group; 4) I/R group, and 5) I/R+miR-30b group. Real-time quantitative polymerase chain reaction, immunohistochemical staining and Western blot analysis were conducted. TUNEL assay was employed for testing cardiomyocyte apoptosis. Our results showed that miR-30b levels were down-regulated in I/R group and I/R + miR-30b group compared with sham-operated group (both P < 0.05). However, miR-30b level in I/R + miR-30b group was higher than I/R group (P < 0.05). Markedly, the apoptotic rate in I/R group showed highest in I/R group (P < 0.05). Additionally, the results illustrated that protein levels of Bcl-2, Bax, and caspase-3 were at higher levels in ischemic regions in I/R group, comparing to sham-operated group (all P < 0.05), while Bcl-2/Bax was reduced (P < 0.05). Bcl-2 level and Bcl-2/Bax were obviously increased in I/R + miR-30b group by comparison with I/R group, and expression levels of Bax and caspase-3 were down-regulated (all P < 0.05). We also found that in I/R + miR-30b group, KRAS level was apparently lower and p-AKT level was higher by comparing with I/R group (both P < 0.05). Our study indicated that miR-30b overexpression had anti-apoptotic effect on early phase of rat myocardial ischemia injury model through targeting KRAS and activating the Ras/Akt pathway.
Collapse
Affiliation(s)
- Chun-Li Song
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Bin Liu
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Jin-Peng Wang
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Bei-Lin Zhang
- Department of Physiology, the College of Basic Medical Sciences of Jilin University, Changchun, 130021, P. R. China
| | - Ji-Chang Zhang
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Li-Yan Zhao
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yong-Feng Shi
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yang-Xue Li
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Guan Wang
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Hong-Ying Diao
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Qian Li
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Xin Xue
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Jun-Duo Wu
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Jia Liu
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yun-Peng Yu
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Dan Cai
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Zhi-Xian Liu
- Department of Cardiology, the Second Hospital of Jilin University, Changchun, 130041, P. R. China
| |
Collapse
|
74
|
Cui L, Houston DA, Farquharson C, MacRae VE. Characterisation of matrix vesicles in skeletal and soft tissue mineralisation. Bone 2016; 87:147-58. [PMID: 27072517 DOI: 10.1016/j.bone.2016.04.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 03/25/2016] [Accepted: 04/06/2016] [Indexed: 12/16/2022]
Abstract
The importance of matrix vesicles (MVs) has been repeatedly highlighted in the formation of cartilage, bone, and dentin since their discovery in 1967. These nano-vesicular structures, which are found in the extracellular matrix, are believed to be one of the sites of mineral nucleation that occurs in the organic matrix of the skeletal tissues. In the more recent years, there have been numerous reports on the observation of MV-like particles in calcified vascular tissues that could be playing a similar role. Therefore, here, we review the characteristics MVs possess that enable them to participate in mineral deposition. Additionally, we outline the content of skeletal tissue- and soft tissue-derived MVs, and discuss their key mineralisation mediators that could be targeted for future therapeutic use.
Collapse
Affiliation(s)
- L Cui
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh Easter Bush Campus, Edinburgh, Midlothian, EH25 9RG, UK.
| | - D A Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh Easter Bush Campus, Edinburgh, Midlothian, EH25 9RG, UK
| | - C Farquharson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh Easter Bush Campus, Edinburgh, Midlothian, EH25 9RG, UK
| | - V E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh Easter Bush Campus, Edinburgh, Midlothian, EH25 9RG, UK
| |
Collapse
|
75
|
Abstract
Vascular disease, such as atherosclerosis and diabetic vasculopathy, is frequently complicated by vascular calcification. Previously believed to be an end-stage process of unregulated mineral precipitation, it is now well established to be a multi-faceted disease influenced by the characteristics of its vascular location, the origins of calcifying cells and numerous regulatory pathways. It reflects the fundamental plasticity of the vasculature that is gradually being revealed by progress in vascular and stem cell biology. This review provides a brief overview of where we stand in our understanding of vascular calcification, facing the challenge of translating this knowledge into viable preventive and therapeutic strategies.
