201
|
Elia L, Quintavalle M. Epigenetics and Vascular Diseases: Influence of Non-coding RNAs and Their Clinical Implications. Front Cardiovasc Med 2017; 4:26. [PMID: 28497038 PMCID: PMC5406412 DOI: 10.3389/fcvm.2017.00026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/12/2017] [Indexed: 01/25/2023] Open
Abstract
Epigenetics refers to heritable mechanisms able to modulate gene expression that do not involve alteration of the genomic DNA sequence. Classically, mechanisms such as DNA methylation and histone modifications were part of this classification. Today, this field of study has been expanded and includes also the large class of non-coding RNAs (ncRNAs). Indeed, with the extraordinary possibilities introduced by the next-generation sequencing approaches, our knowledge of the mammalian transcriptome has greatly improved. Today, we have identifying thousands of ncRNAs, and unsurprisingly, a direct association between ncRNA dysregulation and development of cardiovascular pathologies has been identified. This class of gene modulators is further divided into short-ncRNAs and long-non-coding RNAs (lncRNAs). Among the short-ncRNA sub-group, the best-characterized players are represented by highly conserved RNAs named microRNAs (miRNAs). miRNAs principally inhibit gene expression, and their involvement in cardiovascular diseases has been largely studied. On the other hand, due to the different roles played by lncRNAs, their involvement in cardiovascular pathology development is still limited, and further studies are needed. For instance, in order to define their roles in the cellular processes associated with the development of diseases, we need to better characterize the details of their mechanisms of action; only then might we be able to develop innovative therapeutic strategies. In this review, we would like to give an overview of the current knowledge on the function of ncRNAs and their involvement in the development of vascular diseases.
Collapse
Affiliation(s)
- Leonardo Elia
- Humanitas Clinical and Research Center, Milan, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | |
Collapse
|
202
|
Recent Progress in Research on the Pathogenesis of Pulmonary Thromboembolism: An Old Story with New Perspectives. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6516791. [PMID: 28484717 PMCID: PMC5397627 DOI: 10.1155/2017/6516791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/26/2017] [Accepted: 03/27/2017] [Indexed: 12/25/2022]
Abstract
Pulmonary thromboembolism (PTE) is part of a larger clinicopathological entity, venous thromboembolism. It is also a complex, multifactorial disorder divided into four major disease processes including venous thrombosis, thrombus in transit, acute pulmonary embolism, and pulmonary circulation reconstruction. Even when treated, some patients develop chronic thromboembolic pulmonary hypertension. PTE is also a common fatal type of pulmonary vascular disease worldwide, but earlier studies primarily focused on the pathological changes in the blood component of the disease. With contemporary advances in molecular and cellular biology, people are becoming increasingly aware of coagulation pathways, the function of vascular smooth muscle cells, microparticles, and the inflammatory pathways that play key roles in PTE. Combined hypoxia and immune research has revealed that PTE should be regarded as a class of complex diseases caused by multiple factors involving the vascular microenvironment and vascular cell dysfunction.
Collapse
|
203
|
de Gonzalo-Calvo D, Cenarro A, Garlaschelli K, Pellegatta F, Vilades D, Nasarre L, Camino-Lopez S, Crespo J, Carreras F, Leta R, Catapano AL, Norata GD, Civeira F, Llorente-Cortes V. Translating the microRNA signature of microvesicles derived from human coronary artery smooth muscle cells in patients with familial hypercholesterolemia and coronary artery disease. J Mol Cell Cardiol 2017; 106:55-67. [PMID: 28342976 DOI: 10.1016/j.yjmcc.2017.03.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/13/2017] [Accepted: 03/21/2017] [Indexed: 12/17/2022]
Abstract
AIMS To analyze the impact of atherogenic lipoproteins on the miRNA signature of microvesicles derived from human coronary artery smooth muscle cells (CASMC) and to translate these results to familial hypercholesterolemia (FH) and coronary artery disease (CAD) patients. METHODS Conditioned media was collected after exposure of CASMC to atherogenic lipoproteins. Plasma samples were collected from two independent populations of diagnosed FH patients and matched normocholesterolemic controls (Study population 1, N=50; Study population 2, N=24) and a population of patients with suspected CAD (Study population 3, N=50). Extracellular vesicles were isolated and characterized using standard techniques. A panel of 30 miRNAs related to vascular smooth muscle cell (VSMC) (patho-)physiology was analyzed using RT-qPCR. RESULTS Atherogenic lipoproteins significantly reduced levels of miR-15b-5p, -24-3p, -29b-3p, -130a-3p, -143-3p, -146a-3p, -222-3p, -663a levels (P<0.050) in microvesicles (0.1μm-1μm in diameter) released by CASMC. Two of these miRNAs, miR-24-3p and miR-130a-3p, were reduced in circulating microvesicles from FH patients compared with normocholesterolemic controls in a pilot study (Study population 1) and in different validation studies (Study populations 1 and 2) (P<0.050). Supporting these results, plasma levels of miR-24-3p and miR-130a-3p were also downregulated in FH patients compared to controls (P<0.050). In addition, plasma levels of miR-130a-3p were inversely associated with coronary atherosclerosis in a cohort of suspected CAD patients (Study population 3) (P<0.050). CONCLUSIONS Exposure to atherogenic lipoproteins modifies the miRNA profile of CASMC-derived microvesicles and these alterations are reflected in patients with FH. Circulating miR-130a-3p emerges as a potential biomarker for coronary atherosclerosis.
Collapse
Affiliation(s)
- David de Gonzalo-Calvo
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.
| | - Ana Cenarro
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Lipid Unit and Molecular Research Laboratory, IIS Aragón, Hospital Universitario Miguel Servet, Universidad de Zaragoza, Zaragoza, Spain
| | - Katia Garlaschelli
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello B, Italy
| | - Fabio Pellegatta
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello B, Italy; IRCCS Multimedica, Milan, Italy
| | - David Vilades
- Cardiac Imaging Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Laura Nasarre
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Sandra Camino-Lopez
- Catalan Institute of Cardiovascular Sciences, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Javier Crespo
- Catalan Institute of Cardiovascular Sciences, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Francesc Carreras
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Cardiac Imaging Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rubén Leta
- Cardiac Imaging Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Giuseppe Danilo Norata
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello B, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Fernando Civeira
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Lipid Unit and Molecular Research Laboratory, IIS Aragón, Hospital Universitario Miguel Servet, Universidad de Zaragoza, Zaragoza, Spain
| | - Vicenta Llorente-Cortes
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Institute of Biomedical Research of Barcelona (IIBB) - Spanish National Research Council (CSIC), Barcelona, Spain.
| |
Collapse
|
204
|
Ran X, Xiao CH, Xiang GM, Ran XZ. Regulation of Embryonic Stem Cell Self-Renewal and Differentiation by MicroRNAs. Cell Reprogram 2017; 19:150-158. [PMID: 28277752 DOI: 10.1089/cell.2016.0048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRNAs) are posttranscriptional regulators of gene expression. They play an important role in various cellular processes such as apoptosis, differentiation, secretion, and proliferation. Embryonic stem cells (ESCs) are derived from the inner cell mass of the blastocyst stage of the embryo. miRNAs are critical factors for the self-renewal and differentiation of ESCs. In this review, we will focus on the role of miRNAs in the self-renewal and directional differentiation of ESCs. We will present the current knowledge on key points related to miRNA biogenesis and their function in ESCs.
