1
|
Luo Y, Li M, You J, Jiang J, Zeng M, Luo M. Regulation of vascular smooth muscle cells phenotype by metformin up-regulated miR-1/ CCND1 axis via targeting AMPK/TGF-β signaling pathway. Mol Biol Rep 2025; 52:437. [PMID: 40299098 DOI: 10.1007/s11033-025-10532-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
The phenotypic switch of vascular smooth muscle cells (VSMCs), characterized by the tissue-specific expression of certain microRNAs (miRNAs), is a critical factor in the development of diabetic vascular diseases. Metformin, a widely prescribed anti-diabetic medication for type 2 diabetes treatment, activates the adenosine monophosphate-activated protein kinase (AMPK) pathway and exerts a protective effect on vascular endothelium. Although the regulatory effects of metformin on the switch of the vascular smooth muscle cell phenotype have been identified, the specific role of miRNAs in this process remains unclear. We identified a specific miR-1 in response to metformin treatment and determined its effects on both miR-1 and its targets. Subsequently, we investigated the influence of these factors on the metformin-induced phenotype switch in vascular smooth muscle cells, specifically focusing on proliferation and migration, as well as activation of the AMPK/Transforming Growth Factor (TGF-β) axis. This was achieved using various methodologies, including bioinformatics analysis, quantitative real-time polymerase chain reaction (qRT-PCR), Western blot analysis, wound scratch assays, and Cell Counting Kit-8 assays. Our findings showed that metformin upregulated miR-1, which directly targets cyclin D1 (CCND1) in VSMCs. Metformin was observed to enhance the expression of contractile phenotype proteins, including α-smooth muscle actin (α-SMA) and smooth muscle myosin heavy chain (SMMHC), while simultaneously reducing the expression of proliferative phenotype proteins such as CCND1 and proliferating cell nuclear antigen (PCNA). The inhibition of miR-1 was found to reverse the effects of metformin on the phenotypic switch of VSMCs. This occurs partly through the AMPK/TGF-β signaling pathway and inhibits the migration and proliferation of VSMCs.
Collapse
Affiliation(s)
- Yulin Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Clinical Trial Research Center, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mengting Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Clinical Trial Research Center, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jingcan You
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Clinical Trial Research Center, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Min Zeng
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Clinical Trial Research Center, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China.
| |
Collapse
|
2
|
Uysal O, Erybeh H, Canbek M, Ekenel EQ, Gunes S, Büyükköroğlu G, Semerci Sevimli T, Cemrek F, Sariboyaci AE. Stem Cell-Based or Cell-Free Gene Therapy in Chondrocyte Regeneration: Synovial Fluid-Derived Mesenchymal Stem Cell Exosomes. Curr Mol Med 2024; 24:906-919. [PMID: 37859306 PMCID: PMC11327740 DOI: 10.2174/0115665240266016231014081916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Cartilage injuries are currently the most prevalent joint disease. Previous studies have emphasized the use of stem cells as the effective treatment for regenerating cartilage damage. OBJECTIVE In this study, considering the difficulties of the cellular therapy method, it was hypothesized that human synovial fluid-derived mesenchymal stem cell (hSFMSC) exosomes as a SC source could be used to treat these injuries as a safer and cell-free therapeutic alternative procedure due to its direct relevance to cartilage regeneration. Moreover, this study aimed to determine the miRNA and target genes required for the formation of SC treatment combined with gene therapy in order to reveal the mechanism of cartilage regeneration and increase its effectiveness. METHODS MSCs were characterized by flow cytometry, and immunocytochemical and differentiation analyses were done. To characterize functionally isolated exosomes, in vitro uptake analysis was performed. RT-qPCR was used to examine in terms of the advantages of cellular and cell-free therapy, mature human chondroblasts derived by differentiation from hSF-MSCs and human chondrocyte profiles were compared in order to demonstrate the above profile of hSF-MSCs and exosomes isolated from them, and the effectiveness of SC therapy in repairing cartilage damage. RESULTS According to our findings, the expression level of hsa-miR-155-5p was found to be considerably higher in chondrocytes differentiated from human synovial fluid MSCs than in mature human chondrocytes. These findings were also supported by the TGF-signalling pathway and chondrogenesis marker genes. CONCLUSION It was concluded that hSF-MSCs and exosomes can be used in the treatment of cartilage damage, and hsa-miR-155-5p can be used as a target miRNA in a new gene therapy approach because it increases the therapeutic effect on cartilage damage.
Collapse
Affiliation(s)
- Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| | - Haya Erybeh
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Molecular Biology, Science Faculty, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Mediha Canbek
- Department of Molecular Biology, Science Faculty, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Emilia Qomi Ekenel
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Sibel Gunes
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| | - Gülay Büyükköroğlu
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkiye
| | - Tugba Semerci Sevimli
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Fatih Cemrek
- Department of Statistics, Faculty of Science and Letters, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Ayla Eker Sariboyaci
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| |
Collapse
|
3
|
Fang T, Wang X, Huangfu N. Superfamily II helicases: the potential therapeutic target for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1309491. [PMID: 38152606 PMCID: PMC10752008 DOI: 10.3389/fcvm.2023.1309491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) still maintain high morbidity and mortality globally. Helicases, a unique class of enzymes, are extensively implicated in the processes of nucleic acid (NA) metabolism across various organisms. They play a pivotal role in gene expression, inflammatory response, lipid metabolism, and so forth. However, abnormal helicase expression has been associated with immune response, cancer, and intellectual disability in humans. Superfamily II (SFII) is one of the largest and most diverse of the helicase superfamilies. Increasing evidence has implicated SFⅡ helicases in the pathogenesis of multiple CVDs. In this review, we comprehensively review the regulation mechanism of SFⅡ helicases in CVDs including atherosclerosis, myocardial infarction, cardiomyopathies, and heart failure, which will contribute to the investigation of ideal therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Tianxiang Fang
- Health Science Center, Ningbo University, Ningbo, China
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Cardiology, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Clinical Medicine Research Centre for Cardiovascular Disease of Ningbo, Ningbo, China
| | - Xizhi Wang
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Ning Huangfu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Cardiology, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Clinical Medicine Research Centre for Cardiovascular Disease of Ningbo, Ningbo, China
| |
Collapse
|
4
|
Wang WX, Springer JE, Hatton KW. MicroRNAs as Biomarkers for Predicting Complications following Aneurysmal Subarachnoid Hemorrhage. Int J Mol Sci 2021; 22:ijms22179492. [PMID: 34502401 PMCID: PMC8431281 DOI: 10.3390/ijms22179492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 12/14/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a high mortality hemorrhagic stroke that affects nearly 30,000 patients annually in the United States. Approximately 30% of aSAH patients die during initial hospitalization and those who survive often carry poor prognosis with one in five having permanent physical and/or cognitive disabilities. The poor outcome of aSAH can be the result of the initial catastrophic event or due to the many acute or delayed neurological complications, such as cerebral ischemia, hydrocephalus, and re-bleeding. Unfortunately, no effective biomarker exists to predict or diagnose these complications at a clinically relevant time point when neurologic injury can be effectively treated and managed. Recently, a number of studies have demonstrated that microRNAs (miRNAs) in extracellular biofluids are highly associated with aSAH and complications. Here we provide an overview of the current research on relevant human studies examining the correlation between miRNAs and aSAH complications and discuss the potential application of using miRNAs as biomarkers in aSAH management.
Collapse
Affiliation(s)
- Wang-Xia Wang
- Sanders-Brown Center on Aging, Spinal Cord and Brain Injury Research Center, and the Pathology & Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-859-218-3886
| | - Joe E. Springer
- Spinal Cord and Brain Injury Research Center, and the Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA;
| | - Kevin W. Hatton
- Department of Anesthesiology Critical Care Medicine, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
5
|
Ghasemi M, Rezaei M, Yazdi A, Keikha N, Maruei-Milan R, Asadi-Tarani M, Salimi S. The effects of DICER1 and DROSHA polymorphisms on susceptibility to recurrent spontaneous abortion. J Clin Lab Anal 2019; 34:e23079. [PMID: 31659796 PMCID: PMC7083489 DOI: 10.1002/jcla.23079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 01/08/2023] Open
Abstract
Background Recurrent spontaneous abortion (RSA) is a serious problem in pregnancy. The exact etiology of RSA is unknown in more than 50% of all the patients. However, genetic variations are known as susceptibility factors for idiopathic RSA. Considering the role of miRNA biosynthesis machinery in the miRNA production and effect of miRNAs on various diseases, this study aimed to evaluate the effects of DICER1 rs3742330 and DROSHA rs6877842 polymorphisms on RSA risk. Methods In this case‐control study, 150 RSA patients and 195 age‐matched healthy female controls were recruited. Both polymorphisms were genotyped using PCR‐RFLP method. Results The frequency of DICER1 rs3742330AG genotype was higher in the control group (P = .022). There was a statistically significant association between rs3742330 polymorphism and a reduced RSA risk in dominant and allelic models (P = .013 and P = .007, respectively). No statistically significant association was found between DROSHA rs6877842 variant and RSA risk. The combination of AG and GC genotypes and G‐G alleles of DICER1 rs3742330 and DROSHA rs6877842 polymorphisms led to a decreased RSA risk. However, the synergic effect of rs3742330A and rs6877842G alleles (A‐G) and AA‐GG genotypes was associated with an increased RSA risk. Conclusion the DICER1 rs3742330AG genotype and combination of AG and GC genotypes and G‐G alleles of DICER1 rs3742330 and DROSHA rs6877842 polymorphisms were associated with a reduced RSA risk.