Collapse
|
76
|
Raut SK, Singh GB, Rastogi B, Saikia UN, Mittal A, Dogra N, Singh S, Prasad R, Khullar M. miR-30c and miR-181a synergistically modulate p53–p21 pathway in diabetes induced cardiac hypertrophy. Mol Cell Biochem 2016; 417:191-203. [DOI: 10.1007/s11010-016-2729-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/20/2016] [Indexed: 12/27/2022]
|
77
|
Wu M, Chen G, Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res 2016; 4:16009. [PMID: 27563484 PMCID: PMC4985055 DOI: 10.1038/boneres.2016.9] [Citation(s) in RCA: 1035] [Impact Index Per Article: 129.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) and bone morphogenic protein (BMP) signaling has fundamental roles in both embryonic skeletal development and postnatal bone homeostasis. TGF-βs and BMPs, acting on a tetrameric receptor complex, transduce signals to both the canonical Smad-dependent signaling pathway (that is, TGF-β/BMP ligands, receptors, and Smads) and the non-canonical-Smad-independent signaling pathway (that is, p38 mitogen-activated protein kinase/p38 MAPK) to regulate mesenchymal stem cell differentiation during skeletal development, bone formation and bone homeostasis. Both the Smad and p38 MAPK signaling pathways converge at transcription factors, for example, Runx2 to promote osteoblast differentiation and chondrocyte differentiation from mesenchymal precursor cells. TGF-β and BMP signaling is controlled by multiple factors, including the ubiquitin–proteasome system, epigenetic factors, and microRNA. Dysregulated TGF-β and BMP signaling result in a number of bone disorders in humans. Knockout or mutation of TGF-β and BMP signaling-related genes in mice leads to bone abnormalities of varying severity, which enable a better understanding of TGF-β/BMP signaling in bone and the signaling networks underlying osteoblast differentiation and bone formation. There is also crosstalk between TGF-β/BMP signaling and several critical cytokines’ signaling pathways (for example, Wnt, Hedgehog, Notch, PTHrP, and FGF) to coordinate osteogenesis, skeletal development, and bone homeostasis. This review summarizes the recent advances in our understanding of TGF-β/BMP signaling in osteoblast differentiation, chondrocyte differentiation, skeletal development, cartilage formation, bone formation, bone homeostasis, and related human bone diseases caused by the disruption of TGF-β/BMP signaling.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| | - Guiqian Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA; Department of neurology, Bruke Medical Research Institute, Weil Cornell Medicine of Cornell University, White Plains, USA
| | - Yi-Ping Li
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| |
Collapse
|
78
|
Coll-Bonfill N, de la Cruz-Thea B, Pisano MV, Musri MM. Noncoding RNAs in smooth muscle cell homeostasis: implications in phenotypic switch and vascular disorders. Pflugers Arch 2016; 468:1071-87. [PMID: 27109570 DOI: 10.1007/s00424-016-1821-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/04/2016] [Indexed: 12/16/2022]
Abstract
Vascular smooth muscle cells (SMC) are a highly specialized cell type that exhibit extraordinary plasticity in adult animals in response to a number of environmental cues. Upon vascular injury, SMC undergo phenotypic switch from a contractile-differentiated to a proliferative/migratory-dedifferentiated phenotype. This process plays a major role in vascular lesion formation and during the development of vascular remodeling. Vascular remodeling comprises the accumulation of dedifferentiated SMC in the intima of arteries and is central to a number of vascular diseases such as arteriosclerosis, chronic obstructive pulmonary disease or pulmonary hypertension. Therefore, it is critical to understand the molecular mechanisms that govern SMC phenotype. In the last decade, a number of new classes of noncoding RNAs have been described. These molecules have emerged as key factors controlling tissue homeostasis during physiological and pathological conditions. In this review, we will discuss the role of noncoding RNAs, including microRNAs and long noncoding RNAs, in the regulation of SMC plasticity.
Collapse
Affiliation(s)
- N Coll-Bonfill
- Department of Pulmonary Medicine Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - B de la Cruz-Thea
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - M V Pisano
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - M M Musri
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.
| |
Collapse
|
79
|
Szulc P. Abdominal aortic calcification: A reappraisal of epidemiological and pathophysiological data. Bone 2016; 84:25-37. [PMID: 26688274 DOI: 10.1016/j.bone.2015.12.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 12/16/2022]
Abstract
In men and women, there is a significant association between the risk of cardiovascular event (myocardial infarction, stroke) and risk of major fragility fracture (hip, vertebra). Abdominal aortic calcification (AAC) can be assessed using semiquantitative scores on spine radiographs and spine scans obtained by DXA. Severe AAC is associated with higher risk of major cardiovascular event. Not only does severe AAC reflect poor cardiovascular health status, but also directly disturbs blood flow in the vascular system. Severe (but not mild or moderate) AAC is associated with lower bone mineral density (BMD), faster bone loss and higher risk of major fragility fracture. The fracture risk remains increased after adjustment for BMD and other potential risk factors. The association between severe AAC and fracture risk was found in both sexes, mainly in the follow-ups of less than 10years. Many factors contribute to initiation and progression of AAC: lifestyle, co-morbidities, inorganic ions, dyslipidemia, hormones, cytokines (e.g. inflammatory cytokines, RANKL), matrix vesicles, microRNAs, structural proteins (e.g. elastin), vitamin K-dependent proteins, and medications (e.g. vitamin K antagonists). Osteogenic transdifferentiation of vascular smooth muscle cells (VSMC) and circulating osteoprogenitors penetrating into vascular wall plays a major role in the AAC initiation and progression. Vitamin K-dependent proteins protect vascular tunica media against formation of calcified deposits (matrix GLA protein, GLA-rich protein) and against VSMC apoptosis (Gas6). Further studies are needed to investigate clinical utility of AAC for the assessment of fracture and cardiovascular risk at the individual level and develop new medications permitting to prevent AAC progression.
Collapse
Affiliation(s)
- Pawel Szulc
- INSERM UMR 1033, University of Lyon, Hôpital Edouard Herriot, Lyon, France.
