1
|
Almuwaqqat Z, Liu C, Kim JH, Hammadah M, Alkhoder A, Raggi P, Shah AJ, Bremner JD, Vaccarino V, Sun YV, Quyyumi AA. A novel GWAS locus influences microvascular response to mental stress and predicts adverse cardiovascular events. Sci Rep 2024; 14:23479. [PMID: 39379420 PMCID: PMC11461842 DOI: 10.1038/s41598-024-54566-z] [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: 07/31/2022] [Accepted: 02/14/2024] [Indexed: 10/10/2024] Open
Abstract
Excessive peripheral microvascular constriction during acute psychological stress reflects similar changes in coronary blood flow and is a predictor of adverse cardiovascular outcomes. Among individuals with coronary artery disease (CAD), we sought to determine if genetic factors contribute to the degree of microvascular constriction during mental stress. A total of 580 stable CAD individuals from two prospective cohort studies underwent mental stress testing. Digital pulse wave amplitude was continuously measured and the stress/rest (sPAT) ratio of pulse wave amplitude was calculated. Race stratified genome-wide association studies (GWAS) of sPAT-ratio were conducted using linear regression of additive genetic models. A trans-ethnic meta-analysis integrated the four sets of GWAS results. Participants were followed for the outcome of recurrent cardiovascular events (myocardial infarction, heart failure, revascularization, and CV death) for a median of 5 years. We used Wei-Lin-Weissfeld (WLW) model to assess the association between sPAT-ratio with recurrent events. Mean age was 63 ± 9. We identified three SNPs in linkage disequilibrium, closely related to chr7:111,666,943 T > C (rs6466396) that were associated with sPAT-ratio (p = 6.68E-09). Participants homozygous for the T allele had 80% higher risk of incident adverse events (HR 1.8, 95% CI, 1.4-2.2). Also, participants with a lower sPAT-ratio (< median) had a higher adverse event rate, hazard ratio (HR) = 1.3, [95%confidence interval (CI), 1.1-1.6]. However, adjustment for the genotypes did not substantially alter the impact of sPAT ratio on adverse outcome rate. In conclusion, we have identified a genetic basis for stress-induced vasomotion. The 3 linked variants modulate vasoconstriction during mental stress may have a prognostic importance.
Collapse
Affiliation(s)
- Zakaria Almuwaqqat
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Chang Liu
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jeong Hwan Kim
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
| | - Muhammad Hammadah
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
| | - Ayman Alkhoder
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
| | - Paolo Raggi
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Amit J Shah
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, Georgia
| | - J Douglas Bremner
- Atlanta VA Medical Center, Decatur, Georgia
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, USA
| | - Viola Vaccarino
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA, 30322, USA.
| |
Collapse
|
2
|
Shen Y, Liao D, Shangguan W, Chen L. Variation and significance of serum microRNA-21 level in pediatric pulmonary artery hypertension associated with congenital heart disease. Front Cardiovasc Med 2024; 11:1424679. [PMID: 39309603 PMCID: PMC11413868 DOI: 10.3389/fcvm.2024.1424679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Objective This study strives to the variation and significance of microRNA-21 (miR-21) in children with congenital heart disease (CHD)-related pulmonary artery hypertension (PAH). Methods Children with CHD (n = 179) were selected as subjects, including 101 children without PAH and 78 children with PAH. All children underwent general data collection, laboratory examination, echocardiography and cardiac catheterization. After detection of serum miR-21 expression, the predictive value and the impacts of serum miR-21 for PAH and postoperative critical illness were analyzed. Results Serum creatine kinase isoenzyme (CK-MB), B-type natriuretic peptide (BNP) and miR-21 were elevated, but ejection fraction (EF) and cardiac index (CI) were decreased in the CHD-PAH group. Serum miR-21 assisted in predicting PAH in CHD children, with the area under curve (AUC) of 0.801 (95% CI of 0.735∼0.857), a cut-off value of 2.56, sensitivity of 73.08, and specificity of 72.28%. Serum miR-21 in children with CHD-PAH was correlated with clinicopathological indicators such as systolic pulmonary artery pressure, mean pulmonary arterial pressure, BNP and CI. Serum miR-21 helped predict the development of postoperative critical illness in children with CHD-PAH, with an AUC of 0.859 (95% CI: 0.762-0.927, cut-off value: 4.55, sensitivity: 69.57%, specificity: 92.73%). Increased serum miR-21 was an independent risk factor of postoperative critical illness in children with CHD-PAH. Conclusion Serum miR-21 was upregulated in children with CHD-PAH, which may serve as a predictive biomarker for the onset of PAH and postoperative critical illness in CHD children.