Collapse
Affiliation(s)
- Xi Ran
- 1 Department of Medical Laboratory, Xinqiao Hospital, Third Military Medical University , Chongqing, China .,2 State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University , Chongqing, China
| | - Chun-Hong Xiao
- 3 Qingdao First Sanatorium of Jinan Military Command , Qingdao, China
| | - Gui-Ming Xiang
- 1 Department of Medical Laboratory, Xinqiao Hospital, Third Military Medical University , Chongqing, China
| | - Xin-Ze Ran
- 2 State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University , Chongqing, China
| |
Collapse
|
205
|
Abstract
Pulmonary hypertension (PH) is a multifaceted vascular disease where development and severity are determined by both genetic and environmental factors. Over the past decade, there has been an acceleration of the discovery of molecular effectors that mediate PH pathogenesis, including large numbers of microRNA molecules that are expressed in pulmonary vascular cell types and exert system-wide regulatory functions in all aspects of vascular health and disease. Due to the inherent pleiotropy, overlap, and redundancy of these molecules, it has been challenging to define their integrated effects on overall disease manifestation. In this review, we summarize our current understanding of the roles of microRNAs in PH with an emphasis on potential methods to discern the hierarchical motifs governing their multifunctional and interconnected activities. Deciphering this higher order of regulatory structure will be crucial for overcoming the challenges of developing these molecules as biomarkers or therapeutic targets, in isolation or combination.
Collapse
|
206
|
Gabunia K, Herman AB, Ray M, Kelemen SE, England RN, DeLa Cadena R, Foster WJ, Elliott KJ, Eguchi S, Autieri MV. Induction of MiR133a expression by IL-19 targets LDLRAP1 and reduces oxLDL uptake in VSMC. J Mol Cell Cardiol 2017; 105:38-48. [PMID: 28257760 DOI: 10.1016/j.yjmcc.2017.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.
Collapse
Affiliation(s)
- Khatuna Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Allison B Herman
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Ross N England
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Raul DeLa Cadena
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - William J Foster
- Departments of Ophthalmology & Bioengineering, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Katherine J Elliott
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Satoru Eguchi
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
207
|
Wang N, Chen C, Yang D, Liao Q, Luo H, Wang X, Zhou F, Yang X, Yang J, Zeng C, Wang WE. Mesenchymal stem cells-derived extracellular vesicles, via miR-210, improve infarcted cardiac function by promotion of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2085-2092. [PMID: 28249798 DOI: 10.1016/j.bbadis.2017.02.023] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/18/2017] [Accepted: 02/21/2017] [Indexed: 01/11/2023]
Abstract
Mesenchymal stem cells (MSCs) exert therapeutic effect on treating acute myocardial infarction. Recent evidence showed that paracrine function rather than direct differentiation predominately contributes to the beneficial effects of MSCs, but how the paracrine factors function are not fully elucidated. In the present study, we tested if extracellular vesicles (EVs) secreted by MSC promotes angiogenesis in infracted heart via microRNAs. Immunostaining of CD31 and matrigel plug assay were performed to detect angiogenesis in a mouse myocardial infarction (MI) model. The cardiac function and structure was examined with echocardiographic analysis. Capillary-like tube formation, migration and proliferation of human umbilical vein endothelial cells (HUVECs) were determined. As a result, MSC-EVs significantly improved angiogenesis and cardiac function in post-MI heart. MSC-EVs increased the proliferation, migration and tube formation capacity of HUVECs. MicroRNA (miR)-210 was found to be enriched in MSC-EVs. The EVs collected from MSCs with miR-210 silence largely lost the pro-angiogenic effect both in-vitro and in-vivo. The miR-210 target gene Efna3, which plays a role in angiogenesis, was down-regulated by MSC-EVs treatment in HUVECs. In conclusion, MSC-EVs are sufficient to improve angiogenesis and exert therapeutic effect on MI, its pro- angiogenesis effect might be associated with a miR-210-Efna3 dependent mechanism. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Na Wang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Caiyu Chen
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dezhong Yang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Qiao Liao
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hao Luo
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xinquan Wang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Faying Zhou
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaoli Yang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jian Yang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Chunyu Zeng
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | - Wei Eric Wang
- Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
208
|
Yuan HY, Zhou CB, Chen JM, Liu XB, Wen SS, Xu G, Zhuang J. MicroRNA-34a targets regulator of calcineurin 1 to modulate endothelial inflammation after fetal cardiac bypass in goat placenta. Placenta 2017; 51:49-56. [PMID: 28292468 DOI: 10.1016/j.placenta.2017.01.128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/08/2017] [Accepted: 01/29/2017] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Placental dysfunction characterized by vascular endothelial inflammation is one of the most notable responses to fetal cardiac bypass. Regulator of calcineurin 1 (RCAN1) is an important regulator of inflammatory responses. MicroRNAs (miRNAs) are essential post-transcriptional modulators of gene expression, and miRNA-34a (miR-34a) was showed to activate vascular endothelial inflammation. We hypothesized that miR-34a may be a key regulator of placental dysfunction after fetal cardiac bypass. METHODS We evaluated miRNA expression in goat placentas via small RNA sequencing, quantitative real-time polymerase chain reaction (qRT-PCR) and in situ hybridization. Expression of miRNA target genes was determined via bioinformatics analyses and dual luciferase reporter assays. Furthermore, human umbilical vein endothelial cells (HUVECs) were transfected with miR-34a or a control sequence. The RCAN1, nuclear factor of activated T-cells (NFATC1) and nuclear factor kappa-B (NF-κB) levels in HUVECs and placentas were evaluated via Western blot and qRT-PCR. RESULTS We demonstrated that miR-34a was highly enriched in goat placenta after cardiopulmonary bypass. Moreover, RCAN1 was identified as a novel direct target of miR-34a. Transfection of miR-34a led to decreased RCAN1 expression and increased NFATC1 and NF-κB expression in HUVECs. Conversely, inhibition of miR-34a rescued RCAN1 expression and reduced NFATC1 and NF-κB expression in HUVECs. CONCLUSIONS We demonstrated a remarkable role of miR-34a as a regulator of NFATC1-associated placental inflammation through direct targeting of RCAN1. MiR-34a could serve as a novel therapeutic target for limiting the progression of placental inflammation after fetal cardiac bypass.