| |
Collapse
|
80
|
Zhang BW, Cai HF, Wei XF, Sun JJ, Lan XY, Lei CZ, Lin FP, Qi XL, Plath M, Chen H. miR-30-5p Regulates Muscle Differentiation and Alternative Splicing of Muscle-Related Genes by Targeting MBNL. Int J Mol Sci 2016; 17:ijms17020182. [PMID: 26840300 PMCID: PMC4783916 DOI: 10.3390/ijms17020182] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/26/2015] [Accepted: 01/22/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs), a class of single stranded, small (~22 nucleotides), non-coding RNAs, play an important role in muscle development. We focused on the role of the miR-30-5p family during bovine muscle development from previous high-throughput sequencing results and analyzed their expression profiles. MHC and MyoG mRNAs expression as well as their proteins were suppressed in differentiated C2C12 cells, suggesting the importance of miR-30-5p in muscle development. MBNL, the candidate target of miR-30-5p, is an alternative splicing regulation factor. MBNL1 and MBNL3 have opposite effects on muscle differentiation. Our results confirmed that miR-30a-5p and miR-30e-5p repress the expression of MBNL1, MBNL2 and MBNL3, whereas miR-30b-5p inhibits MBNL1 and MBNL2 expression. This provides direct evidence that MBNL expression can be flexibly regulated by miR-30-5p. Previous studies showed that MBNL1 promotes exon inclusion of two muscle-related genes (Trim55 and INSR). Through RNA splicing studies, we found that miR-30-5p had an effect on their alternative splicing, which means miR-30-5p via MBNL1 could be integrated into muscle signaling pathways in which INSR or Trim55 are located. In conclusion, miR-30-5p could inhibit muscle cell differentiation and regulate the alternative splicing of Trim55 and INSR by targeting MBNL. These results promote the understanding of the function of miRNAs in muscle development.
Collapse
Affiliation(s)
- Bo-Wen Zhang
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Han-Fang Cai
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xue-Feng Wei
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jia-Jie Sun
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xian-Yong Lan
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Chu-Zhao Lei
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Feng-Peng Lin
- Department of Animal Husbandry, Bureau of Biyang County of Henan province, Biyang 463700, Henan, China.
| | - Xing-Lei Qi
- Department of Animal Husbandry, Bureau of Biyang County of Henan province, Biyang 463700, Henan, China.
| | - Martin Plath
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Hong Chen
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
81
|
Ekman M, Albinsson S, Uvelius B, Swärd K. MicroRNAs in Bladder Outlet Obstruction: Relationship to Growth and Matrix Remodelling. Basic Clin Pharmacol Toxicol 2016; 119 Suppl 3:5-17. [DOI: 10.1111/bcpt.12534] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/12/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Mari Ekman
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | | | - Bengt Uvelius
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - Karl Swärd
- Department of Experimental Medical Science; Lund University; Lund Sweden
| |
Collapse
|
82
|
Zhao YG, Meng FX, Li BW, Sheng YM, Liu MM, Wang B, Li HW, Xiu RJ. Gelatinases promote calcification of vascular smooth muscle cells by up-regulating bone morphogenetic protein-2. Biochem Biophys Res Commun 2016; 470:287-293. [PMID: 26797522 DOI: 10.1016/j.bbrc.2016.01.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 01/10/2016] [Indexed: 02/01/2023]
Abstract
Matrix metalloproteinase-2 (MMP-2), also known as gelatinase A, is involved in vascular calcification. Another member of gelatinases is MMP-9 (gelatinase B). However, the role of gelatinases in the pathogenesis of vascular calcification is not well understood. The current study aims to clarify the relationship between gelatinases and vascular calcification and to elucidate the underlying mechanism. Beta-glycerophosphate (β-GP) was used to induce calcification of vascular smooth muscle cells (VSMCs) with or without 2-[[(4-Phenoxyphenyl)sulfonyl]methyl]-thiirane (SB-3CT), a specific gelatinases inhibitor. Levels of calcification were determined by assessing calcium content and calcification area of VSMCs. Phenotype transition of VSMCs was observed by assessing expressions of alkaline phosphatase (ALP), smooth muscle α-actin (SM-α-actin) and desmin. Gelatin zymography was applied to determine the activities of gelatinases, and western blot was applied to determine expressions of gelatinases, bone morphogenetic protein-2 (BMP-2), Runt-related transcription factor 2 (RUNX2) and msh homeobox homolog 2 (Msx-2). Gelatinases inhibition by SB-3CT alleviated calcification and phenotype transition of VSMCs induced by β-GP. Increased gelatinases expression and active MMP-2 were observed in calcifying VSMCs. Gelatinases inhibition reduced expression of RUNX2, Msx-2 and BMP-2. BMP-2 treatment increased expressions of RUNX2 and Msx-2, while noggin, an antagonist of BMP-2, decreased expressions of RUNX2 and Msx-2. Gelatinases promote vascular calcification by upregulating BMP-2 which induces expression of RUNX2 and Msx-2, two proteins associated with phenotype transition of VSMCs in vascular calcification. Interventions targeting gelatinases inhibition might be a proper candidate for ameliorating vascular calcification.
Collapse
Affiliation(s)
- Yong-Gang Zhao
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Fan-Xing Meng
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Bing-Wei Li
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - You-Ming Sheng
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Ming-Ming Liu
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Bing Wang
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Hong-Wei Li
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Rui-Juan Xiu
- Institute of Microcirculation and Key Laboratory, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
83
|
Abstract
Vascular calcification can lead to cardiovascular morbidity and mortality. The initiating factors and clinical consequences depend on the underlying disease state and location of the calcification. The pathogenesis of vascular calcification is complex and involves a transformation of vascular smooth muscle cells to an osteo/chondrocytic cell that expresses RUNX2 and produces matrix vesicles. The imbalance of promoters (such as hyperphosphatemia and hypercalcemia) and inhibitors (e.g., fetuin-A) is critical in the development of vascular calcification. The altered mineral metabolism and deficiency in inhibitors are common in patients with chronic kidney disease (CKD) and is one reason why vascular calcification is so prevalent in that population.