Collapse
Affiliation(s)
- Yanming Shen
- Cardiac Surgery, Fujian Medical University, Fuzhou, Fujian, China
| | - Dongshan Liao
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery, (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Wenlin Shangguan
- Thoracic and Cardiovascular Surgery, Fuzhou Changle District People’s Hospital, Fuzhou, Fujian, China
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery, (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Zhang H, Zhang K, Gu Y, Tu Y, Ouyang C. Roles and Mechanisms of miRNAs in Abdominal Aortic Aneurysm: Signaling Pathways and Clinical Insights. Curr Atheroscler Rep 2024; 26:273-287. [PMID: 38709435 DOI: 10.1007/s11883-024-01204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE OF REVIEW Abdominal aortic aneurysm refers to a serious medical condition that can cause the irreversible expansion of the abdominal aorta, which can lead to ruptures that are associated with up to 80% mortality. Currently, surgical and interventional procedures are the only treatment options available for treating abdominal aortic aneurysm patients. In this review, we focus on the upstream and downstream molecules of the microRNA-related signaling pathways and discuss the roles, mechanisms, and targets of microRNAs in abdominal aortic aneurysm modulation to provide novel insights for precise and targeted drug therapy for the vast number of abdominal aortic aneurysm patients. RECENT FINDINGS Recent studies have highlighted that microRNAs, which are emerging as novel regulators of gene expression, are involved in the biological activities of regulating abdominal aortic aneurysms. Accumulating studies suggested that microRNAs modulate abdominal aortic aneurysm development through various signaling pathways that are yet to be comprehensively summarized. A total of six signaling pathways (NF-κB signaling pathway, PI3K/AKT signaling pathway, MAPK signaling pathway, TGF-β signaling pathway, Wnt signaling pathway, and P53/P21 signaling pathway), and a total of 19 miRNAs are intimately associated with the biological properties of abdominal aortic aneurysm through targeting various essential molecules. MicroRNAs modulate the formation, progression, and rupture of abdominal aortic aneurysm by regulating smooth muscle cell proliferation and phenotype change, vascular inflammation and endothelium function, and extracellular matrix remodeling. Because of the broad crosstalk among signaling pathways, a comprehensive analysis of miRNA-mediated signaling pathways is necessary to construct a well-rounded upstream and downstream regulatory network for future basic and clinical research of AAA therapy.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Ke Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Yuanrui Gu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Yanxia Tu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| |
Collapse
|
4
|
de Vries PS, Conomos MP, Singh K, Nicholson CJ, Jain D, Hasbani NR, Jiang W, Lee S, Lino Cardenas CL, Lutz SM, Wong D, Guo X, Yao J, Young EP, Tcheandjieu C, Hilliard AT, Bis JC, Bielak LF, Brown MR, Musharoff S, Clarke SL, Terry JG, Palmer ND, Yanek LR, Xu H, Heard-Costa N, Wessel J, Selvaraj MS, Li RH, Sun X, Turner AW, Stilp AM, Khan A, Newman AB, Rasheed A, Freedman BI, Kral BG, McHugh CP, Hodonsky C, Saleheen D, Herrington DM, Jacobs DR, Nickerson DA, Boerwinkle E, Wang FF, Heiss G, Jun G, Kinney GL, Sigurslid HH, Doddapaneni H, Hall IM, Bensenor IM, Broome J, Crapo JD, Wilson JG, Smith JA, Blangero J, Vargas JD, Mosquera JV, Smith JD, Viaud-Martinez KA, Ryan KA, Young KA, Taylor KD, Lange LA, Emery LS, Bittencourt MS, Budoff MJ, Montasser ME, Yu M, Mahaney MC, Mahamdeh MS, Fornage M, Franceschini N, Lotufo PA, Natarajan P, Wong Q, Mathias RA, Gibbs RA, Do R, Mehran R, Tracy RP, Kim RW, Nelson SC, Damrauer SM, Kardia SL, Rich SS, Fuster V, Napolioni V, Zhao W, Tian W, Yin X, Min YI, Manning AK, Peloso G, Kelly TN, O’Donnell CJ, Morrison AC, Curran JE, Zapol WM, Bowden DW, Becker LC, Correa A, Mitchell BD, Psaty BM, Carr JJ, Pereira AC, Assimes TL, Stitziel NO, Hokanson JE, Laurie CA, Rotter JI, Vasan RS, Post WS, Peyser PA, Miller CL, Malhotra R. Whole-genome sequencing uncovers two loci for coronary artery calcification and identifies ARSE as a regulator of vascular calcification. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1159-1172. [PMID: 38817323 PMCID: PMC11138106 DOI: 10.1038/s44161-023-00375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/25/2023] [Indexed: 06/01/2024]
Abstract
Coronary artery calcification (CAC) is a measure of atherosclerosis and a well-established predictor of coronary artery disease (CAD) events. Here we describe a genome-wide association study (GWAS) of CAC in 22,400 participants from multiple ancestral groups. We confirmed associations with four known loci and identified two additional loci associated with CAC (ARSE and MMP16), with evidence of significant associations in replication analyses for both novel loci. Functional assays of ARSE and MMP16 in human vascular smooth muscle cells (VSMCs) demonstrate that ARSE is a promoter of VSMC calcification and VSMC phenotype switching from a contractile to a calcifying or osteogenic phenotype. Furthermore, we show that the association of variants near ARSE with reduced CAC is likely explained by reduced ARSE expression with the G allele of enhancer variant rs5982944. Our study highlights ARSE as an important contributor to atherosclerotic vascular calcification, and a potential drug target for vascular calcific disease.