Collapse
Affiliation(s)
- Hai-Yun Yuan
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Department of Maternal Fetal Medicine and Fetal Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Cheng-Bin Zhou
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Department of Maternal Fetal Medicine and Fetal Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Ji-Mei Chen
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Xiao-Bing Liu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shu-Sheng Wen
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Gang Xu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jian Zhuang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China.
| |
Collapse
|
209
|
Abstract
PURPOSE OF REVIEW Noncoding RNAs regulate many aspects of cardiovascular biology and are potential therapy targets. In this review, we summarize and highlight current discoveries in the field of microRNAs, a class of noncoding RNAs. RECENT FINDINGS miRNAs regulate posttranscriptional gene expression and have been shown to control cardiac development, hypertrophy, fibrosis, and regeneration. Of note are the miRNAs that regulate cardiac contractility (for example, miR-25 and miR-22), cardiac regeneration (like miR-302-367 and miR99/100 families), and fibrosis (as miR-125b). Consistently with these roles of miRNAs, pharmacological intervention using anti-miRNA oligonucleotides (antagomirs or LNA-anti-miRs) has been shown to improve cardiac contractility and mitigate fibrosis, alleviating cardiac dysfunction in the setting of heart failure. SUMMARY miRNAs are crucial regulators of cardiac phenotype and have enthused both basic scientists and clinicians alike. With advancement of technology and better understanding of mechanisms governing miRNA deregulation, we are at the crossroads for deciphering miRNA function and modulating it for therapeutics.
Collapse
|
210
|
Chun HJ, Bonnet S, Chan SY. Translational Advances in the Field of Pulmonary Hypertension. Translating MicroRNA Biology in Pulmonary Hypertension. It Will Take More Than "miR" Words. Am J Respir Crit Care Med 2017; 195:167-178. [PMID: 27648944 PMCID: PMC5394787 DOI: 10.1164/rccm.201604-0886pp] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/10/2016] [Indexed: 12/17/2022] Open
Affiliation(s)
- Hyung J. Chun
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, University of Laval, Quebec City, Quebec, Canada; and
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
211
|
Zhao L, Luo H, Li X, Li T, He J, Qi Q, Liu Y, Yu Z. Exosomes Derived from Human Pulmonary Artery Endothelial Cells Shift the Balance between Proliferation and Apoptosis of Smooth Muscle Cells. Cardiology 2017; 137:43-53. [DOI: 10.1159/000453544] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/16/2016] [Indexed: 11/19/2022]
Abstract
Background: The overproliferation of pulmonary vascular cells is noted in pulmonary hypertension. The role of exosomes from pulmonary artery endothelial cells (PAEC) in the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMC) remains unclear. Methods: Exosomes were isolated and purified from the culture medium of PAEC using a commercial kit. Lipopolysaccharide (LPS), hypoxia, and hydrogen peroxide were utilized to induce PAEC injury. Coculture of PAEC and PASMC was conducted using Transwell plates, and GW4869 was applied to inhibit exosome release. The proliferation and apoptosis level of PASMC was assayed by MTT assay, apoptosis staining, and cleaved caspase-3 immunoblotting. Plasma exosomes were isolated by differential ultracentrifugation. Results: LPS or hypoxia enhance exosome release from PAEC. Release of PAEC-derived exosomes positively correlates with LPS concentration. The coculture of LPS-disposed PAEC with PASMC leads to overproliferation and apoptosis resistance in PASMC, and the exosome inhibitor GW4869 can partly cancel out this effect. Exosomes derived from PAEC could be internalized into PASMC, and thus promote proliferation and induce apoptosis resistance in PASMC. Idiopathic pulmonary arterial hypertension patients exhibit a higher circulation level of endothelium-derived exosomes. Conclusions: Inflammation and hypoxia could induce PAEC to release exosomes. PAEC- derived exosomes are involved in overproliferation and apoptosis resistance in PASMC, by which they may contribute to the pathogenesis of pulmonary hypertension.
Collapse
|
212
|
Brock M, Schuoler C, Leuenberger C, Bühlmann C, Haider TJ, Vogel J, Ulrich S, Gassmann M, Kohler M, Huber LC. Analysis of hypoxia-induced noncoding RNAs reveals metastasis-associated lung adenocarcinoma transcript 1 as an important regulator of vascular smooth muscle cell proliferation. Exp Biol Med (Maywood) 2017; 242:487-496. [PMID: 28056547 DOI: 10.1177/1535370216685434] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vascular remodeling, a pathogenic hallmark in pulmonary hypertension, is mainly driven by a dysbalance between proliferation and apoptosis of human pulmonary artery smooth muscle cells. It has previously been shown that microRNAs are involved in the pathogenesis of pulmonary hypertension. However, the role of long noncoding RNAs has not been evaluated. long noncoding RNA expression was quantified in human pulmonary artery smooth muscle cells using PCR arrays and quantitative PCR. Knockdown of genes was performed by transfection of siRNA or GapmeR. Proliferation and migration were measured using BrdU incorporation and wound healing assays. The mouse model of hypoxia-induced PH was used to determine the physiological meaning of identified long noncoding RNAs. The expression of 84 selected long noncoding RNAs was assessed in hypoxic human pulmonary artery smooth muscle cells and the levels of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) were significantly increased. Depletion of hypoxia-inducible factor 1α abolished the hypoxia-induced upregulation of metastasis-associated lung adenocarcinoma transcript 1 expression. Silencing of MALAT1 significantly decreased proliferation and migration of human pulmonary artery smooth muscle cells. In vivo, MALAT1 expression was significantly increased in lungs of hypoxic mice. Of note, targeting of MALAT1 by GapmeR ameliorated heart hypertrophy in mice with pulmonary hypertension. This is the first report on functional characterization of MALAT1 in the pulmonary vasculature. Our data provide evidence that MALAT1 expression is significantly increased by hypoxia, probably by hypoxia-inducible factor 1α. Intervention experiments confirmed that MALAT1 regulates the proliferative phenotype of smooth muscle cells and silencing of MALAT1 reduced heart hypertrophy in mice with pulmonary hypertension. These data indicate a potential role of MALAT1 in the pathogenesis of pulmonary hypertension. Impact statement Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long noncoding RNA that mediates several biological processes. In the context of vascular biology, MALAT1 has been shown to be inducible by hypoxia and to control cell proliferation. These processes are of major importance for the pathophysiology of hypoxia-induced pulmonary hypertension (PH). Until now, the physiological role of MALAT1 in PH remains unclear. By using smooth muscle cells and by employing an established PH mouse model, we provide evidence that hypoxia induces MALAT1 expression. Moreover, depletion of MALAT1 inhibited migration and proliferation of smooth muscle cells, probably by the induction of cyclin-dependent kinase inhibitors. Of note, MALAT1 was significantly increased in mice exposed to hypoxia and silencing of MALAT1 ameliorated heart hypertrophy in mice with hypoxia-induced PH. Since vascular remodeling and right heart failure as a consequence of pulmonary pressure overload is a major problem in PH, these data have implications for our pathogenetic understanding.