Collapse
Affiliation(s)
- Neal X Chen
- Department of Medicine, Indiana University School of Medicine, 950 W. Walnut Street, R2-202, Indianapolis, IN, 46202, USA.
| | - Sharon M Moe
- Department of Medicine, Indiana University School of Medicine, 950 W. Walnut Street, R2-202, Indianapolis, IN, 46202, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 950 W. Walnut Street, R2-202, Indianapolis, IN, 46202, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| |
Collapse
|
84
|
Avogaro A, Fadini GP. Mechanisms of ectopic calcification: implications for diabetic vasculopathy. Cardiovasc Diagn Ther 2015; 5:343-52. [PMID: 26543821 DOI: 10.3978/j.issn.2223-3652.2015.06.05] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular calcification (VC) is the deposition of calcium/phosphate in the vasculature, which portends a worse clinical outcome and predicts major adverse cardiovascular events. VC is an active process initiated and regulated via a variety of molecular signalling pathways. There are mainly two types of calcifications: the media VC and the intima VC. All major risk factors for cardiovascular disease (CVD) have been linked to the presence/development of VC. Besides the risk factors, a genetic component is also operative to determine arterial calcification. Several events take place before VC is established, including inflammation, trans-differentiation of vascular cells and homing of circulating pro-calcific cells. Diabetes is an important predisposing factor for VC. Compared with non-diabetic subjects, patients with diabetes show increased VC and higher expression of bone-related proteins in the medial layer of the vessels. In this review we will highlight the mechanisms underlying vascular calcification in diabetic patients.
Collapse
Affiliation(s)
- Angelo Avogaro
- 1 Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy ; 2 Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gian Paolo Fadini
- 1 Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy ; 2 Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
85
|
Panizo S, Naves-Díaz M, Carrillo-López N, Martínez-Arias L, Fernández-Martín JL, Ruiz-Torres MP, Cannata-Andía JB, Rodríguez I. MicroRNAs 29b, 133b, and 211 Regulate Vascular Smooth Muscle Calcification Mediated by High Phosphorus. J Am Soc Nephrol 2015; 27:824-34. [PMID: 26187577 DOI: 10.1681/asn.2014050520] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 06/11/2015] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification is a frequent cause of morbidity and mortality in patients with CKD and the general population. The common association between vascular calcification and osteoporosis suggests a link between bone and vascular disorders. Because microRNAs (miRs) are involved in the transdifferentiation of vascular smooth muscle cells into osteoblast-like cells, we investigated whether miRs implicated in osteoblast differentiation and bone formation are involved in vascular calcification. Different levels of uremia, hyperphosphatemia, and aortic calcification were induced by feeding nephrectomized rats a normal or high-phosphorus diet for 12 or 20 weeks, at which times the levels of eight miRs (miR-29b, miR-125, miR-133b, miR-135, miR-141, miR-200a, miR-204, and miR-211) in the aorta were analyzed. Compared with controls and uremic rats fed a normal diet, uremic rats fed a high-phosphorous diet had lower levels of miR-133b and miR-211 and higher levels of miR-29b that correlated respectively with greater expression of osteogenic RUNX2 and with lower expression of several inhibitors of osteoblastic differentiation. Uremia per se mildly reduced miR-133b levels only. Similar results were obtained in two in vitro models of vascular calcification (uremic serum and high-calcium and -phosphorus medium), and experiments using antagomirs and mimics to modify miR-29b, miR-133b, and miR-211 expression levels in these models confirmed that these miRs regulate the calcification process. We conclude that miR-29b, miR-133b, and miR-211 have direct roles in the vascular smooth muscle calcification induced by high phosphorus and may be new therapeutic targets in the management of vascular calcification.
Collapse
Affiliation(s)
- Sara Panizo
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - José Luis Fernández-Martín
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - María Piedad Ruiz-Torres
- Department of Systems Biology, Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Faculty of Medicine, University of Alcalá, Alcalá de Henares, Madrid, Spain; and
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain; Department of Medicine, University of Oviedo, Oviedo, Asturias, Spain
| | - Isabel Rodríguez
- Bone and Mineral Research Unit, Reina Sofia Institute of Renal Research (IRSIN), Renal Research Network (REDinREN) from Carlos III Health Institute (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain;
| |
Collapse
|
86
|
Di Y, Zhang D, Hu T, Li D. miR-23 regulate the pathogenesis of patients with coronary artery disease. Int J Clin Exp Med 2015; 8:11759-11769. [PMID: 26380016 PMCID: PMC4565399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/23/2015] [Indexed: 06/05/2023]
Abstract
To study whether miR-23 is regulated in coronary artery disease (CAD) patients and what is the possible mechanism of miR-23 in regulating CAD progression. Method Three different cohorts (including 13 AMI patients, 176 angina pectoris patients and 127 control subjects) were enrolled to investigate the expression levels of circulating miR-23 in patients with myocardial ischemia and also the relationship between plasma miR-23 and severity of coronary stenosis. Plasma miR-23 levels of participants were examined by real-time quantitative PCR. We further detected the correlation of miR-23 and VEGF by molecular and animal assays. Result miR-23 was enriched in not only diseased endothelial progenitor cells (EPCs) but also the plasma of CAD patients. Besides, we found out miR-23 was able to suppress VEGF expression and EPC activities. Reporter assays confirmed the direct binding and repression of miR-23 to the 3'-UTR of VEGF mRNA. Knock down of miR-23 not only restored VEGF levels and angiogenic activities of diseased EPCs in vitro, but further promoted blood flow recovery in ischemic limbs of mice. Conclusion Circulating miR-23 may be a new biomarker for CAD and as a potential diagnostic tool. And increased miR-23 level may be used to predict the presence and severity of coronary lesions in CAD patients.