Collapse
Affiliation(s)
- Marzieh Ghasemi
- Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.,Moloud Infertility Center, Ali ibn Abitaleb Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahnaz Rezaei
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Atefeh Yazdi
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Narjes Keikha
- Moloud Infertility Center, Ali ibn Abitaleb Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Rostam Maruei-Milan
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mina Asadi-Tarani
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeedeh Salimi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
6
|
Leeper NJ, Maegdefessel L. Non-coding RNAs: key regulators of smooth muscle cell fate in vascular disease. Cardiovasc Res 2019; 114:611-621. [PMID: 29300828 DOI: 10.1093/cvr/cvx249] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/28/2017] [Indexed: 01/02/2023] Open
Abstract
The vascular smooth muscle cell (SMC) is one of the most plastic cells in the body. Understanding how non-coding RNAs (ncRNAs) regulate SMC cell-fate decision making in the vasculature has significantly enhanced our understanding of disease development, and opened up exciting new avenues for potential therapeutic applications. Recent studies on SMC physiology have in addition challenged our traditional view on their role and contribution to vascular disease, mainly in the setting of atherosclerosis as well as aneurysm disease, and restenosis after angioplasties. The impact of SMC behaviour on vascular disease is now recognized to be context dependent; SMC proliferation and migration can be harmful or beneficial, whereas their apoptosis, senescence, and switching into a more macrophage-like phenotype can promote inflammation and disease progression. This is in particular true for atherosclerosis-related diseases, where proliferation of SMCs was believed to promote lesion formation, but may also prevent plaque rupture by stabilizing the fibrous cap. Based on newer findings of genetic lineage tracing studies, it was revealed that SMC phenotypic switching can result in less-differentiated forms that lack classical SMC markers while exhibiting functions more related to macrophage-like cells. This switching can directly promote atherogenesis. The aim of this current review is to summarize and discuss how ncRNAs (mainly microRNAs and long ncRNAs) are involved in SMC plasticity, and how they directly affect vascular disease development and progression. Finally, we want to critically assess where potential future therapies could be useful to influence the burden of vascular diseases.
Collapse
Affiliation(s)
- Nicholas J Leeper
- Division of Vascular Surgery, Stanford University, Stanford, CA, USA
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, and German Center for Cardiovascular Research Center (DZHK) Partner Site Munich, 81675 Munich, Germany.,Karolinska Institute, Center for Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
7
|
Lu Y, Thavarajah T, Gu W, Cai J, Xu Q. Impact of miRNA in Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 38:e159-e170. [PMID: 30354259 DOI: 10.1161/atvbaha.118.310227] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yao Lu
- From the Center of Clinical Pharmacology (Y.L.)
| | - Tanuja Thavarajah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Jingjing Cai
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingbo Xu
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| |
Collapse
|
8
|
Guo WT, Wang Y. Dgcr8 knockout approaches to understand microRNA functions in vitro and in vivo. Cell Mol Life Sci 2019; 76:1697-1711. [PMID: 30694346 PMCID: PMC11105204 DOI: 10.1007/s00018-019-03020-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/11/2019] [Accepted: 01/17/2019] [Indexed: 01/07/2023]
Abstract
Biologic function of the majority of microRNAs (miRNAs) is still unknown. Uncovering the function of miRNAs is hurdled by redundancy among different miRNAs. The deletion of Dgcr8 leads to the deficiency in producing all canonical miRNAs, therefore, overcoming the redundancy issue. Dgcr8 knockout strategy has been instrumental in understanding the function of miRNAs in a variety of cells in vitro and in vivo. In this review, we will first give a brief introduction about miRNAs, miRNA biogenesis pathway and the role of Dgcr8 in miRNA biogenesis. We will then summarize studies performed with Dgcr8 knockout cell models with a focus on embryonic stem cells. After that, we will summarize results from various in vivo Dgcr8 knockout models. Given significant phenotypic differences in various tissues between Dgcr8 and Dicer knockout, we will also briefly review current progresses on understanding miRNA-independent functions of miRNA biogenesis factors. Finally, we will discuss the potential use of a new strategy to stably express miRNAs in Dgcr8 knockout cells. In future, Dgcr8 knockout approaches coupled with innovations in miRNA rescue strategy may provide further insights into miRNA functions in vitro and in vivo.
Collapse
Affiliation(s)
- Wen-Ting Guo
- Central Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, People's Republic of China
| | - Yangming Wang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, People's Republic of China.
| |
Collapse
|
9
|
Zhou D, Zhao Y, Hook M, Zhao W, Starlard-Davenport A, Cook MN, Jones BC, Hamre KM, Lu L. Ethanol's Effect on Coq7 Expression in the Hippocampus of Mice. Front Genet 2018; 9:602. [PMID: 30564271 PMCID: PMC6288283 DOI: 10.3389/fgene.2018.00602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/16/2018] [Indexed: 01/16/2023] Open
Abstract
Coenzyme Q (CoQ) is a well-studied molecule, present in every cell membrane in the body, best known for its roles as a mitochondrial electron transporter and a potent membrane anti-oxidant. Much of the previous work was done in vitro in yeast and more recent work has suggested that CoQ may have additional roles prompting calls for a re-assessment of its role using in vivo systems in mammals. Here we investigated the putative role of Coenzyme Q in ethanol-induced effects in vivo using BXD RI mice. We examined hippocampal expression of Coq7 in saline controls and after an acute ethanol treatment, noting enriched biologic processes and pathways following ethanol administration. We also identified 45 ethanol-related phenotypes that were significantly correlated with Coq7 expression, including six phenotypes related to conditioned taste aversion and ethanol preference. This analysis highlights the need for further investigation of Coq7 and related genes in vivo as well as previously unrecognized roles that it may play in the hippocampus.
Collapse
Affiliation(s)
- Diana Zhou
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yinghong Zhao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Michael Hook
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wenyuan Zhao
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Athena Starlard-Davenport
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Melloni N Cook
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Psychology, The University of Memphis, Memphis, TN, United States
| | - Byron C Jones
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kristin M Hamre
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
10
|
Gong YY, Luo JY, Wang L, Huang Y. MicroRNAs Regulating Reactive Oxygen Species in Cardiovascular Diseases. Antioxid Redox Signal 2018; 29:1092-1107. [PMID: 28969427 DOI: 10.1089/ars.2017.7328] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Oxidative stress caused by overproduction of reactive oxygen species (ROS) in cells is one of the most important contributors to the pathogenesis of cardiovascular and metabolic diseases such as hypertension and atherosclerosis. Excessive accumulation of ROS impairs, while limiting oxidative stress protects cardiovascular and metabolic function through various cellular mechanisms. Recent Advances: MicroRNAs (miRNAs) are novel regulators of oxidative stress in cardiovascular cells that modulate the expression of redox-related genes. This article summarizes recent advances in our understanding of how miRNAs target major ROS generators, antioxidant signaling pathways, and effectors in cells of the cardiovascular system. CRITICAL ISSUES The role of miRNAs in regulating ROS in cardiovascular cells is complicated because miRNAs can target multiple redox-related genes, act on redox regulatory pathways indirectly, and display context-dependent pro- or antioxidant effects. The complex regulatory network of ROS and the plethora of targets make it difficult to pin point the role of miRNAs and develop them as therapeutics. Therefore, these properties should be considered when designing strategies for therapeutic or diagnostic development. FUTURE DIRECTIONS Future studies can gain a better understanding of redox-related miRNAs by investigating their own regulatory mechanisms and the dual role of ROS in the cardiovascular systems. The combination of improved study design and technical advancements will reveal newer pathophysiological importance of redox-related miRNAs.