Collapse
Affiliation(s)
- Matthias Brock
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Claudio Schuoler
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland.,2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland
| | - Caroline Leuenberger
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Carlo Bühlmann
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Thomas J Haider
- 2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland.,3 Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich CH-8057, Switzerland
| | - Johannes Vogel
- 2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland.,3 Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich CH-8057, Switzerland
| | - Silvia Ulrich
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Max Gassmann
- 2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland.,3 Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich CH-8057, Switzerland
| | - Malcolm Kohler
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Lars C Huber
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| |
Collapse
|
213
|
Wang C, Zhang C, Liu L, A X, Chen B, Li Y, Du J. Macrophage-Derived mir-155-Containing Exosomes Suppress Fibroblast Proliferation and Promote Fibroblast Inflammation during Cardiac Injury. Mol Ther 2017; 25:192-204. [PMID: 28129114 PMCID: PMC5363311 DOI: 10.1016/j.ymthe.2016.09.001] [Citation(s) in RCA: 271] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022] Open
Abstract
Inflammation plays an important role in cardiac injuries. Here, we examined the role of miRNA in regulating inflammation and cardiac injury during myocardial infarction. We showed that mir-155 expression was increased in the mouse heart after myocardial infarction. Upregulated mir-155 was primarily presented in macrophages and cardiac fibroblasts of injured hearts, while pri-mir-155 was only expressed in macrophages. mir-155 was also presented in exosomes derived from macrophages, and it can be transferred into cardiac fibroblasts by macrophage-derived exosomes. A mir-155 mimic or mir-155 containing exosomes inhibited cardiac fibroblast proliferation by downregulating Son of Sevenless 1 expression and promoted inflammation by decreasing Suppressor of Cytokine Signaling 1 expression. These effects were reversed by the addition of a mir-155 inhibitor. In vivo, mir-155-deficient mice showed a significant reduction of the incidence of cardiac rupture and an improved cardiac function compared with wild-type mice. Moreover, transfusion of wild-type macrophage exosomes to mir-155-/- mice exacerbated cardiac rupture. Finally, the mir-155-deficient mice exhibited elevated fibroblast proliferation and collagen production, along with reduced cardiac inflammation in injured heart. Taken together, our results demonstrate that activated macrophages secrete mir-155-enriched exosomes and identify macrophage-derived mir-155 as a paracrine regulator for fibroblast proliferation and inflammation; thus, a mir-155 inhibitor (i.e., mir-155 antagomir) has the potential to be a therapeutic agent for reducing acute myocardial-infarction-related adverse events.
Collapse
Affiliation(s)
- Chunxiao Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Congcong Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Luxin Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Xi A
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Boya Chen
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China.
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing 100029, China.
| |
Collapse
|
214
|
Xu T, Liu S, Ma T, Jia Z, Zhang Z, Wang A. Aldehyde dehydrogenase 2 protects against oxidative stress associated with pulmonary arterial hypertension. Redox Biol 2016; 11:286-296. [PMID: 28030785 PMCID: PMC5192477 DOI: 10.1016/j.redox.2016.12.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/09/2016] [Accepted: 12/17/2016] [Indexed: 12/18/2022] Open
Abstract
The cardioprotective benefits of aldehyde dehydrogenase 2 (ALDH2) are well established, although the regulatory role of ALDH2 in vascular remodeling in pulmonary arterial hypertension (PAH) is largely unknown. ALDH2 potently regulates the metabolism of aldehydes such as 4-hydroxynonenal (4-HNE), the endogenous product of lipid peroxidation. Thus, we hypothesized that ALDH2 ameliorates the proliferation and migration of human pulmonary artery smooth muscle cells (HPASMCs) by inhibiting 4-HNE accumulation and regulating downstream signaling pathways, thereby ameliorating pulmonary vascular remodeling. We found that low concentrations of 4-HNE (0.1 and 1μM) stimulated cell proliferation by enhancing cyclin D1 and c-Myc expression in primary HPASMCs. Low 4-HNE concentrations also enhanced cell migration by activating the nuclear factor kappa B (NF-κB) signaling pathway, thereby regulating matrix metalloprotein (MMP)-9 and MMP2 expression in vitro. In vivo, Alda-1, an ALDH2 agonist, significantly stimulated ALDH2 activity, reducing elevated 4-HNE and malondialdehyde levels and right ventricular systolic pressure in a monocrotaline-induced PAH animal model to the level of control animals. Our findings indicate that 4-HNE plays an important role in the abnormal proliferation and migration of HPASMCs, and that ALDH2 activation can attenuate 4-HNE-induced PASMC proliferation and migration, possibly by regulating NF-κB activation, in turn ameliorating vascular remodeling in PAH. This mechanism might reflect a new molecular target for treating PAH.