Collapse
Affiliation(s)
- Yunfeng Di
- Department of Cardiology, Sichuan Mianyang 404 Hospital (The Second Affiliated Hospital of North Sichuan Medical College) Sichuan, China
| | - Dayong Zhang
- Department of Cardiology, Sichuan Mianyang 404 Hospital (The Second Affiliated Hospital of North Sichuan Medical College) Sichuan, China
| | - Teng Hu
- Department of Cardiology, Sichuan Mianyang 404 Hospital (The Second Affiliated Hospital of North Sichuan Medical College) Sichuan, China
| | - Decai Li
- Department of Cardiology, Sichuan Mianyang 404 Hospital (The Second Affiliated Hospital of North Sichuan Medical College) Sichuan, China
| |
Collapse
|
87
|
Kurakula K, Goumans MJ, Ten Dijke P. Regulatory RNAs controlling vascular (dys)function by affecting TGF-ß family signalling. EXCLI JOURNAL 2015; 14:832-50. [PMID: 26862319 PMCID: PMC4743484 DOI: 10.17179/excli2015-423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/15/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Over the last few years, microRNAs (miRNAs) have emerged as master regulators of gene expression in cardiovascular biology and disease. miRNAs are small endogenous non-coding RNAs that usually bind to 3′ untranslated region (UTR) of their target mRNAs and inhibit mRNA stability or translation of their target genes. miRNAs play a dynamic role in the pathophysiology of many CVDs through their effects on target mRNAs in vascular cells. Recently, numerous miRNAs have been implicated in the regulation of the transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signalling pathway which plays crucial roles in diverse biological processes, and is involved in pathogenesis of many diseases including CVD. This review gives an overview of current literature on the role of miRNAs targeting TGF-β/BMP signalling in vascular cells, including endothelial cells and smooth muscle cells. We also provide insight into how this miRNA-mediated regulation of TGF-β/BMP signalling might be used to harness CVD.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-Jose Goumans
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
88
|
MicroRNA-204 Targets Runx2 to Attenuate BMP-2-induced Osteoblast Differentiation of Human Aortic Valve Interstitial Cells. J Cardiovasc Pharmacol 2015; 66:63-71. [DOI: 10.1097/fjc.0000000000000244] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
89
|
Abstract
Arterial calcification is highly prevalent and correlated with cardiovascular mortality, especially in patients with ESRD or diabetes. The pathogenesis of arterial calcification is multifactorial, with both genetic and environmental factors being implicated. In recent years, several mechanisms contributing to arterial calcification have been proposed. However, these can only explain a small proportion of the variability in arterial calcification, which is a major obstacle for its prevention and management. Epigenetics has emerged as one of the most promising areas that may fill in some of the gaps in our current knowledge of the interaction between the environmental insults with gene regulation in the development of diseases. Epigenetics refers to heritable and acquired changes in gene transcription that occur independently of the DNA sequence. Well-known components of epigenetic regulation include DNA methylation, histone modifications, and microRNAs. Epigenetics research in the regulation of arterial calcification has only recently been elucidated. In this review, we will summarise recent progress in epigenetic pathways involved in arterial calcification and discuss potential therapeutic interventions based on epigenetic mechanisms.
Collapse
|
90
|
Miano JM, Long X. The short and long of noncoding sequences in the control of vascular cell phenotypes. Cell Mol Life Sci 2015; 72:3457-88. [PMID: 26022065 DOI: 10.1007/s00018-015-1936-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022]
Abstract
The two principal cell types of importance for normal vessel wall physiology are smooth muscle cells and endothelial cells. Much progress has been made over the past 20 years in the discovery and function of transcription factors that coordinate proper differentiation of these cells and the maintenance of vascular homeostasis. More recently, the converging fields of bioinformatics, genomics, and next generation sequencing have accelerated discoveries in a number of classes of noncoding sequences, including transcription factor binding sites (TFBS), microRNA genes, and long noncoding RNA genes, each of which mediates vascular cell differentiation through a variety of mechanisms. Alterations in the nucleotide sequence of key TFBS or deviations in transcription of noncoding RNA genes likely have adverse effects on normal vascular cell phenotype and function. Here, the subject of noncoding sequences that influence smooth muscle cell or endothelial cell phenotype will be summarized as will future directions to further advance our understanding of the increasingly complex molecular circuitry governing normal vascular cell differentiation and how such information might be harnessed to combat vascular diseases.
Collapse
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642, USA,
| | | |
Collapse
|
91
|
Qi F, He T, Jia L, Song N, Guo L, Ma X, Wang C, Xu M, Fu Y, Li L, Luo Y. The miR-30 Family Inhibits Pulmonary Vascular Hyperpermeability in the Premetastatic Phase by Direct Targeting of Skp2. Clin Cancer Res 2015; 21:3071-80. [PMID: 25810374 DOI: 10.1158/1078-0432.ccr-14-2785] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/22/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Before metastasis, primary tumor can create a premetastatic niche in distant organ to facilitate the dissemination of tumor cells. In the premetastatic phase, the permeability of pulmonary vasculatures is increased to accelerate the extravasation of circulating tumor cells. However, it is not clear whether local miRNAs contribute to the vascular hyperpermeability of the premetastatic niche. EXPERIMENTAL DESIGN The expression of total miRNAs was determined using microarray in series of premetastatic lungs from tumor-bearing mice. Significantly differentially expressed miRNAs were identified and validated with qRT-PCR. Vascular permeability assays, vascular mimic systems, and orthotopic tumor models were used to investigate roles of selected miRNAs and target genes in premetastatic hyperpermeability. RESULTS We identified a miRNA signature in premetastatic lungs. Among these miRNAs, miR-30a, b, c, d, and e were significantly attenuated. Subsequent investigations elucidated that lung fibroblast-derived miR-30s stabilized pulmonary vessels. Overexpression of miR-30s in lungs postponed metastasis and extended overall survival of B16 tumor-bearing mice. Following studies uncovered that Skp2 was directly targeted by miR-30s. Overexpression of Skp2 could disrupt pulmonary vessels, promote lung metastasis, and decrease overall survival of B16 tumor-bearing mice. CONCLUSIONS These findings illuminate a novel mechanism for the modulation of premetastatic niches by miR-30s, which suggest that miR-30s represent not only promising targets for antimetastasis therapy but also indicators for metastasis.