Collapse
Affiliation(s)
- Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew P. Conomos
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Kuldeep Singh
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Christopher J. Nicholson
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Deepti Jain
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Natalie R. Hasbani
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Wanlin Jiang
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Sujin Lee
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Christian L Lino Cardenas
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Sharon M. Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR)
Center, Department of Population Medicine, Harvard Medical School and Harvard
Pilgrim Health Care Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of
Public Health, Boston, MA, USA
| | - Doris Wong
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Erica P. Young
- Cardiovascular Division, Department of Internal Medicine,
Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine Tcheandjieu
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, USA
| | - Austin T. Hilliard
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA,
USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of
Medicine, University of Washington, Seattle, WA, USA
| | - Lawrence F. Bielak
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Michael R. Brown
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Shaila Musharoff
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Genetics, Stanford University School of
Medicine, Stanford, CA, USA
| | - Shoa L. Clarke
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, USA
| | - James G. Terry
- Department of Radiology, Vanderbilt Translational and
Clinical Cardiovascular Research Center, Vanderbilt University Medical Center,
Nashville, TN, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of
Medicine, Winston-Salem, NC, USA
| | - Lisa R. Yanek
- Division of General Internal Medicine, Department of
Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huichun Xu
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
| | - Nancy Heard-Costa
- Boston University School of Medicine, Boston, MA,
USA
- Boston University and National Heart, Lung, and Blood
Institute’s Framingham Heart Study, Framingham, MA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Fairbanks School of Public
Health, Indiana University, Indianapolis, IN, USA
- Diabetes Translational Research Center, Indiana
University, Indianapolis, IN, USA
| | - Margaret Sunitha Selvaraj
- Cardiovascular Research Center and Center for Genomic
Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad
Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston,
MA, USA
| | - Rebecca H. Li
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Xiao Sun
- School of Public Health and Tropical Medicine, Department
of Epidemiology, Tulane University, New Orleans, LA, USA
- College of Medicine, Department of Medicine, Division of
Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Adrienne M. Stilp
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Alyna Khan
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Anne B. Newman
- Department of Epidemiology, Graduate School of Public
Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Asif Rasheed
- Center For Non-Communicable Diseases, Karachi,
Pakistan
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Brian G. Kral
- Division of Cardiology, Department of Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD, USA
| | - Caitlin P. McHugh
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Chani Hodonsky
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Danish Saleheen
- Center For Non-Communicable Diseases, Karachi,
Pakistan
- Department of Medicine, Columbia University Irving
Medical Center, New York, NY, USA
- Department of Cardiology, Columbia University Irving
Medical Center, New York, NY, USA
| | - David M. Herrington
- Department of Internal Medicine, Section of
Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC,
USA
| | - David R. Jacobs
- Division of Epidemiology and Community Health, University
of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Deborah A. Nickerson
- Department of Genome Sciences, University of Washington,
Seattle, WA, USA
- Northwest Genomics Center, University of Washington,
Seattle, WA, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of
Medicine, Houston, TX, USA
| | - Fei Fei Wang
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global
Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Goo Jun
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Greg L. Kinney
- Department of Epidemiology, Colorado School of Public
Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Haakon H. Sigurslid
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | | | - Ira M. Hall
- Yale Center for Genomic Health, Yale School of Medicine,
New Haven, CT, USA
| | - Isabela M. Bensenor
- Center for Clinical and Epidemiological Research,
University Hospital, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Jai Broome
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - James D. Crapo
- Department of Medicine, National Jewish Health, Denver,
CO, USA
| | - James G. Wilson
- Division of Cardiology, Beth Israel Deaconess Medical
Center, Boston, MA, USA
| | - Jennifer A. Smith
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research,
University of Michigan, Ann Arbor, MI, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio
Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of
Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Jose D. Vargas
- Medstar Heart and Vascular Institute, Medstar Georgetown
University Hospital, Washington, DC, USA
| | - Jose Verdezoto Mosquera
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Joshua D. Smith
- Department of Genome Sciences, University of Washington,
Seattle, WA, USA
- Northwest Genomics Center, University of Washington,
Seattle, WA, USA
| | | | - Kathleen A. Ryan
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
| | - Kendra A. Young
- Department of Epidemiology, Colorado School of Public
Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie A. Lange
- Department of Medicine, University of Colorado Denver,
Anschutz Medical Campus, Aurora, CO, USA
| | - Leslie S. Emery
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Marcio S. Bittencourt
- Center for Clinical and Epidemiological Research,
University Hospital, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Matthew J. Budoff
- Department of Medicine, The Lundquist Institute for
Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - May E. Montasser
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
| | - Miao Yu
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Michael C. Mahaney
- Department of Human Genetics, University of Texas Rio
Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of
Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Mohammed S Mahamdeh
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
- Institute of Molecular Medicine, McGovern Medical School,
The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global
Public health, University of North Carolina, Chapel Hill, NC, USA
| | - Paulo A. Lotufo
- Center for Clinical and Epidemiological Research,
University Hospital, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic
Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad
Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston,
MA, USA
| | - Quenna Wong
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Rasika A. Mathias
- Division of General Internal Medicine, Department of
Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Allergy and Clinical Immunology, Department
of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of
Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor
College of Medicine, Houston, TX, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine,
Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School
of Medicine at Mount Sinai, New York, NY, USA
| | - Roxana Mehran
- Icahn School of Medicine at Mount Sinai, New York, NY,
USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Robert
Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Sarah C. Nelson
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Scott M. Damrauer
- Corporal Michael J. Crescenz VA Medical Center,
Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon L.R. Kardia
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares
Carlos III, Madrid, Spain
- Mount Sinai Heart Center, New York, NY, USA
| | - Valerio Napolioni
- Genomic And Molecular Epidemiology (GAME) Lab, School of
Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Wei Zhao
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Wenjie Tian
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical
Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Yuan-I Min
- Jackson Heart Study, Department of Medicine, University
of Mississippi Medical Center, Jackson, MS, USA
| | - Alisa K. Manning
- Clinical and Translation Epidemiology Unit, Department of
Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical and Population
Genetics, Broad Institute, Cambridge, MA, USA
| | - Gina Peloso
- Department of Biostatistics, Boston University School of
Public Health, Boston, MA, USA
| | - Tanika N. Kelly
- College of Medicine, Department of Medicine, Division of
Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Christopher J. O’Donnell
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital,
Boston, MA, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Joanne E. Curran
- Department of Human Genetics, University of Texas Rio
Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of
Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Warren M. Zapol
- Department of Anesthesia, Critical Care and Pain Medicine
at Massachusetts General Hospital, Boston, MA, USA
| | - Donald W. Bowden
- Department of Biochemistry, Wake Forest School of
Medicine, Winston-Salem, NC, USA
| | - Lewis C. Becker
- Division of Cardiology, Department of Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University
of Mississippi Medical Center, Jackson, MS, USA
- Department of Population Health Science, University of
Mississippi Medical Center, Jackson, MS, USA
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
- Geriatrics Research and Education Clinical Center,
Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of
Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington,
Seattle, WA, USA
- Department of Health Services, University of Washington,
Seattle, WA, USA
| | - John Jeffrey Carr
- Department of Radiology, Vanderbilt Translational and
Clinical Cardiovascular Research Center, Vanderbilt University Medical Center,
Nashville, TN, USA
| | - Alexandre C. Pereira
- Department of Genetics, Harvard Medical School, Boston,
MA, USA
- Laboratory of Genetics and Molecular Cardiology, Heart
Institute, University of São Paulo, São Paulo, Brazil
| | - Themistocles L. Assimes
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, USA