Collapse
Affiliation(s)
- Yao-Yu Gong
- 1 School of Life Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China .,2 School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China
| | - Jiang-Yun Luo
- 2 School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China .,3 Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China
| | - Li Wang
- 2 School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China .,3 Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China
| | - Yu Huang
- 2 School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China .,3 Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, China
| |
Collapse
|
11
|
Gu W, Hong X, Potter C, Qu A, Xu Q. Mesenchymal stem cells and vascular regeneration. Microcirculation 2018; 24. [PMID: 27681821 DOI: 10.1111/micc.12324] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022]
Abstract
In recent years, MSCs have emerged as a promising therapeutic cell type in regenerative medicine. They hold great promise for treating cardiovascular diseases, such as myocardial infarction and limb ischemia. MSCs may be utilized in both cell-based therapy and vascular graft engineering to restore vascular function, thereby providing therapeutic benefits to patients. The efficacy of MSCs lies in their multipotent differentiation ability toward vascular smooth muscle cells, endothelial cells and other cell types, as well as their capacity to secrete various trophic factors, which are potent in promoting angiogenesis, inhibiting apoptosis and modulating immunoreaction. Increasing our understanding of the mechanisms of MSC involvement in vascular regeneration will be beneficial in boosting present therapeutic approaches and developing novel ones to treat cardiovascular diseases. In this review, we aim to summarize current progress in characterizing the in vivo identity of MSCs, to discuss mechanisms involved in cell-based therapy utilizing MSCs, and to explore current and future strategies for vascular regeneration.
Collapse
Affiliation(s)
- Wenduo Gu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Xuechong Hong
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Claire Potter
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
12
|
Zou Y, Chen Z, Jennings BL, Zhao G, Gu Q, Bhattacharya A, Cui Y, Yu B, Malik KU, Yue J. Deletion of DGCR8 in VSMCs of adult mice results in loss of vascular reactivity, reduced blood pressure and neointima formation. Sci Rep 2018; 8:1468. [PMID: 29362439 PMCID: PMC5780492 DOI: 10.1038/s41598-018-19660-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/04/2018] [Indexed: 12/28/2022] Open
Abstract
DiGeorge syndrome chromosomal region 8 (DGCR8), a double-stranded-RNA-binding protein, participates in the miRNA biogenesis pathway and contributes to miRNA maturation by interacting with the RNAase III enzyme Drosha in cell nuclei. To investigate the role of DGCR8 in vascular smooth muscle cells (VSMCs) at the postnatal stages, we generated tamoxifen-inducible VSMC specific knockout (iKO) mice by crossing DGCR8loxp/loxp with VSMC specific tamoxifen-inducible Cre transgenic mice SMA-Cre-ERT2. DGCR8iKO mice display reduced body weight one month following tamoxifen treatment and died around 3 months. Blood pressure and vascular reactivity were significantly reduced in DGCR8iKO mice compared to control. Furthermore, loss of DGCR8 in VSMCs inhibited cell proliferation, migration and neointima formation. VSMC differentiation marker genes, including SMA and SM22, were downregulated in DGCR8 iKO mice. The majority of miRNAs were downregulated in DGCR8iKO mice. Disruption of the DGCR8-mediated miRNA biogenesis pathway attenuated multiple signaling pathways including ERK1/2 and AKT. Our results demonstrate that the DGCR8-mediated miRNA pathway is required for maintaining blood pressure, vascular reactivity and vascular wall remodeling at the postnatal stages.
Collapse
Affiliation(s)
- Yanan Zou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, P.R. China.,Departments of Pathology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA
| | - Zixuan Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, P.R. China.,Departments of Pathology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA.,School of Life Science, Beijing Institute of Technology, Beijing, P.R. China
| | - Brett L Jennings
- Departments of Pharmacology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA
| | - Guannan Zhao
- Departments of Pathology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA
| | - Qingqing Gu
- Departments of Pathology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA
| | - Anindya Bhattacharya
- Department of Genetics Genomics and Informatics, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA
| | - Yan Cui
- Department of Genetics Genomics and Informatics, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, P.R. China
| | - Kafait U Malik
- Departments of Pharmacology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA.
| | - Junming Yue
- Departments of Pathology, The University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN, 38163, USA.
| |
Collapse
|
13
|
Huang J, Song J, Qu M, Wang Y, An Q, Song Y, Yan W, Wang B, Wang X, Zhang S, Chen X, Zhao B, Liu P, Xu T, Zhang Z, Greenberg DA, Wang Y, Gao P, Zhu W, Yang GY. MicroRNA-137 and microRNA-195* inhibit vasculogenesis in brain arteriovenous malformations. Ann Neurol 2017; 82:371-384. [PMID: 28802071 DOI: 10.1002/ana.25015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Brain arteriovenous malformations (AVMs) are the most common cause of nontraumatic intracerebral hemorrhage in young adults. The genesis of brain AVM remains enigmatic. We investigated microRNA (miRNA) expression and its contribution to the pathogenesis of brain AVMs. METHODS We used a large-scale miRNA analysis of 16 samples including AVMs, hemangioblastoma, and controls to identify a distinct AVM miRNA signature. AVM smooth muscle cells (AVMSMCs) were isolated and identified by flow cytometry and immunohistochemistry, and candidate miRNAs were then tested in these cells. Migration, tube formation, and CCK-8-induced proliferation assays were used to test the effect of the miRNAs on phenotypic properties of AVMSMCs. A quantitative proteomics approach was used to identify protein expression changes in AVMSMCs treated with miRNA mimics. RESULTS A distinct AVM miRNA signature comprising a large portion of lowly expressed miRNAs was identified. Among these miRNAs, miR-137 and miR-195* levels were significantly decreased in AVMs and constituent AVMSMCs. Experimentally elevating the level of these microRNAs inhibited AVMSMC migration, tube formation, and survival in vitro and the formation of vascular rings in vivo. Proteomics showed the protein expression signature of AVMSMCs and identified downstream proteins regulated by miR-137 and miR-195* that were key signaling proteins involved in vessel development. INTERPRETATION Our results indicate that miR-137 and miR-195* act as vasculogenic suppressors in AVMs by altering phenotypic properties of AVMSMCs, and that the absence of miR-137 and miR-195* expression leads to abnormal vasculogenesis. Ann Neurol 2017;82:371-384.
Collapse
Affiliation(s)
- Jun Huang
- Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Meijie Qu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qingzhu An
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Song
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Yan
- Department of Biostatistics, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingshun Wang
- Institute of Systemic Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojin Wang
- Institute of Systemic Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Song Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing Zhao
- Emergency Department, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Tongyi Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Pingjin Gao
- Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Rujijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Yang S, Dong F, Li D, Sun H, Wu B, Sun T, Wang Y, Shen P, Ji F, Zhou D. Persistent distention of colon damages interstitial cells of Cajal through Ca 2+ -ERK-AP-1-miR-34c-SCF deregulation. J Cell Mol Med 2017; 21:1881-1892. [PMID: 28580775 PMCID: PMC5571545 DOI: 10.1111/jcmm.13108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/02/2017] [Indexed: 12/30/2022] Open
Abstract
Gastrointestinal motility disorders (GMDs) are attributed to loss of interstitial cells of Cajal (ICC), whose survival and function are deeply dependent on the activation of KIT/SCF signalling. Based on the facts that gastrointestinal distention is common in GMD patients and SCF produced by smooth muscle cells (SMCs) is usually decreased before ICC loss, we considered a possible contribution of persistent gastrointestinal distention/stretch to SCF deficiency. In this study, chronic colonic distention mouse model, diabetic gastrointestinal paresis mouse model, cultured mouse colonic SMCs and colon specimens from Hirschsprung's disease patients were used. The results showed that SCF was clearly decreased in distent colon of mice and patients, and microRNA array and real-time PCR indicated a concomitant increase of miR-34c in distent colon. A negative regulation of miR-34c on SCF expression was confirmed by luciferase reporter assays together with knock-down and overexpression of miR-34c in cultured colonic SMCs. Using EMSA and ChIP assays, we further consolidated that in response to persistent stretch, the transcription factor AP-1/c-Jun was highly activated in colonic SMCs and significantly promoted miR-34c transcription by binding to miR-34c promoter. Knock-down or overexpression of AP-1/c-Jun in cultured colonic SMCs leads to down- or up-regulation of miR-34c, respectively. In addition, the activation of AP-1/c-Jun was through ERK1/2 signalling provoked by Ca2+ overload in colonic SMCs that were subject to persistent stretch. In conclusion, our data demonstrated that persistent distention/stretch on colonic SMCs could suppress SCF production probably through Ca2+ -ERK-AP-1-miR-34c deregulation, resulting in ICC loss or impairment and GMD progress.