Collapse
Affiliation(s)
- Feifei Qi
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ting He
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lin Jia
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nan Song
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lifang Guo
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuhui Ma
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Chunying Wang
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Min Xu
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yan Fu
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lin Li
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yongzhang Luo
- National Engineering Laboratory for Anti-tumor Protein Therapeutics, Beijing, China. Beijing Key Laboratory for Protein Therapeutics, Beijing, China. Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
92
|
Han F, Huo Y, Huang CJ, Chen CL, Ye J. MicroRNA-30b promotes axon outgrowth of retinal ganglion cells by inhibiting Semaphorin3A expression. Brain Res 2015; 1611:65-73. [PMID: 25791621 DOI: 10.1016/j.brainres.2015.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/08/2015] [Accepted: 03/06/2015] [Indexed: 10/23/2022]
Abstract
Semaphorin3A (Sema3A) is a major inhibitory factor of optic nerve (ON) regeneration post-injury. Many microRNAs (miRNAs) are expressed specifically in the mammalian brain and retina and are dynamically regulated during development, suggesting that this group of miRNAs may be associated with neural development. We found that microRNA-30b (miR-30b) bound to the three prime untranslated region (3' UTR) of Sema3A and inhibited the expression of Sema3A mRNA. The mRNA expression level of miR-30b and the protein expression levels of Sema3A, Neuropilin1 (NRP1), PlexinA1 (PlexA1), phosphorylated p38MAPK (p-p38MAPK), and active caspase-3 were all upregulated in retinas from rats with a damaged ON relative to those with an intact ON. Transfection of cultured retinal ganglion cells (RGCs) with an miR-30b mimic led to decreased levels of Sema3A, NRP1, PlexA1, p-p38MAPK, and active caspase-3 protein expression, as well as axon elongation and reduced levels of apoptosis. These findings provide evidence that miR-30b inhibits Sema3A expression. Decreased Sema3A expression promotes axon outgrowth in RGCs due to reduced levels of Sema3A binding to NRP1 and PlexA1 and simultaneously reduces apoptosis by inhibiting the p38MAPK and caspase-3 pathways. Our findings provide the first evidence that miR-30b-mediated Sema3A downregulation may serve as a new strategy for the clinical treatment of ON injury.
Collapse
Affiliation(s)
- F Han
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y Huo
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - C-J Huang
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - C-L Chen
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - J Ye
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
93
|
Li T, Sun ZL, Xie QY. Protective effect of microRNA-30b on hypoxia/reoxygenation-induced apoptosis in H9C2 cardiomyocytes. Gene 2015; 561:268-75. [PMID: 25701595 DOI: 10.1016/j.gene.2015.02.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/30/2015] [Accepted: 02/14/2015] [Indexed: 12/23/2022]
Abstract
We examined the protective role of microRNA-30b (miR-30b) in ischemia-reperfusion (I/R)-induced injury in rat H9C2 cardiomyocytes. H9C2 cells were subjected to hypoxia-reoxygenation (H/R) treatment to simulate ischemia-reperfusion (I/R) injury. H9C2 cells were divided into: vehicle control (VC) group; scrambled inhibitors (INC) group; scrambled mimics (MNC) group; H/R+VC group; H/R+INC group; H/R+mimics group. H/R induced apoptosis was detected by flow cytometry and the pathways involved in miR-30b-mediated protection were examined by analyzing the expression of miR-30b, Bcl-2, Bax, Caspase-3, KRAS, p-AKT and total AKT in H9C2 cells. Overexpression of miR-30b mimic (H/R+mimics group) significantly increased Bcl-2 and Bcl-2/Bax levels and decreased Bax and Caspase-3 levels, compared with the H/R+VC group (all P<0.05). Consistent with this, the apoptosis rate was significantly decreased in the H/R+mimics group (P<0.05) compared with the H/R+VC group. Western blot analysis revealed that overexpression of miR-30b mimic resulted in significantly increase in AKT activation and decreased KRAS, compared to the H/R+VC group (both P<0.05). In conclusion, the H/R induced apoptosis decreased miR-30b expression, but over-expression of miR-30b inhibited H/R induced apoptosis. The observed miR-30b-mediated protection against H/R induced apoptosis involved the upregulation of Ras-PI3K-Akt pathway.