Collapse
Affiliation(s)
- Tao Xu
- Life Science Institute, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China.
| | - Shuangyue Liu
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China
| | - Tingting Ma
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China
| | - Ziyi Jia
- College of Economics and Management, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Zhifei Zhang
- Department of Physiology and Pathophysiology, Capital Medical University, School of Basic Medical Sciences, Beijing 100069, PR China
| | - Aimei Wang
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China.
| |
Collapse
|
215
|
TGF-β signaling controls FSHR signaling-reduced ovarian granulosa cell apoptosis through the SMAD4/miR-143 axis. Cell Death Dis 2016; 7:e2476. [PMID: 27882941 PMCID: PMC5260897 DOI: 10.1038/cddis.2016.379] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/09/2016] [Accepted: 10/17/2016] [Indexed: 01/13/2023]
Abstract
Follicle-stimulating hormone receptor (FSHR) and its intracellular signaling control mammalian follicular development and female infertility. Our previous study showed that FSHR is downregulated during follicular atresia of porcine ovaries. However, its role and regulation in follicular atresia remain unclear. Here, we showed that FSHR knockdown induced porcine granulosa cell (pGC) apoptosis and follicular atresia, and attenuated the levels of intracellular signaling molecules such as PKA, AKT and p-AKT. FSHR was identified as a target of miR-143, a microRNA that was upregulated during porcine follicular atresia. miR-143 enhanced pGC apoptosis by targeting FSHR, and reduced the levels of intracellular signaling molecules. SMAD4, the final molecule in transforming growth factor (TGF)-β signaling, bound to the promoter and induced significant downregulation of miR-143 in vitro and in vivo. Activated TGF-β signaling rescued miR-143-reduced FSHR and intracellular signaling molecules, and miR-143-induced pGC apoptosis. Overall, our findings offer evidence to explain how TGF-β signaling influences and FSHR signaling for regulation of pGC apoptosis and follicular atresia by a specific microRNA, miR-143.
Collapse
|
216
|
Singh J, Boopathi E, Addya S, Phillips B, Rigoutsos I, Penn RB, Rattan S. Aging-associated changes in microRNA expression profile of internal anal sphincter smooth muscle: Role of microRNA-133a. Am J Physiol Gastrointest Liver Physiol 2016; 311:G964-G973. [PMID: 27634012 PMCID: PMC5130548 DOI: 10.1152/ajpgi.00290.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/13/2016] [Indexed: 01/31/2023]
Abstract
A comprehensive genomic and proteomic, computational, and physiological approach was employed to examine the (previously unexplored) role of microRNAs (miRNAs) as regulators of internal anal sphincter (IAS) smooth muscle contractile phenotype and basal tone. miRNA profiling, genome-wide expression, validation, and network analyses were employed to assess changes in mRNA and miRNA expression in IAS smooth muscles from young vs. aging rats. Multiple miRNAs, including rno-miR-1, rno-miR-340-5p, rno-miR-185, rno-miR-199a-3p, rno-miR-200c, rno-miR-200b, rno-miR-31, rno-miR-133a, and rno-miR-206, were found to be upregulated in aging IAS. qPCR confirmed the upregulated expression of these miRNAs and downregulation of multiple, predicted targets (Eln, Col3a1, Col1a1, Zeb2, Myocd, Srf, Smad1, Smad2, Rhoa/Rock2, Fn1, Tagln v2, Klf4, and Acta2) involved in regulation of smooth muscle contractility. Subsequent studies demonstrated an aging-associated increase in the expression of miR-133a, corresponding decreases in RhoA, ROCK2, MYOCD, SRF, and SM22α protein expression, RhoA-signaling, and a decrease in basal and agonist [U-46619 (thromboxane A2 analog)]-induced increase in the IAS tone. Moreover, in vitro transfection of miR-133a caused a dose-dependent increase of IAS tone in strips, which was reversed by anti-miR-133a. Last, in vivo perianal injection of anti-miR-133a reversed the loss of IAS tone associated with age. This work establishes the important regulatory effect of miRNA-133a on basal and agonist-stimulated IAS tone. Moreover, reversal of age-associated loss of tone via anti-miR delivery strongly implicates miR dysregulation as a causal factor in the aging-associated decrease in IAS tone and suggests that miR-133a is a feasible therapeutic target in aging-associated rectoanal incontinence.
Collapse
Affiliation(s)
- Jagmohan Singh
- 1Department of Medicine, Division of Gastroenterology & Hepatology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania;
| | - Ettickan Boopathi
- 2Center for Translational Medicine, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania;
| | - Sankar Addya
- 3Kimmel Cancer Center, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania;
| | - Benjamin Phillips
- 4Department of Surgery, Division of Colorectal Surgery, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Isidore Rigoutsos
- 5Computational Medicine Center, Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Raymond B. Penn
- 2Center for Translational Medicine, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania;
| | - Satish Rattan
- 1Department of Medicine, Division of Gastroenterology & Hepatology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania;
| |
Collapse
|
217
|
Li J, Qu H, Jiang H, Zhao Z, Zhang Q. Transcriptome-Wide Comparative Analysis of microRNA Profiles in the Telogen Skins of Liaoning Cashmere Goats (Capra hircus) and Fine-Wool Sheep (Ovis aries) by Solexa Deep Sequencing. DNA Cell Biol 2016; 35:696-705. [PMID: 27754706 DOI: 10.1089/dna.2015.3161] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Compare the microRNA (miRNA) trancriptomes of goat and sheep skin using Solexa sequencing to understand the development of skin and hair follicles (HFs). miRNA expression patterns vary in the two small RNA libraries from goat (G library) and sheep (S library) telogen skin samples. Analysis of the size distribution of 25.32 million clean reads revealed that most are 21-23 nucleotides. A total of 1910 known miRNAs and 2261 novel mature miRNAs were identified in this study. Among them, 107 novel miRNAs and 1246 known miRNAs were differentially expressed in the two libraries; 10 of the known miRNAs were identified using stem-loop quantitative real-time PCR. Furthermore, GO and KEGG pathway analysis of predicted miRNA targets illustrated the roles of these differentially expressed miRNAs in telogen HF development and growth. This study provides important information about the role of miRNAs in the regulation of HF development and their function in the telogen phase. This observation may help future investigations of the regulation of miRNAs during wool quality improvement.