Collapse
Affiliation(s)
- Shu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Fang Dong
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dandan Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Haimei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Bo Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Tingyi Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Yaxi Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ping Shen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fengqing Ji
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| | - Deshan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
| |
Collapse
|
15
|
Zhou DX, Zhao Y, Baker JA, Gu Q, Hamre KM, Yue J, Jones BC, Cook MN, Lu L. The effect of alcohol on the differential expression of cluster of differentiation 14 gene, associated pathways, and genetic network. PLoS One 2017; 12:e0178689. [PMID: 28575045 PMCID: PMC5456352 DOI: 10.1371/journal.pone.0178689] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/17/2017] [Indexed: 12/13/2022] Open
Abstract
Alcohol consumption affects human health in part by compromising the immune system. In this study, we examined the expression of the Cd14 (cluster of differentiation 14) gene, which is involved in the immune system through a proinflammatory cascade. Expression was evaluated in BXD mice treated with saline or acute 1.8 g/kg i.p. ethanol (12.5% v/v). Hippocampal gene expression data were generated to examine differential expression and to perform systems genetics analyses. The Cd14 gene expression showed significant changes among the BXD strains after ethanol treatment, and eQTL mapping revealed that Cd14 is a cis-regulated gene. We also identified eighteen ethanol-related phenotypes correlated with Cd14 expression related to either ethanol responses or ethanol consumption. Pathway analysis was performed to identify possible biological pathways involved in the response to ethanol and Cd14. We also constructed a genetic network for Cd14 using the top 20 correlated genes and present several genes possibly involved in Cd14 and ethanol responses based on differential gene expression. In conclusion, we found Cd14, along with several other genes and pathways, to be involved in ethanol responses in the hippocampus, such as increased susceptibility to lipopolysaccharides and neuroinflammation.
Collapse
Affiliation(s)
- Diana X. Zhou
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Yinghong Zhao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jessica A. Baker
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Qingqing Gu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Kristin M. Hamre
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Junming Yue
- Department of Pathology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Byron C. Jones
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Melloni N. Cook
- Department of Psychology, University of Memphis, Memphis, Tennessee, United States of America
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
16
|
Gu Q, Zhao G, Wang Y, Xu B, Yue J. Silencing miR-16 Expression Promotes Angiotensin II Stimulated Vascular Smooth Muscle Cell Growth. ACTA ACUST UNITED AC 2017; 6. [PMID: 29104843 DOI: 10.4172/2168-9296.1000181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
miRNAs are a class of non-coding endogenous small RNAs that control gene expression at the posttranscriptional level and involved in cell proliferation, migration and differentiation. Dysregulation of miRNA expression is involved in a variety of human diseases including cardiovascular diseases. miRNAs have been shown to regulate vascular smooth muscle cell (VSMC) function and play vital roles in hypertension, restenosis and atherosclerosis. Here we reported that miR-16 as one of miRNAs in the miR-15 family was highly expressed in vascular smooth muscle cells (VSMCs) and involved in angiotensin II (Ang II) mediated VSMC signaling pathways. Ang II downregulated miR-16 expression in VSMCs. Lentiviral vector mediated miR-16 knockdown promoted Ang II-induced cell proliferation and migration. Moreover, silencing miR-16 enhanced Ang II induced cell cycle associated gene expression and promoted Ang II-activated cell proliferative pathways ERK1/2 and p38. Our finding demonstrated for the first time that miR-16 was a potential therapeutic target by participating in the Ang II-associated multiple signaling pathways in cardiovascular diseases.
Collapse
Affiliation(s)
- Qingqing Gu
- Department of Cardiology, the Affiliated Hospital of Nantong University, Nantong, Jiangsu.,Department of Pathology, the University of Tennessee Health Science Center, TN, Memphis, USA
| | - Guannan Zhao
- Department of Pathology, the University of Tennessee Health Science Center, TN, Memphis, USA
| | - Yinan Wang
- Department of Pathology, the University of Tennessee Health Science Center, TN, Memphis, USA
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, P. R. China
| | - Junming Yue
- Department of Pathology, the University of Tennessee Health Science Center, TN, Memphis, USA
| |
Collapse
|
17
|
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: 2.7] [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
|
18
|
Cushing L, Jiang Z, Kuang P, Lü J. The roles of microRNAs and protein components of the microRNA pathway in lung development and diseases. Am J Respir Cell Mol Biol 2016; 52:397-408. [PMID: 25211015 DOI: 10.1165/rcmb.2014-0232rt] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Decades of studies have shown evolutionarily conserved molecular networks consisting of transcriptional factors, diffusing growth factors, and signaling pathways that regulate proper lung development. Recently, microRNAs (miRNAs), small, noncoding regulatory RNAs, have been integrated into these networks. Significant advances have been made in characterizing the developmental stage- or cell type-specific miRNAs during lung development by using approaches such as genome-wide profiling and in situ hybridization. Results from gain- or loss-of-function studies revealed pivotal roles of protein components of the miRNA pathway and individual miRNAs in regulating proliferation, apoptosis, differentiation, and morphogenesis during lung development. Aberrant expression or functions of these components have been associated with pulmonary disorders, suggesting their involvement in pathogenesis of these diseases. Moreover, genetically modified mice generated in these studies have become useful models of human lung diseases. Challenges in this field include characterization of collective function and responsible targets of miRNAs specifically expressed during lung development, and translation of these basic findings into clinically relevant information for better understanding of human diseases. The goal of this review is to discuss the recent progress on the understanding of how the miRNA pathway regulates lung development, how dysregulation of miRNA activities contributes to pathogenesis of related pulmonary diseases, and to identify relevant questions and future directions.
Collapse
Affiliation(s)
- Leah Cushing
- Columbia Center for Human Development, Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Columbia University, College of Physicians & Surgeons, New York, New York
| | | | | | | |
Collapse
|
19
|
DICER1 and microRNA regulation in post-traumatic stress disorder with comorbid depression. Nat Commun 2015; 6:10106. [PMID: 26632874 PMCID: PMC4686835 DOI: 10.1038/ncomms10106] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/04/2015] [Indexed: 12/23/2022] Open
Abstract
DICER1 is an enzyme that generates mature microRNAs (miRNAs), which regulate gene expression post-transcriptionally in brain and other tissues and is involved in synaptic maturation and plasticity. Here, through genome-wide differential gene expression survey of post-traumatic stress disorder (PTSD) with comorbid depression (PTSD&Dep), we find that blood DICER1 expression is significantly reduced in cases versus controls, and replicate this in two independent cohorts. Our follow-up studies find that lower blood DICER1 expression is significantly associated with increased amygdala activation to fearful stimuli, a neural correlate for PTSD. Additionally, a genetic variant in the 3′ un-translated region of DICER1, rs10144436, is significantly associated with DICER1 expression and with PTSD&Dep, and the latter is replicated in an independent cohort. Furthermore, genome-wide differential expression survey of miRNAs in blood in PTSD&Dep reveals miRNAs to be significantly downregulated in cases versus controls. Together, our novel data suggest DICER1 plays a role in molecular mechanisms of PTSD&Dep through the DICER1 and the miRNA regulation pathway. DICER1 is required for the maturation of miRNAs which regulate expression of thousands of genes. Here the authors show significantly reduced levels of DICER1 in individuals having post-traumatic stress disorder and comorbid depression suggestive of a role in the molecular mechanism of the condition.
Collapse
|
20
|
Silencing the double-stranded RNA binding protein DGCR8 inhibits ovarian cancer cell proliferation, migration, and invasion. Pharm Res 2015; 32:769-78. [PMID: 25823356 DOI: 10.1007/s11095-013-1219-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE To evaluate the role of DiGeorge Critical Region 8 (DGCR8), a key component of miRNA biogenesis pathway in ovarian cancer. METHODS The expression of DGCR8 in ovarian cancer was detected by immunostaining and DGCR8 knockdown in ovarian cancer cells was achieved using lentiviral shRNA. Differential expression of miRNAs was determined using Nanostring miRNA arrays and validated by real-time RT-PCR. RESULTS DGCR8 was highly expressed in ovarian cancer. Knockdown of DGCR8 expression inhibits cell proliferation, migration, and invasion, as well as sensitizes cells to apoptosis induced by the chemotherapeutic drug cisplatin. Cellular survival pathways including ERK1/2 mitogen-activated protein kinase and phosphatidylinositol 3-kinase/AKT were attenuated in DGCR8 knockdown cells. DGCR8 knockdown results in dysregulated miRNA gene expression. miR-27b was identified as the most highly down-regulated miRNA in DGCR8 knockdown cells and promoted cell proliferation in ovarian cancer cells. CONCLUSIONS DGCR8 functions as an oncogene in ovarian cancer, which is in part mediated by miR-27b.
Collapse
|
21
|
Shi L, Liao J, Liu B, Zeng F, Zhang L. Mechanisms and therapeutic potential of microRNAs in hypertension. Drug Discov Today 2015; 20:1188-204. [PMID: 26004493 DOI: 10.1016/j.drudis.2015.05.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 01/08/2023]
Abstract
Hypertension is the major risk factor for the development of stroke, coronary artery disease, heart failure and renal disease. The underlying cellular and molecular mechanisms of hypertension are complex and remain largely elusive. MicroRNAs (miRNAs) are short, noncoding RNA fragments of 22-26 nucleotides and regulate protein expression post-transcriptionally by targeting the 3'-untranslated region of mRNA. A growing body of recent research indicates that miRNAs are important in the pathogenesis of arterial hypertension. Herein, we summarize the current knowledge regarding the mechanisms of miRNAs in cardiovascular remodeling, focusing specifically on hypertension. We also review recent progress of the miRNA-based therapeutics including pharmacological and nonpharmacological therapies (such as exercise training) and their potential applications in the management of hypertension.