Collapse
Affiliation(s)
- Tong Li
- Department of Emergency, The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Ze-Lin Sun
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Ying Xie
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
94
|
Prandi F, Piola M, Soncini M, Colussi C, D’Alessandra Y, Penza E, Agrifoglio M, Vinci MC, Polvani G, Gaetano C, Fiore GB, Pesce M. Adventitial vessel growth and progenitor cells activation in an ex vivo culture system mimicking human saphenous vein wall strain after coronary artery bypass grafting. PLoS One 2015; 10:e0117409. [PMID: 25689822 PMCID: PMC4331547 DOI: 10.1371/journal.pone.0117409] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/21/2014] [Indexed: 01/09/2023] Open
Abstract
Saphenous vein graft disease is a timely problem in coronary artery bypass grafting. Indeed, after exposure of the vein to arterial blood flow, a progressive modification in the wall begins, due to proliferation of smooth muscle cells in the intima. As a consequence, the graft progressively occludes and this leads to recurrent ischemia. In the present study we employed a novel ex vivo culture system to assess the biological effects of arterial-like pressure on the human saphenous vein structure and physiology, and to compare the results to those achieved in the presence of a constant low pressure and flow mimicking the physiologic vein perfusion. While under both conditions we found an activation of Matrix Metallo-Proteases 2/9 and of microRNAs-21/146a/221, a specific effect of the arterial-like pressure was observed. This consisted in a marked geometrical remodeling, in the suppression of Tissue Inhibitor of Metallo-Protease-1, in the enhanced expression of TGF-β1 and BMP-2 mRNAs and, finally, in the upregulation of microRNAs-138/200b/200c. In addition, the veins exposed to arterial-like pressure showed an increase in the density of the adventitial vasa vasorum and of cells co-expressing NG2, CD44 and SM22α markers in the adventitia. Cells with nuclear expression of Sox-10, a transcription factor characterizing multipotent vascular stem cells, were finally found in adventitial vessels. Our findings suggest, for the first time, a role of arterial-like wall strain in the activation of pro-pathologic pathways resulting in adventitial vessels growth, activation of vasa vasorum cells, and upregulation of specific gene products associated to vascular remodeling and inflammation.
Collapse
Affiliation(s)
- Francesca Prandi
- Unità di Ingegneria Tissutale, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Marco Piola
- Politecnico di Milano, Dipartimento di Elettronica, Informazione e Bioingegneria, Milan, Italy
| | - Monica Soncini
- Politecnico di Milano, Dipartimento di Elettronica, Informazione e Bioingegneria, Milan, Italy
| | - Claudia Colussi
- Istituto di Patologia Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Yuri D’Alessandra
- Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Eleonora Penza
- II Divisione di Cardiochirurgia, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Marco Agrifoglio
- Dipartimento di Scienze Cliniche e di Comunità, Università di Milano, Milan, Italy
| | | | - Gianluca Polvani
- Dipartimento di Scienze Cliniche e di Comunità, Università di Milano, Milan, Italy
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Goethe University, Frankfurt-am-Main, Germany
| | | | - Maurizio Pesce
- Unità di Ingegneria Tissutale, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- * E-mail:
| |
Collapse
|
95
|
MiR-214 regulates the pathogenesis of patients with coronary artery disease by targeting VEGF. Mol Cell Biochem 2015; 402:111-22. [PMID: 25575606 DOI: 10.1007/s11010-014-2319-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/23/2014] [Indexed: 12/11/2022]
Abstract
Circulating microRNAs (miRNAs) in patient body fluids have recently been considered to hold the potential of being novel disease biomarkers and drug targets. We aimed to investigate the correlation between the levels of circulating miR-214 and the expression of vascular endothelial growth factor (VEGF) in the pathogenesis of coronary heart disease patients to further explore the mechanism involved in the vasculogenesis. Three different cohorts, including 13 acute myocardial infarction patients, 176 angina pectoris patients, and 127 control subjects, were enrolled to investigate the expression levels of circulating miR-214 in patients with myocardial ischemia and also the relationship between plasma miR-214 and severity of coronary stenosis. Plasma miR-214 levels of participants were examined by real-time quantitative PCR. Simultaneously, plasma cardiac troponin I concentrations were measured by ELISA assays. We further detected the correlation of miR-214 and VEGF by molecular and animal assays. MiR-214 was enriched in not only diseased endothelial progenitor cells (EPCs) but also the plasma of coronary artery disease (CAD) patients. Besides, we found out miR-214 was able to suppress VEGF expression and EPC activities. Reporter assays confirmed the direct binding and repression of miR-214 to the 39-UTR of VEGF mRNA. Knockdown of miR-214 not only restored VEGF levels and angiogenic activities of diseased EPCs in vitro, but also further promoted blood flow recovery in ischemic limbs of mice. Circulating miR-214 may be a new biomarker for CAD and as a potential diagnostic tool. And increased miR-214 level may be used to predict the presence and severity of coronary lesions in CAD patients.
Collapse
|
96
|
Hata A, Kang H. Functions of the bone morphogenetic protein signaling pathway through microRNAs (review). Int J Mol Med 2015; 35:563-8. [PMID: 25571950 PMCID: PMC6904101 DOI: 10.3892/ijmm.2015.2060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) have emerged as key regulators of gene expression in essential cellular processes, such as cell growth, differentiation and development. Recent findings have established that the levels of miRNAs are modulated by cell signaling mechanisms, including the bone morphogenetic protein (BMP) signaling pathway. The BMP signaling pathway controls diverse cellular activities by modulating the levels of miRNAs, indicating the complexity of gene regulation by the BMP signaling pathway. The tight regulation of the levels of miRNAs is critical for maintaining normal physiological conditions, and dysregulated miRNA levels contribute to the development of diseases. In the present review, we discuss different insights (provided over the past decade) into the regulation of miRNAs governed by the BMP signaling pathway and the implications of this regulation on the understanding of the cellular differentiation of vascular smooth muscle cells (VSMCs), osteoblasts and neuronal cells.