Collapse
Affiliation(s)
- Jianping Li
- 1 College of Veterinary Medicine, Jilin University, Changchun, China .,2 Henan University of Animal Husbandry and Economy, Henan, Zhengzhou, China
| | - HaiE Qu
- 1 College of Veterinary Medicine, Jilin University, Changchun, China
| | - Huaizhi Jiang
- 3 College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zhihui Zhao
- 4 College of Animal Science and Technology, Jilin University, Changchun, China
| | - Qiaoling Zhang
- 1 College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
218
|
Abstract
PURPOSE OF REVIEW Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are becoming fundamentally important in the pathophysiology relating to injury-induced vascular remodelling. We highlight recent studies that demonstrate the involvement of ncRNAs in vein graft disease, in in-stent restenosis and in pulmonary arterial hypertension, with a particular focus on endothelial cell and vascular smooth muscle cell function. We also briefly discuss the emerging role of exosomal-derived ncRNAs and how this mechanism impacts on vascular function. RECENT FINDINGS ncRNAs have been described as novel regulators in the pathophysiology of vascular injury, inflammation, and vessel wall remodelling. In particular, several studies have demonstrated that manipulation of miRNAs can reduce the burden of pathological vascular remodelling. Such studies have also shown that exosomal miRNA-mediated, cell-to-cell communication between endothelial cells and vascular smooth muscle cells is critical in the disease process. In addition to miRNAs, lncRNAs are emerging as regulators of vascular function in health and disease. Although lncRNAs are complex in both their sheer numbers and mechanisms of action, identifying their contribution to vascular disease is essential. SUMMARY Given the important roles of ncRNAs in vascular injury and remodelling together will their capacity for cell-to-cell communication, manipulating ncRNA might provide novel therapeutic interventions.
Collapse
Affiliation(s)
- Lin Deng
- aBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow bCentre for Cardiovascular Science, Queen's Medical Research Institute, BHF/University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
219
|
Gao Y, Chen T, Raj JU. Endothelial and Smooth Muscle Cell Interactions in the Pathobiology of Pulmonary Hypertension. Am J Respir Cell Mol Biol 2016; 54:451-60. [PMID: 26744837 DOI: 10.1165/rcmb.2015-0323tr] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the pulmonary vasculature, the endothelial and smooth muscle cells are two key cell types that play a major role in the pathobiology of pulmonary vascular disease and pulmonary hypertension. The normal interactions between these two cell types are important for the homeostasis of the pulmonary circulation, and any aberrant interaction between them may lead to various disease states including pulmonary vascular remodeling and pulmonary hypertension. It is well recognized that the endothelial cell can regulate the function of the underlying smooth muscle cell by releasing various bioactive agents such as nitric oxide and endothelin-1. In addition to such paracrine regulation, other mechanisms exist by which there is cross-talk between these two cell types, including communication via the myoendothelial injunctions and information transfer via extracellular vesicles. Emerging evidence suggests that these nonparacrine mechanisms play an important role in the regulation of pulmonary vascular tone and the determination of cell phenotype and that they are critically involved in the pathobiology of pulmonary hypertension.
Collapse
Affiliation(s)
- Yuansheng Gao
- 1 Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing, China; and
| | - Tianji Chen
- 2 Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - J Usha Raj
- 2 Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
220
|
Protective Roles of Endothelial AMP-Activated Protein Kinase Against Hypoxia-Induced Pulmonary Hypertension in Mice. Circ Res 2016; 119:197-209. [DOI: 10.1161/circresaha.115.308178] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Rationale:
Endothelial AMP-activated protein kinase (AMPK) plays an important role for vascular homeostasis, and its role is impaired by vascular inflammation. However, the role of endothelial AMPK in the pathogenesis of pulmonary arterial hypertension (PAH) remains to be elucidated.
Objective:
To determine the role of endothelial AMPK in the development of PAH.
Methods and Results:
Immunostaining showed that endothelial AMPK is downregulated in the pulmonary arteries of patients with PAH and hypoxia mouse model of pulmonary hypertension (PH). To elucidate the role of endothelial AMPK in PH, we used endothelial-specific AMPK-knockout mice (
eAMPK
–/–
), which were exposed to hypoxia. Under normoxic condition,
eAMPK
–/–
mice showed the normal morphology of pulmonary arteries compared with littermate controls (
eAMPK
flox/flox
). In contrast, development of hypoxia-induced PH was accelerated in
eAMPK
–/–
mice compared with controls. Furthermore, the exacerbation of PH in
eAMPK
–/–
mice was accompanied by reduced endothelial function, upregulation of growth factors, and increased proliferation of pulmonary artery smooth muscle cells. Importantly, conditioned medium from endothelial cells promoted pulmonary artery smooth muscle cell proliferation, which was further enhanced by the treatment with AMPK inhibitor. Serum levels of inflammatory cytokines, including tumor necrosis factor-α and interferon-γ were significantly increased in patients with PAH compared with healthy controls. Consistently, endothelial AMPK and cell proliferation were significantly reduced by the treatment with serum from patients with PAH compared with controls. Importantly, long-term treatment with metformin, an AMPK activator, significantly attenuated hypoxia-induced PH in mice.
Conclusions:
These results indicate that endothelial AMPK is a novel therapeutic target for the treatment of PAH.
Collapse
|
221
|
Wang P, Xu J, Hou Z, Wang F, Song Y, Wang J, Zhu H, Jin H. miRNA-34a promotes proliferation of human pulmonary artery smooth muscle cells by targeting PDGFRA. Cell Prolif 2016; 49:484-93. [PMID: 27302634 DOI: 10.1111/cpr.12265] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/07/2016] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Pulmonary arterial hypertension (PAH) is a fast progressing vascular disease characterized by uncontrolled cell proliferation of pulmonary artery smooth muscle cells (PASMCs). Some studies have suggested that PAH and cancers share an apoptosis-resistant state, featuring excessive cell proliferation. The miR-34 family consists of tumour-suppressive miRNAs, and its reduced expression has been reported in numerous cancers; however, its role in hypoxia-induced PAH has not been previously studied. MATERIALS AND METHODS miR-34 family expression was evaluated in a rat model with hypoxia and in cultured hypoxic PASMCs, using real-time quantitative PCR (RT-qPCR). Function of miR-34 family was assessed by transfecting miR-34 mimics and inhibitors. Dual luciferase reporter gene assays, RT-qPCR and Western blotting were performed to validate target genes of miR-34. RESULTS Significant down-regulation of miR-34a in hypoxic lung tissue, pulmonary artery and PASMCs was identified and then effects of miR-34a in modulating cell proliferation in human pulmonary artery smooth muscle cells (hPASMCs) was investigated in vitro. Reduction of miR-34a levels in hPASMCs caused increased proliferation and these effects were reversed by overexpression of miR-34a. miR-34a overexpression down-regulated platelet-derived growth factor receptor alpha (PDGFRA) expression, which is a key factor in PAH development. These results suggest that miR-34a is a potential regulator of proliferation in PASMCs, and that it could be used as a novel treatment strategy in PAH.