Collapse
Affiliation(s)
- Lijun Shi
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China.
| | - Jingwen Liao
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Bailin Liu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Fanxing Zeng
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
22
|
Zhao H, Wen G, Wen G, Huang Y, Yu X, Chen Q, Afzal TA, Luong LA, Zhu J, Ye S, Shu Y, Zhang L, Xiao Q. MicroRNA-22 regulates smooth muscle cell differentiation from stem cells by targeting methyl CpG-binding protein 2. Arterioscler Thromb Vasc Biol 2015; 35:918-29. [PMID: 25722434 DOI: 10.1161/atvbaha.114.305212] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In this study, we attempted to uncover the functional impact of microRNA-22 (miR-22) and its target gene in smooth muscle cell (SMC) differentiation and delineate the molecular mechanism involved. APPROACH AND RESULTS miR-22 was found to be significantly upregulated during SMC differentiation from embryonic stem cells and adventitia stem/progenitor cells. Enforced expression of miR-22 by its mimic, while knockdown of miR-22 by its antagomiR, promotes or inhibits SMC differentiation from embryonic stem cells and adventitia stem/progenitor cells, respectively. Expectedly, miR-22 overexpression in stem cells promoted SMC differentiation in vivo. Methyl CpG-binding protein 2 (MECP2) was predicted as one of the top targets of miR-22. Interestingly, the gene expression levels of MECP2 were significantly decreased during SMC differentiation, and MECP2 was dramatically decreased in miR-22 overexpressing cells but significantly increased when miR-22 was knockdown in the differentiating stem cells. Importantly, luciferase assay showed that miR-22 substantially inhibited wild-type, but not mutant MECP2-3' untranslated region-luciferase activity. In addition, modulation of MECP2 expression levels affects multiple SMC-specific gene expression in differentiated embryonic stem cells. Mechanistically, our data showed that MECP2 could transcriptionally repress SMC gene expression through modulating various SMC transcription factors, as well as several proven SMC differentiation regulators. Evidence also revealed that enrichment of H3K9 trimethylation around the promoter regions of the SMC differentiation regulators genes were significantly increased by MECP2 overexpression. Finally, miR-22 was upregulated by platelet-derived growth factor-BB and transforming growth factor-β through a transcriptional mechanism during SMC differentiation. CONCLUSIONS miR-22 plays an important role in SMC differentiation, and epigenetic regulation through MECP2 is required for miR-22 mediated SMC differentiation.
Collapse
Affiliation(s)
- Hanqing Zhao
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | | | - Guammei Wen
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Yuan Huang
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Xiaotian Yu
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Qishan Chen
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Tayyab Adeel Afzal
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Le Anh Luong
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Jianhua Zhu
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | | | - Ye Shu
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| | - Li Zhang
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.).
| | - Qingzhong Xiao
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (H.Z., G.W., Y.H., X.Y., Q.C., T.A.A., L.A.L., Y.S., Q.X.); and Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China (Y.H., Q.C., J.Z., L.Z.)
| |
Collapse
|
23
|
MicroRNA processing machinery in the developing chick embryo. Gene Expr Patterns 2014; 16:114-21. [DOI: 10.1016/j.gep.2014.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/12/2014] [Accepted: 09/24/2014] [Indexed: 12/21/2022]
|
24
|
Reduced intestinal brain-derived neurotrophic factor increases vagal sensory innervation of the intestine and enhances satiation. J Neurosci 2014; 34:10379-93. [PMID: 25080597 DOI: 10.1523/jneurosci.1042-14.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is produced by developing and mature gastrointestinal (GI) tissues that are heavily innervated by autonomic neurons and may therefore control their development or function. To begin investigating this hypothesis, we compared the morphology, distribution, and density of intraganglionic laminar endings (IGLEs), the predominant vagal GI afferent, in mice with reduced intestinal BDNF (INT-BDNF(-/-)) and controls. Contrary to expectations of reduced development, IGLE density and longitudinal axon bundle number in the intestine of INT-BDNF(-/-) mice were increased, but stomach IGLEs were normal. INT-BDNF(-/-) mice also exhibited increased vagal sensory neuron numbers, suggesting that their survival was enhanced. To determine whether increased intestinal IGLE density or other changes to gut innervation in INT-BDNF(-/-) mice altered feeding behavior, meal pattern and microstructural analyses were performed. INT-BDNF(-/-) mice ate meals of much shorter duration than controls, resulting in reduced meal size. Increased suppression of feeding in INT-BDNF(-/-) mice during the late phase of a scheduled meal suggested that increased satiation signaling contributed to reduced meal duration and size. Furthermore, INT-BDNF(-/-) mice demonstrated increases in total daily intermeal interval and satiety ratio, suggesting that satiety signaling was augmented. Compensatory responses maintained normal daily food intake and body weight in INT-BDNF(-/-) mice. These findings suggest a target organ-derived neurotrophin suppresses development of that organ's sensory innervation and sensory neuron survival and demonstrate a role for BDNF produced by peripheral tissues in short-term controls of feeding, likely through its regulation of development or function of gut innervation, possibly including augmented intestinal IGLE innervation.
Collapse
|
25
|
miRNAs and lncRNAs in vascular injury and remodeling. SCIENCE CHINA-LIFE SCIENCES 2014; 57:826-35. [PMID: 25104456 DOI: 10.1007/s11427-014-4698-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/20/2014] [Indexed: 01/17/2023]
Abstract
Vascular injury, remodeling, as well as angiogenesis, are the leading causes of coronary or cerebrovascular disease. The blood vessel functional imbalance trends to induce atherosclerosis, hypertension, and pulmonary arterial hypertension. As several genes have been identified to be dynamically regulated during vascular injury and remodeling, it is becoming widely accepted that several types of non-coding RNA, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are involved in regulating the endothelial cell and vascular smooth muscle cell (VSMC) behaviors. Here, we review the progress of the extant studies on mechanistic, clinical and diagnostic implications of miRNAs and lncRNAs in vascular injury and remodeling, as well as angiogenesis, emphasizing the important roles of miRNAs and lncRNAs in vascular diseases. Furthermore, we introduce the interaction between miRNAs and lncRNAs, and highlight the mechanism through which lncRNAs are regulating the miRNA function. We envisage that continuous in-depth research of non-coding RNAs in vascular disease will have significant implications for the treatment of coronary or cerebrovascular diseases.
Collapse
|
26
|
Xin H, Deng K, Fu M. Post-transcriptional gene regulation by RNA-binding proteins in vascular endothelial dysfunction. SCIENCE CHINA. LIFE SCIENCES 2014; 57:836-44. [PMID: 25104457 PMCID: PMC7089175 DOI: 10.1007/s11427-014-4703-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022]
Abstract
Endothelial cell dysfunction is a term which implies the dysregulation of normal endothelial cell functions, including impairment of the barrier functions, control of vascular tone, disturbance of proliferative and migratory capacity of endothelial cells, as well as control of leukocyte trafficking. Endothelial dysfunction is an early step in vascular inflammatory diseases such as atherosclerosis, diabetic vascular complications, sepsis-induced or severe virus infection-induced organ injuries. The expressions of inflammatory cytokines and vascular adhesion molecules induced by various stimuli, such as modified lipids, smoking, advanced glycation end products and bacteria toxin, significantly contribute to the development of endothelial dysfunction. The transcriptional regulation of inflammatory cytokines and vascular adhesion molecules has been well-studied. However, the regulation of those gene expressions at post-transcriptional level is emerging. RNA-binding proteins have emerged as critical regulators of gene expression acting predominantly at the post-transcriptional level in microRNA-dependent or independent manners. This review summarizes the latest insights into the roles of RNA-binding proteins in controlling vascular endothelial cell functions and their contribution to the pathogenesis of vascular inflammatory diseases.
Collapse
Affiliation(s)
- HongBo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| | - KeYu Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| | - MinGui Fu
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| |
Collapse
|
27
|
Chu R, Mo G, Duan Z, Huang M, Chang J, Li X, Liu P. miRNAs affect the development of hepatocellular carcinoma via dysregulation of their biogenesis and expression. Cell Commun Signal 2014; 12:45. [PMID: 25012758 PMCID: PMC4117189 DOI: 10.1186/s12964-014-0045-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/24/2014] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of hepatocellular carcinoma (HCC) is not fully understood, which has affected the early diagnosis and treatment of HCC and the survival time of patients. MicroRNAs (miRNAs) are a class of evolutionarily conserved small, non-coding RNAs, which regulate the expression of various genes post-transcriptionally. Emerging evidence indicates that the key enzymes involved in the miRNA biosynthesis pathway and some tumor-specific miRNAs are widely deregulated or upregulated in HCC and closely associated with the occurrence and development of various cancers, including HCC. Early studies have shown that miRNAs have critical roles in HCC progression by targeting many critical protein-coding genes, thereby contributing to the promotion of cell proliferation; the avoidance of apoptosis, inducing via angiogenesis; and the activation of invasion and metastasis pathways. Experimental data indicate that discovery of increasing numbers of aberrantly expressed miRNAs has opened up a new field for investigating the molecular mechanism of HCC progression. In this review, we describe the current knowledge about the roles and validated targets of miRNAs in the above pathways that are known to be hallmarks of HCC, and we also describe the influence of genetic variations in miRNA biosynthesis and genes.