Collapse
Affiliation(s)
- Akiko Hata
- Cardiovascular Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406-772, Republic of Korea
| |
Collapse
|
97
|
Evrard S, Delanaye P, Kamel S, Cristol JP, Cavalier E. Vascular calcification: from pathophysiology to biomarkers. Clin Chim Acta 2015; 438:401-14. [PMID: 25236333 DOI: 10.1016/j.cca.2014.08.034] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/27/2014] [Accepted: 08/29/2014] [Indexed: 02/06/2023]
Abstract
The link between vascular calcification (VC) and increased mortality is now well established. Over time, as clinical importance of this phenomenon has begun to be fully considered, scientists have highlighted more and more physiopathological mechanisms and signaling pathways that underlie VC. Several conditions such as diabetes, dyslipidemia and renal diseases are undoubtedly identified as predisposing factors. But even if the process is better understood, many questions still remain unanswered. This review briefly develops the various theories that attempt to explain mineralization genesis. Nonetheless, the main purpose of the article is to provide a profile of the various existing biomarkers of VC. Indeed, in the past years, a lot of inhibitors and promoters, which form a dense and interconnected network, were identified. Given importance to assess and control mineralization process, a focusing on accumulated knowledge of each marker seemed to be necessary. Therefore, we tried to define their respective role in the physiopathology and how they can contribute to calcification risk assessment. Among these, Klotho/fibroblast growth factor-23, fetuin-A, Matrix Gla protein, Bone morphogenetic protein-2, osteoprotegerin, osteopontin, osteonectin, osteocalcin, pyrophosphate and sclerostin are specifically discussed.
Collapse
Affiliation(s)
- Séverine Evrard
- Department of Clinical Chemistry, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - Pierre Delanaye
- Department of Nephrology, Dialysis and Hypertension, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - Said Kamel
- Laboratoire de Biochimie, CHU Amiens, Amiens, France; INSERM U1088, Université de Picardie Jules-Verne, Amiens, France
| | - Jean-Paul Cristol
- Laboratoire de Biochimie, CHRU de Montpellier, Hôpital Lapeyronie, Montpellier, France
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart-Tilman, Liège, Belgium.
| |
Collapse
|
98
|
Hamburg NM, Leeper NJ. Therapeutic Potential of Modulating MicroRNA in Peripheral Artery Disease. Curr Vasc Pharmacol 2015; 13:316-23. [PMID: 23713861 PMCID: PMC4886469 DOI: 10.2174/15701611113119990014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 12/16/2022]
Abstract
Peripheral artery disease (PAD) produces significant disability attributable to lower extremity ischemia. Limited treatment modalities exist to ameliorate clinical symptoms in patients with PAD. Growing evidence links microRNAs to key processes that govern disease expression in PAD including angiogenesis, endothelial function, inflammation, vascular regeneration, vascular smooth muscle cell function, restenosis, and mitochondrial function. MicroRNAs have been identified in circulation and may serve as novel biomarkers in PAD. This article reviews the potential contribution of microRNA to key pathways of disease development in PAD that may lead to microRNA-based diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Naomi M Hamburg
- Section of Cardiology, Boston Medical Center, 88 East Newton St., Boston, MA, 02118.
| | | |
Collapse
|
99
|
Vascular calcification: Mechanisms of vascular smooth muscle cell calcification. Trends Cardiovasc Med 2014; 25:267-74. [PMID: 25435520 DOI: 10.1016/j.tcm.2014.10.021] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/02/2014] [Accepted: 10/25/2014] [Indexed: 11/24/2022]
Abstract
Vascular calcification is highly prevalent and, when present, is associated with major adverse cardiovascular events. Vascular smooth muscle cells play an integral role in mediating vessel calcification by undergoing differentiation to osteoblast-like cells and generating matrix vesicles that serve as a nidus for calcium-phosphate deposition in the vessel wall. Once believed to be a passive process, it is now recognized that vascular calcification is a complex and highly regulated process that involves activation of cellular signaling pathways, circulating inhibitors of calcification, genetic factors, and hormones. This review will examine several of the key mechanisms linking vascular smooth muscle cells to vessel calcification that may be targeted to reduce vessel wall mineralization and, thereby, reduce cardiovascular risk.
Collapse
|
100
|
MicroRNAs Regulate Vascular Medial Calcification. Cells 2014; 3:963-80. [PMID: 25317928 PMCID: PMC4276909 DOI: 10.3390/cells3040963] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 01/08/2023] Open
Abstract
Vascular calcification is highly prevalent in patients with coronary artery disease and, when present, is associated with major adverse cardiovascular events, including an increased risk of cardiovascular mortality. The pathogenesis of vascular calcification is complex and is now recognized to recapitulate skeletal bone formation. Vascular smooth muscle cells (SMC) play an integral role in this process by undergoing transdifferentiation to osteoblast-like cells, elaborating calcifying matrix vesicles and secreting factors that diminish the activity of osteoclast-like cells with mineral resorbing capacity. Recent advances have identified microRNAs (miRs) as key regulators of this process by directing the complex genetic reprogramming of SMCs and the functional responses of other relevant cell types relevant for vascular calcification. This review will detail SMC and bone biology as it relates to vascular calcification and relate what is known to date regarding the regulatory role of miRs in SMC-mediated vascular calcification.
Collapse
|