Collapse
Affiliation(s)
- Peng Wang
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China.,Department of Physiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, China
| | - Jie Xu
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Zhiling Hou
- Department of Emergency Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Fangfang Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jiamusi University, Jiamusi, China
| | - Yingli Song
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jiao Wang
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hui Zhu
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hongbo Jin
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
222
|
Ballantyne MD, Pinel K, Dakin R, Vesey AT, Diver L, Mackenzie R, Garcia R, Welsh P, Sattar N, Hamilton G, Joshi N, Dweck MR, Miano JM, McBride MW, Newby DE, McDonald RA, Baker AH. Smooth Muscle Enriched Long Noncoding RNA (SMILR) Regulates Cell Proliferation. Circulation 2016; 133:2050-65. [PMID: 27052414 PMCID: PMC4872641 DOI: 10.1161/circulationaha.115.021019] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 03/28/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells from a contractile to a synthetic state is implicated in diverse vascular pathologies, including atherogenesis, plaque stabilization, and neointimal hyperplasia. However, very little is known about the role of long noncoding RNA (lncRNA) during this process. Here, we investigated a role for lncRNAs in vascular smooth muscle cell biology and pathology. METHODS AND RESULTS Using RNA sequencing, we identified >300 lncRNAs whose expression was altered in human saphenous vein vascular smooth muscle cells following stimulation with interleukin-1α and platelet-derived growth factor. We focused on a novel lncRNA (Ensembl: RP11-94A24.1), which we termed smooth muscle-induced lncRNA enhances replication (SMILR). Following stimulation, SMILR expression was increased in both the nucleus and cytoplasm, and was detected in conditioned media. Furthermore, knockdown of SMILR markedly reduced cell proliferation. Mechanistically, we noted that expression of genes proximal to SMILR was also altered by interleukin-1α/platelet-derived growth factor treatment, and HAS2 expression was reduced by SMILR knockdown. In human samples, we observed increased expression of SMILR in unstable atherosclerotic plaques and detected increased levels in plasma from patients with high plasma C-reactive protein. CONCLUSIONS These results identify SMILR as a driver of vascular smooth muscle cell proliferation and suggest that modulation of SMILR may be a novel therapeutic strategy to reduce vascular pathologies.
Collapse
Affiliation(s)
- Margaret D Ballantyne
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Karine Pinel
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Rachel Dakin
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Alex T Vesey
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Louise Diver
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Ruth Mackenzie
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Raquel Garcia
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Paul Welsh
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Naveed Sattar
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Graham Hamilton
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Nikhil Joshi
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Marc R Dweck
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Joseph M Miano
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Martin W McBride
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - David E Newby
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Robert A McDonald
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Andrew H Baker
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.).
| |
Collapse
|
223
|
Escudero CA, Herlitz K, Troncoso F, Acurio J, Aguayo C, Roberts JM, Truong G, Duncombe G, Rice G, Salomon C. Role of Extracellular Vesicles and microRNAs on Dysfunctional Angiogenesis during Preeclamptic Pregnancies. Front Physiol 2016; 7:98. [PMID: 27047385 PMCID: PMC4796029 DOI: 10.3389/fphys.2016.00098] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/01/2016] [Indexed: 01/08/2023] Open
Abstract
Preeclampsia is a syndrome characterized by hypertension during pregnancy, which is a leading cause of morbidity and mortality in both mother and newborn in developing countries. Some advances have increased the understanding of pathophysiology of this disease. For example, reduced utero-placental blood flow associated with impaired trophoblast invasion may lead to a hypoxic placenta that releases harmful materials into the maternal and feto-placental circulation and impairs endothelial function. Identification of these harmful materials is one of the hot topics in the literature, since these provide potential biomarkers. Certainty, such knowledge will help us to understand the miscommunication between mother and fetus. In this review we highlight how placental extracellular vesicles and their cargo, such as small RNAs (i.e., microRNAs), might be involved in endothelial dysfunction, and then in the angiogenesis process, during preeclampsia. Currently only a few reports have addressed the potential role of endothelial regulatory miRNA in the impaired angiogenesis in preeclampsia. One of the main limitations in this area is the variability of the analyses performed in the current literature. This includes variability in the size of the particles analyzed, and broad variation in the exosomes considered. The quantity of microRNA targets genes suggest that practically all endothelial cell metabolic functions might be impaired. More studies are required to investigate mechanisms underlying miRNA released from placenta upon endothelial function involved in the angiogenenic process.
Collapse
Affiliation(s)
- Carlos A Escudero
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Universidad del Bío-BíoChillán, Chile; Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Universidad del Bío-BíoChillán, Chile
| | - Kurt Herlitz
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Universidad del Bío-Bío Chillán, Chile
| | - Felipe Troncoso
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Universidad del Bío-Bío Chillán, Chile
| | - Jesenia Acurio
- Group of Investigation in Tumor Angiogenesis, Vascular Physiology Laboratory, Universidad del Bío-Bío Chillán, Chile
| | - Claudio Aguayo
- Group of Research and Innovation in Vascular Health, Department of Basic Sciences, Universidad del Bío-BíoChillán, Chile; Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of ConcepciónConcepción, Chile
| | - James M Roberts
- Departments of Obstetrics, Gynecology and Reproductive Sciences, Epidemiology, and the Clinical and Translational Science Institute, Magee-Womens Research Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Grace Truong
- Exosome Biology Laboratory, Faculty of Medicine and Biomedical Sciences, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, The University of Queensland Brisbane, QLD, Australia
| | - Gregory Duncombe
- Exosome Biology Laboratory, Faculty of Medicine and Biomedical Sciences, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, The University of Queensland Brisbane, QLD, Australia
| | - Gregory Rice
- Exosome Biology Laboratory, Faculty of Medicine and Biomedical Sciences, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, The University of QueenslandBrisbane, QLD, Australia; Ochsner Clinic Foundation, Maternal-Fetal Medicine, Department of Obstetrics and GynecologyNew Orleans, LA, USA
| | - Carlos Salomon
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of ConcepciónConcepción, Chile; Exosome Biology Laboratory, Faculty of Medicine and Biomedical Sciences, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, The University of QueenslandBrisbane, QLD, Australia; Ochsner Clinic Foundation, Maternal-Fetal Medicine, Department of Obstetrics and GynecologyNew Orleans, LA, USA
| |
Collapse
|
224
|
Kapustin AN, Shanahan CM. Emerging roles for vascular smooth muscle cell exosomes in calcification and coagulation. J Physiol 2016; 594:2905-14. [PMID: 26864864 DOI: 10.1113/jp271340] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/25/2015] [Indexed: 12/26/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) phenotypic conversion from a contractile to 'synthetic' state contributes to vascular pathologies including restenosis, atherosclerosis and vascular calcification. We have recently found that the secretion of exosomes is a feature of 'synthetic' VSMCs and that exosomes are novel players in vascular repair processes as well as pathological vascular thrombosis and calcification. Pro-inflammatory cytokines and growth factors as well as mineral imbalance stimulate exosome secretion by VSMCs, most likely by the activation of sphingomyelin phosphodiesterase 3 (SMPD3) and cytoskeletal remodelling. Calcium stress induces dramatic changes in VSMC exosome composition and accumulation of phosphatidylserine (PS), annexin A6 and matrix metalloproteinase-2, which converts exosomes into a nidus for calcification. In addition, by presenting PS, VSMC exosomes can also provide the catalytic surface for the activation of coagulation factors. Recent data showing that VSMC exosomes are loaded with proteins and miRNA regulating cell adhesion and migration highlight VSMC exosomes as potentially important communication messengers in vascular repair. Thus, the identification of signalling pathways regulating VSMC exosome secretion, including activation of SMPD3 and cytoskeletal rearrangements, opens up novel avenues for a deeper understanding of vascular remodelling processes.