Collapse
|
28
|
Cheong AWY, Pang RTK, Liu WM, Kottawatta KSA, Lee KF, Yeung WSB. MicroRNA Let-7a and dicer are important in the activation and implantation of delayed implanting mouse embryos. Hum Reprod 2014; 29:750-62. [PMID: 24419497 DOI: 10.1093/humrep/det462] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Does Let-7a have a functional role in modulating dicer expression to activate dormant mouse blastocysts for implantation? SUMMARY ANSWER Let-7a post-transcriptionally regulates dicer expression altering microRNA expression to affect the implantation competency of the activated blastocysts. WHAT IS KNOWN ALREADY The Let-7a microRNA is up-regulated during blastocyst dormancy and its forced-expression suppresses embryo implantation in vitro and in vivo. Dicer is a Let-7 target, which processes pre-microRNA to mature microRNA. STUDY DESIGN, SIZE, DURATION The effects on the expression of Let-7a and dicer in dormant blastocysts during the first 12 h after estradiol-induced activation, and the relationship between Let-7a and dicer in preimplantation embryos were determined. The effects on the microRNA expression and embryo implantation in vivo in dicer-knockdown mouse 5-8 cell embryos and dormant blastocysts at 1 h post estradiol activation were also studied. PARTICIPANTS/MATERIALS, SETTING, METHODS ICR female mice at 6 weeks of age were ovariectomized on Day 4 of pregnancy to generate the delayed implantation model. Mouse 5-8 cell embryos and/or dormant blastocysts at 1 h after estradiol injection were electroporated with dicer siRNA and Let-7a precursor or Let-7a inhibitor. At 48 h post electroporation, the Let-7a expression, dicer transcripts and proteins in the embryos were determined using qPCR and immunostaining/western blotting, respectively. All experiments were repeated at least three times. MAIN RESULTS AND THE ROLE OF CHANCE Estradiol injection down-regulated Let-7a and up-regulated dicer in the dormant blastocysts during the first 12 h post-activation. Dicer knockdown at 1 h post-activation of blastocysts suppressed EGFR expression, attenuated EGF binding and compromised implantation of the transferred embryos. Let-7a transcriptionally regulated dicer by binding to the 3'-UTR of dicer in trophoblast cells. Dicer knockdown in blastocysts suppressed mature Let-7a expression and compromised implantation. LIMITATIONS, REASONS FOR CAUTION Gain- and loss-of-function approaches were used by analyzing transient expressions of transfected microRNA modulators or genes. The consequence of the Let-7a-dicer interaction on pregnancy remains to be determined. The study used the mouse as a model and the applicability of the observed phenomena in humans warrants further investigation. WIDER IMPLICATIONS OF THE FINDINGS Our results indicate that the Let-7a-dicer interaction leads to differential microRNA expression in dormant blastocysts after estradiol activation. Because the expression pattern of Let-7a in human blastocysts is similar to that in mouse blastocysts, our observation that the Let-7a-dicer interaction has a role in regulating the implantation potential of the mouse blastocysts could be applicable to humans. STUDY FUNDING/COMPETING INTEREST(S) This project is supported partly by a research grant from the Research Grant Council to W.S.B.Y. The authors have no competing interests to declare.
Collapse
Affiliation(s)
- Ana W Y Cheong
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
29
|
Fox EA, Biddinger JE, Baquet ZC, Jones KR, McAdams J. Loss of neurotrophin-3 from smooth muscle disrupts vagal gastrointestinal afferent signaling and satiation. Am J Physiol Regul Integr Comp Physiol 2013; 305:R1307-22. [PMID: 24068045 PMCID: PMC3882559 DOI: 10.1152/ajpregu.00337.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A large proportion of vagal afferents are dependent on neurotrophin-3 (NT-3) for survival. NT-3 is expressed in developing gastrointestinal (GI) smooth muscle, a tissue densely innervated by vagal mechanoreceptors, and thus could regulate their survival. We genetically ablated NT-3 from developing GI smooth muscle and examined the pattern of loss of NT-3 expression in the GI tract and whether this loss altered vagal afferent signaling or feeding behavior. Meal-induced c-Fos activation was reduced in the solitary tract nucleus and area postrema in mice with a smooth muscle-specific NT-3 knockout (SM-NT-3(KO)) compared with controls, suggesting a decrease in vagal afferent signaling. Daily food intake and body weight of SM-NT-3(KO) mice and controls were similar. Meal pattern analysis revealed that mutants, however, had increases in average and total daily meal duration compared with controls. Mutants maintained normal meal size by decreasing eating rate compared with controls. Although microstructural analysis did not reveal a decrease in the rate of decay of eating in SM-NT-3(KO) mice, they ate continuously during the 30-min meal, whereas controls terminated feeding after 22 min. This led to a 74% increase in first daily meal size of SM-NT-3(KO) mice compared with controls. The increases in meal duration and first meal size of SM-NT-3(KO) mice are consistent with reduced satiation signaling by vagal afferents. This is the first demonstration of a role for GI NT-3 in short-term controls of feeding, most likely involving effects on development of vagal GI afferents that regulate satiation.
Collapse
Affiliation(s)
- Edward A Fox
- Behavioral Neurogenetics Laboratory, Department of Psychological Sciences, Purdue University, West Lafayette, Indiana; and
| | | | | | | | | |
Collapse
|
30
|
Ro S, Ma HY, Park C, Ortogero N, Song R, Hennig GW, Zheng H, Lin YM, Moro L, Hsieh JT, Yan W. The mitochondrial genome encodes abundant small noncoding RNAs. Cell Res 2013; 23:759-774. [PMID: 23478297 PMCID: PMC3674384 DOI: 10.1038/cr.2013.37] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/08/2013] [Accepted: 01/28/2013] [Indexed: 02/07/2023] Open
Abstract
Small noncoding RNAs identified thus far are all encoded by the nuclear genome. Here, we report that the murine and human mitochondrial genomes encode thousands of small noncoding RNAs, which are predominantly derived from the sense transcripts of the mitochondrial genes (host genes), and we termed these small RNAs mitochondrial genome-encoded small RNAs (mitosRNAs). DICER inactivation affected, but did not completely abolish mitosRNA production. MitosRNAs appear to be products of currently unidentified mitochondrial ribonucleases. Overexpression of mitosRNAs enhanced expression levels of their host genes in vitro, and dysregulated mitosRNA expression was generally associated with aberrant mitochondrial gene expression in vivo. Our data demonstrate that in addition to 37 known mitochondrial genes, the mammalian mitochondrial genome also encodes abundant mitosRNAs, which may play an important regulatory role in the control of mitochondrial gene expression in the cell.
Collapse
Affiliation(s)
- Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Hsiu-Yen Ma
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- Department of Urology, College of Medicine, National Cheng Kung University, 138 Sheng Li Road, Tainan 70403, China
| | - Chanjae Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Nicole Ortogero
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Rui Song
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- Current address: Department of Molecular and Cell Biology, University of California, Berkeley, 142 Life Sciences Addition # 3200, Berkeley, CA 94720-3200, USA
| | - Grant W Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Yung-Ming Lin
- Department of Urology, College of Medicine, National Cheng Kung University, 138 Sheng Li Road, Tainan 70403, China
| | - Loredana Moro
- Institute of Biomembranes and Bioenergetics, National Research Council (CNR), Bari 70123, Italy
| | - Jer-Tsong Hsieh
- Department of Urology, Southwestern Medical Center, University of Texas, Dallas, TX 75235, USA
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| |
Collapse
|
31
|
Albinsson S, Swärd K. Targeting smooth muscle microRNAs for therapeutic benefit in vascular disease. Pharmacol Res 2013; 75:28-36. [PMID: 23611811 DOI: 10.1016/j.phrs.2013.04.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 12/16/2022]
Abstract
In view of the bioinformatic projection that a third of all protein coding genes and essentially all biological pathways are under control of microRNAs (miRNAs), it is not surprising that this class of small RNAs plays roles in vascular disease progression. MiRNAs have been shown to be involved in cholesterol turnover, thrombosis, glucose homeostasis and vascular function. Some miRNAs appear to be specific for certain cells, and the role that such cell-specific miRNAs play in vascular disease is only beginning to be appreciated. A notable example is the miR-143/145 cluster which is enriched in mature and highly differentiated smooth muscle cells (SMCs). Here we outline and discuss the recent literature on SMC-expressed miRNAs in major vascular diseases, including atherosclerosis, neointima formation, aortic aneurysm formation, and pulmonary arterial hypertension. Forced expression of miR-145 emerges as a promising strategy for reduction and stabilization of atherosclerotic plaques as well as for reducing neointimal hyperplasia. It is concluded that if obstacles in the form of delivery and untoward effects of antimirs and mimics can be overcome, the outlook for targeting of SMC-specific miRNAs for therapeutic benefit in vascular disease is bright.
Collapse
|
32
|
miRNA biogenesis enzyme Drosha is required for vascular smooth muscle cell survival. PLoS One 2013; 8:e60888. [PMID: 23637774 PMCID: PMC3630177 DOI: 10.1371/journal.pone.0060888] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/04/2013] [Indexed: 12/03/2022] Open
Abstract
miRNA biogenesis enzyme Drosha cleaves double-stranded primary miRNA by interacting with double-stranded RNA binding protein DGCR8 and processes primary miRNA into precursor miRNA to participate in the miRNA biogenesis pathway. The role of Drosha in vascular smooth muscle cells (VSMCs) has not been well addressed. We generated Drosha conditional knockout (cKO) mice by crossing VSMC-specific Cre mice, SM22-Cre, with Drosha loxp/loxp mice. Disruption of Drosha in VSMCs resulted in embryonic lethality at E14.5 with severe liver hemorrhage in mutant embryos. No obvious developmental delay was observed in Drosha cKO embryos. The vascular structure was absent in the yolk sac of Drosha homozygotes at E14.5. Loss of Drosha reduced VSMC proliferation in vitro and in vivo. The VSMC differentiation marker genes, including αSMA, SM22, and CNN1, and endothelial cell marker CD31 were significantly downregulated in Drosha cKO mice compared to controls. ERK1/2 mitogen-activated protein kinase and the phosphatidylinositol 3-kinase/AKT were attenuated in VSMCs in vitro and in vivo. Disruption of Drosha in VSMCs of mice leads to the dysregulation of miRNA expression. Using bioinformatics approach, the interactions between dysregulated miRNAs and their target genes were analyzed. Our data demonstrated that Drosha is required for VSMC survival by targeting multiple signaling pathways.
Collapse
|
33
|
Fox EA, Biddinger JE, Jones KR, McAdams J, Worman A. Mechanism of hyperphagia contributing to obesity in brain-derived neurotrophic factor knockout mice. Neuroscience 2012; 229:176-99. [PMID: 23069761 DOI: 10.1016/j.neuroscience.2012.09.078] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 09/28/2012] [Accepted: 09/28/2012] [Indexed: 10/27/2022]
Abstract
Global-heterozygous and brain-specific homozygous knockouts (KOs) of brain-derived neurotrophic factor (BDNF) cause late- and early-onset obesity, respectively, both involving hyperphagia. Little is known about the mechanism underlying this hyperphagia or whether BDNF loss from peripheral tissues could contribute to overeating. Since global-homozygous BDNF-KO is perinatal lethal, a BDNF-KO that spared sufficient brainstem BDNF to support normal health was utilized to begin to address these issues. Meal pattern and microstructure analyses suggested overeating of BDNF-KO mice was mediated by deficits in both satiation and satiety that resulted in increased meal size and frequency and implicated a reduction of vagal signaling from the gut to the brain. Meal-induced c-Fos activation in the nucleus of the solitary tract, a more direct measure of vagal afferent signaling, however, was not decreased in BDNF-KO mice, and thus was not consistent with a vagal afferent role. Interestingly though, meal-induced c-Fos activation was increased in the dorsal motor nucleus of the vagus nerve (DMV) of BDNF-KO mice. This could imply that augmentation of vago-vagal digestive reflexes occurred (e.g., accommodation), which would support increased meal size and possibly increased meal number by reducing the increase in intragastric pressure produced by a given amount of ingesta. Additionally, vagal sensory neuron number in BDNF-KO mice was altered in a manner consistent with the increased meal-induced activation of the DMV. These results suggest reduced BDNF causes satiety and satiation deficits that support hyperphagia, possibly involving augmentation of vago-vagal reflexes mediated by central pathways or vagal afferents regulated by BDNF levels.
Collapse
Affiliation(s)
- E A Fox
- Behavioral Neurogenetics Laboratory, Department of Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | | | | | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW In this review, we summarize the recent advances regarding microRNA (miRNA) functions in the regulation of vascular smooth muscle cell (VSMC) differentiation and phenotypic modulation. RECENT FINDINGS Multiple miRNAs are found to be responsible for VSMC differentiation and proliferation under physiological or pathological condition. A single miRNA downregulates multiple targets, whereas a single gene is regulated by multiple miRNAs to modulate a specific aspect of VSMC phenotype. SUMMARY The phenotype of VSMCs is dynamically regulated in response to environmental stimuli. Deregulation of phenotype switching is associated with vascular diseases. Several miRNAs have been found to be highly expressed in the vasculature, to modulate VSMC phenotype, and to be dysregulated in vascular diseases. By regulating mRNA and/or protein levels posttranscriptionally, miRNAs provide a delicate regulation in the complex molecular networks that regulate the vascular system. Understanding the functions of miRNAs in the regulation of VSMC differentiation and phenotype switching provides new insights into the mechanisms of vascular development, function, and dysfunction.
Collapse
|
35
|
Wu Q, Song R, Ortogero N, Zheng H, Evanoff R, Small CL, Griswold MD, Namekawa SH, Royo H, Turner JM, Yan W. The RNase III enzyme DROSHA is essential for microRNA production and spermatogenesis. J Biol Chem 2012; 287:25173-90. [PMID: 22665486 DOI: 10.1074/jbc.m112.362053] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
DROSHA is a nuclear RNase III enzyme responsible for cleaving primary microRNAs (miRNAs) into precursor miRNAs and thus is essential for the biogenesis of canonical miRNAs. DICER is a cytoplasmic RNase III enzyme that not only cleaves precursor miRNAs to produce mature miRNAs but also dissects naturally formed/synthetic double-stranded RNAs to generate small interfering RNAs (siRNAs). To investigate the role of canonical miRNA and/or endogenous siRNA production in spermatogenesis, we generated Drosha or Dicer conditional knock-out (cKO) mouse lines by inactivating Drosha or Dicer exclusively in spermatogenic cells in postnatal testes using the Cre-loxp strategy. Both Drosha and Dicer cKO males were infertile due to disrupted spermatogenesis characterized by depletion of spermatocytes and spermatids leading to oligoteratozoospermia or azoospermia. The developmental course of spermatogenic disruptions was similar at morphological levels between Drosha and Dicer cKO males, but Drosha cKO testes appeared to be more severe in spermatogenic disruptions than Dicer cKO testes. Microarray analyses revealed transcriptomic differences between Drosha- and Dicer-null pachytene spermatocytes or round spermatids. Although levels of sex-linked mRNAs were mildly elevated, meiotic sex chromosome inactivation appeared to have occurred normally. Our data demonstrate that unlike DICER, which is required for the biogenesis of several small RNA species, DROSHA is essential mainly for the canonical miRNA production, and DROSHA-mediated miRNA production is essential for normal spermatogenesis and male fertility.
Collapse
Affiliation(s)
- Qiuxia Wu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen Z, Wu J, Yang C, Fan P, Balazs L, Jiao Y, Lu M, Gu W, Li C, Pfeffer LM, Tigyi G, Yue J. DiGeorge syndrome critical region 8 (DGCR8) protein-mediated microRNA biogenesis is essential for vascular smooth muscle cell development in mice. J Biol Chem 2012; 287:19018-28. [PMID: 22511778 DOI: 10.1074/jbc.m112.351791] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
DiGeorge Critical Region 8 (DGCR8) is a double-stranded RNA-binding protein that interacts with Drosha and facilitates microRNA (miRNA) maturation. However, the role of DGCR8 in vascular smooth muscle cells (VSMCs) is not well understood. To investigate whether DGCR8 contributes to miRNA maturation in VSMCs, we generated DGCR8 conditional knockout (cKO) mice by crossing VSMC-specific Cre mice (SM22-Cre) with DGCR8(loxp/loxp) mice. We found that loss of DGCR8 in VSMCs resulted in extensive liver hemorrhage and embryonic mortality between embryonic days (E) 12.5 and E13.5. DGCR8 cKO embryos displayed dilated blood vessels and disarrayed vascular architecture. Blood vessels were absent in the yolk sac of DGCR8 KOs after E12.5. Disruption of DGCR8 in VSMCs reduced VSMC proliferation and promoted apoptosis in vitro and in vivo. In DGCR8 cKO embryos and knockout VSMCs, differentiation marker genes, including αSMA, SM22, and CNN1, were significantly down-regulated, and the survival pathways of ERK1/2 mitogen-activated protein kinase and the phosphatidylinositol 3-kinase/AKT were attenuated. Knockout of DGCR8 in VSMCs has led to down-regulation of the miR-17/92 and miR-143/145 clusters. We further demonstrated that the miR-17/92 cluster promotes VSMC proliferation and enhances VSMC marker gene expression, which may contribute to the defects of DGCR8 cKO mutants. Our results indicate that the DGCR8 gene is required for vascular development through the regulation of VSMC proliferation, apoptosis, and differentiation.