Collapse
Affiliation(s)
- A N Kapustin
- BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - C M Shanahan
- BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
225
|
Glucocorticoid-induced fetal origins of adult hypertension: Association with epigenetic events. Vascul Pharmacol 2016; 82:41-50. [PMID: 26903240 DOI: 10.1016/j.vph.2016.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 02/05/2023]
Abstract
Hypertension is a predominant risk factor for cardiovascular diseases and a major health care burden. Accumulating epidemiological and experimental evidence suggest that adult-onset hypertension may have its origins during early development. Upon exposure to glucocorticoids, the fetus develops hypertension, and the offspring may be programmed to continue the hypertensive trajectory into adulthood. Elevated oxidative stress and deranged nitric oxide system are not only hallmarks of adult hypertension but are also observed earlier in life. Endothelial dysfunction and remodeling of the vasculature, which are robustly associated with increased incidence of hypertension, are likely to have been pre-programmed during fetal life. Apparently, genomic, non-genomic, and epigenomic factors play a significant role in the development of hypertension, including glucocorticoid-driven effects on blood pressure. In this review, we discuss the involvement of the aforementioned participants in the pathophysiology of hypertension and suggest therapeutic opportunities for targeting epigenome modifiers, potentially for personalized medicine.
Collapse
|
226
|
Caporali A, Miscianinov V, Saif J, Emanueli C. MicroRNA transport in cardiovascular complication of diabetes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2111-2120. [PMID: 26806392 DOI: 10.1016/j.bbalip.2016.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/17/2016] [Accepted: 01/18/2016] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) are post-transcriptional inhibitory regulators of gene expression by binding to complementary messenger RNA (mRNA) transcripts. Extracellular miRNAs are transported by membrane-derived vesicles (exosomes and microparticles), lipoproteins, and other ribonucleoprotein complexes. Extracellular microRNAs are emerging as important mediators of intercellular communications, being involved in the transmission of biological signals between cells. Several miRNAs have been identified as having a primary impact on many biological processes that are of direct relevance to cardiovascular complications of diabetes. Whether the extracellular miRNAs are directly involved in the regulation of these processes is yet to be established. Here, we review recent progresses in extracellular miRNA biology and the role of extracellular miRNA in diabetes induced cardiovascular disease, describing the regulators affecting miRNA transport and the mechanisms for different miRNA transporters. In addition, we discuss the advancement of the research in this field and identify the associated challenges. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Andrea Caporali
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Vladislav Miscianinov
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Jaimy Saif
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Costanza Emanueli
- Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
227
|
Rajput C, Tauseef M, Farazuddin M, Yazbeck P, Amin MR, Avin Br V, Sharma T, Mehta D. MicroRNA-150 Suppression of Angiopoetin-2 Generation and Signaling Is Crucial for Resolving Vascular Injury. Arterioscler Thromb Vasc Biol 2016; 36:380-8. [PMID: 26743170 DOI: 10.1161/atvbaha.115.306997] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/17/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Increased vascular permeability is a hallmark of sepsis and acute respiratory distress syndrome. Angiopoietin (Ang2) induces vascular leak, and excess Ang2 generation is associated with patient mortality from these diseases. However, mechanisms dampening Ang2 generation during injury remain unclear. Interestingly, microRNA (miR)-150 levels were decreased in septic patients. miR regulate signaling networks by silencing mRNAs containing complementary sequences. Thus, we hypothesized that miR-150 suppresses Ang2 generation and thereby resolves vascular injury. APPROACH AND RESULTS Wild-type or miR-150(-/-) mice or endothelial cells were exposed to lipopolysaccharide or sepsis, and Ang2 levels, adherens junction reannealing, endothelial barrier function, and mortality were determined. Although Ang2 transiently increased during lipopolysaccharide-induced injury in wild-type endothelial cells and lungs, miR-150 expression was elevated only during recovery from injury. Deletion of miR-150 caused a persistent increase in Ang2 levels and impaired adherens junctions reannealing after injury, resulting thereby in an irreversible increase in vascular permeability. Also, miR-150(-/-) mice died rapidly after sepsis. Rescuing miR-150 expression in endothelial cells prevented Ang2 generation, thereby restoring vascular barrier function in miR-150(-/-) mice. miR-150 terminated Ang2 generation by targeting the transcription factor, early growth response 2. Thus, early growth response 2 or Ang2 depletion in miR-150(-/-) endothelial cells restored junctional reannealing and reinstated barrier function. Importantly, upregulating miR-150 expression by injecting a chemically synthesized miR-150 mimic into wild-type mice vasculature decreased early growth response 2 and Ang2 levels and hence mortality from sepsis. CONCLUSIONS miR-150 is a novel suppressor of Ang2 generation with a key role in resolving vascular injury and reducing mortality resulting from sepsis.
Collapse
Affiliation(s)
- Charu Rajput
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Mohammad Tauseef
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Mohammad Farazuddin
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Pascal Yazbeck
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Md-Ruhul Amin
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Vijay Avin Br
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Tiffany Sharma
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago
| | - Dolly Mehta
- From the Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago.
| |
Collapse
|