Collapse
Affiliation(s)
- Zixuan Chen
- Department of Physiology, Campbell Clinic, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The discovery of the regulatory role of noncoding RNAs, and micro (mi)RNAs in particular, has added a new layer of complexity to our understanding of cardiovascular development. miRNAs regulate and modulate various steps of cardiovascular morphogenesis, cell proliferation, differentiation, and phenotype modulation. miRNAs simultaneously regulate multiple targets, and many miRNAs can bind to the same target, allowing for a complex pattern of regulation of gene expression. miRNA families are continuously added during evolution paralleling the increased complexity of the cardiovascular system in vertebrates compared with invertebrates. Several lines of evidence suggest that the appearance of miRNAs is at least in part responsible for the formation of complex organ systems and stable regulatory mechanisms in vertebrates. We review the current understanding of miRNAs during cardiovascular development. Further progress in this area will help to decipher quantitative changes in gene expression that provide robustness to cellular phenotypes and regulatory options to diseases processes. miRNAs might also provide clues to better understand congenital heart defects, which are the most common birth defects in human newborns.
Collapse
Affiliation(s)
- Thomas Boettger
- From the Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Bad Nauheim, Germany
| | - Thomas Braun
- From the Max-Planck-Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Bad Nauheim, Germany
| |
Collapse
|
38
|
Zhang L, Zhou Y, Zhu J, Xu Q. An updated view on stem cell differentiation into smooth muscle cells. Vascul Pharmacol 2012; 56:280-7. [PMID: 22421140 DOI: 10.1016/j.vph.2012.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/17/2012] [Accepted: 02/28/2012] [Indexed: 12/16/2022]
Abstract
Stem cells possess the ability of self-renewal and give rise to specific cell types. The differentiation of stem cells involves environmental factors, transduction of extra and intra-cellular signals, regulation of gene expression by transcriptional factors, microRNAs and chromosome structural modifiers. Vascular SMCs play a profound role in blood vessel physiology and participate in a number of cardiovascular diseases such as atherosclerosis, hypertension and restenosis. In addition, SMCs could be a crucial cell component for vascular tissue engineering. In this review, we aim to update the recent progress on the mechanisms of SMC differentiation from stem cells, which involve reactive oxygen species, epigenetic modifiers, transcription factors and microRNAs coordinately regulated during stem cell differentiation. We will also discuss the potential application of stem cell therapy for patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou 310003, PR China
| | | | | | | |
Collapse
|
39
|
Abstract
MicroRNAs play essential roles during animal development, including in developing muscle. Many microRNAs are expressed during muscle development and some, like miR-1 and miR-133, are muscle specific. Muscle microRNAs are integrated into myogenic regulatory networks: their expression is under the transcriptional and posttranscriptional control of myogenic factors, and they in turn have widespread control of muscle gene expression. This review summarizes recent work characterizing the function of microRNAs in muscle biology and specifically focuses on the genetic analysis of muscle microRNAs in a variety of model organisms including worms, flies, zebrafish, and mice.
Collapse
Affiliation(s)
- Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
40
|
McDonald RA, Hata A, MacLean MR, Morrell NW, Baker AH. MicroRNA and vascular remodelling in acute vascular injury and pulmonary vascular remodelling. Cardiovasc Res 2011; 93:594-604. [PMID: 22065733 DOI: 10.1093/cvr/cvr299] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vascular remodelling is an integral pathological process central to a number of cardiovascular diseases. The complex interplay between distinct cell populations in the vessel wall following vascular injury leads to inflammation, cellular dysfunction, pro-growth signals in the smooth muscle cell (SMC) compartment, and the acquisition of a synthetic phenotype. Although the signals for vascular remodelling are diverse in different pathological contexts, SMC proliferation and migration are consistently observed. It is therefore critical to elucidate key mechanisms central to these processes. MicroRNAs (miRNAs) are small non-coding sequences of RNA that have the capacity to regulate many genes, pathways, and complex biological networks within cells, acting either alone or in concert with one another. In diseases such as cancer and cardiac disease, the role of miRNA in disease pathogenesis has been documented in detail. In contrast, despite a great deal of interest in miRNA, relatively few studies have directly assessed the role of miRNA in vascular remodelling. The potential for modulation of miRNA to achieve therapeutic benefits in this setting is attractive. Here, we focus on the role of miRNA in vascular inflammation and remodelling associated with acute vascular injury (vein graft disease, angioplasty restenosis, and in-stent restenosis) as well as in vascular remodelling associated with the development of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Robert A McDonald
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | | | | | | | | |
Collapse
|
41
|
Singh MK, Lu MM, Massera D, Epstein JA. MicroRNA-processing enzyme Dicer is required in epicardium for coronary vasculature development. J Biol Chem 2011; 286:41036-45. [PMID: 21969379 DOI: 10.1074/jbc.m111.268573] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The epicardium is a sheet of epithelial cells covering the heart during early cardiac development. In recent years, the epicardium has been identified as an important contributor to cardiovascular development, and epicardium-derived cells have the potential to differentiate into multiple cardiac cell lineages. Some epicardium-derived cells that undergo epithelial-to-mesenchymal transition and delaminate from the surface of the developing heart subsequently invade the myocardium and differentiate into vascular smooth muscle of the developing coronary vasculature. MicroRNAs (miRNAs) have been implicated broadly in tissue patterning and development, including in the heart, but a role in epicardium is unknown. To examine the role of miRNAs during epicardial development, we conditionally deleted the miRNA-processing enzyme Dicer in the proepicardium using Gata5-Cre mice. Epicardial Dicer mutant mice are born in expected Mendelian ratios but die immediately after birth with profound cardiac defects, including impaired coronary vessel development. We found that loss of Dicer leads to impaired epicardial epithelial-to-mesenchymal transition and a reduction in epicardial cell proliferation and differentiation into coronary smooth muscle cells. These results demonstrate a critical role for Dicer, and by implication miRNAs, in murine epicardial development.
Collapse
Affiliation(s)
- Manvendra K Singh
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
42
|
MicroRNAs regulating cell pluripotency and vascular differentiation. Vascul Pharmacol 2011; 55:69-78. [PMID: 21854874 DOI: 10.1016/j.vph.2011.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/26/2011] [Accepted: 08/02/2011] [Indexed: 12/12/2022]
Abstract
Human embryonic stem cells (hESC) offer broad potential for regenerative medicine owing to their capacity for self renewal, exponential scale up and differentiation into any cell type in the adult body. hESC have been proposed as a potentially unlimited source for the generation of transplantable, healthy, functional vascular cells for repair of ischemic tissues. To optimally harness this potential necessitates precise control over biological processes that govern maintenance, pluripotency and cell differentiation including signalling cascades, gene expression profiles and epigenetic modification. Such control may be elicited by microRNAs, which are powerful negative regulators of gene expression. Here, we review the role for miRNAs in both the maintenance of pluripotency and differentiation of cells to a cardiovascular lineage including endothelial cells, vascular smooth muscle cells and cardiomyocytes and put this into context for regenerative medicine in the cardiovascular system.
Collapse
|
43
|
Quintavalle C, Garofalo M, Croce CM, Condorelli G. "ApoptomiRs" in vascular cells: their role in physiological and pathological angiogenesis. Vascul Pharmacol 2011; 55:87-91. [PMID: 21798370 DOI: 10.1016/j.vph.2011.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/04/2011] [Accepted: 07/11/2011] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNAs) have emerged as crucial players regulating the magnitude of gene expression in a variety of organisms. This class of short (22 nucleotides) noncoding RNA molecules have been shown to participate in almost every cellular process investigated so far, and their deregulation is observed in different human pathologies including cancer, heart disease, and neurodegeneration. These new molecular regulators have been identified also in endothelial cells (ECs), and their role in the regulation of different aspects of the angiogenic process has been recently investigated in a variety of laboratories. The current review focuses on the research progress regarding the roles of miRNAs in vascular pathology and their potential therapeutic applications for vascular diseases associated with abnormal angiogenesis, such as cancer.
Collapse
Affiliation(s)
- Cristina Quintavalle
- Department of Cellular and Molecular Biology and Pathology, Federico II University of Naples, Biotechnological Science Faculty, and IEOS, CNR Naples, Italy
| | | | | | | |
Collapse
|