| | - Nathan O. Stitziel
- Cardiovascular Division, Department of Internal Medicine,
Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of
Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School
of Medicine, St. Louis, MO, USA
| | - John E. Hokanson
- Department of Epidemiology, Colorado School of Public
Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cecelia A. Laurie
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ramachandran S. Vasan
- Boston University and National Heart, Lung, and Blood
Institute’s Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of
Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of
Public Health, Boston, MA, USA
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patricia A. Peyser
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Rajeev Malhotra
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| |
Collapse
|
5
|
Wang G, Luo Y, Gao X, Liang Y, Yang F, Wu J, Fang D, Luo M. MicroRNA regulation of phenotypic transformations in vascular smooth muscle: relevance to vascular remodeling. Cell Mol Life Sci 2023; 80:144. [PMID: 37165163 PMCID: PMC11071847 DOI: 10.1007/s00018-023-04793-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023]
Abstract
Alterations in the vascular smooth muscle cells (VSMC) phenotype play a critical role in the pathogenesis of several cardiovascular diseases, including hypertension, atherosclerosis, and restenosis after angioplasty. MicroRNAs (miRNAs) are a class of endogenous noncoding RNAs (approximately 19-25 nucleotides in length) that function as regulators in various physiological and pathophysiological events. Recent studies have suggested that aberrant miRNAs' expression might underlie VSMC phenotypic transformation, appearing to regulate the phenotypic transformations of VSMCs by targeting specific genes that either participate in the maintenance of the contractile phenotype or contribute to the transformation to alternate phenotypes, and affecting atherosclerosis, hypertension, and coronary artery disease by altering VSMC proliferation, migration, differentiation, inflammation, calcification, oxidative stress, and apoptosis, suggesting an important regulatory role in vascular remodeling for maintaining vascular homeostasis. This review outlines recent progress in the discovery of miRNAs and elucidation of their mechanisms of action and functions in VSMC phenotypic regulation. Importantly, as the literature supports roles for miRNAs in modulating vascular remodeling and for maintaining vascular homeostasis, this area of research will likely provide new insights into clinical diagnosis and prognosis and ultimately facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Gang Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yulin Luo
- GCP Center, Affiliated Hospital (Traditional Chinese Medicine) of Southwest Medical University, Luzhou, China
| | - Xiaojun Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Feifei Yang
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
6
|
Pan Y, Jiang Z, Ye Y, Zhu D, Li N, Yang G, Wang Y. Role and Mechanism of BMP4 in Regenerative Medicine and Tissue Engineering. Ann Biomed Eng 2023:10.1007/s10439-023-03173-6. [PMID: 37014581 DOI: 10.1007/s10439-023-03173-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/21/2023] [Indexed: 04/05/2023]
Abstract
Bone morphogenetic protein 4 (BMP4) is emerging as a promising cytokine for regenerative medicine and tissue engineering. BMP4 has been shown to promote the regeneration of teeth, periodontal tissue, bone, cartilage, the thymus, hair, neurons, nucleus pulposus, and adipose tissue, as well as the formation of skeletal myotubes and vessels. BMP4 can also contribute to the formation of tissues in the heart, lung, and kidney. However, there are certain deficiencies, including the insufficiency of the mechanism of BMP4 in some fields and an appropriate carrier of BMP4 for clinical use. There has also been a lack of in vivo experiments and orthotopic transplantation studies in some fields. BMP4 has great distance from the clinical application. Therefore, there are many BMP4-related studies waiting to be explored. This review mainly discusses the effects, mechanisms, and applications of BMP4 in regenerative medicine and tissue engineering over the last 10 years in various domains and possible improvements. BMP4 has shown great potential in regenerative medicine and tissue engineering. The research of BMP4 has broad development space and great value.
Collapse
Affiliation(s)
- Yiqi Pan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Yuer Ye
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
7
|
Direct targeting of DOCK4 by miRNA-181d in oxygen-glucose deprivation/reoxygenation-mediated neuronal injury. Lipids Health Dis 2023; 22:34. [PMID: 36882763 PMCID: PMC9990210 DOI: 10.1186/s12944-023-01794-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
The miRNA-181 (miR-181) family regulates neuronal persistence during cerebral ischemia/reperfusion injury (CI/RI). Since the effect of miR-181d on CI/RI has never been studied, the current work sought to determine the involvement of miR-181d in neuronal apoptosis after brain I/R injury. To replicate in vivo and in vitro CI/RI, a transient middle cerebral artery occlusion (tMCAO) model in rats and an oxygen-glucose deficiency/reoxygenation (OGD/R) model in neuro 2A cells were developed. In both in vivo and in vitro stroke models, the expression of miR-181d was considerably higher. miR-181d suppression reduced apoptosis and oxidative stress in OGD/R-treated neuroblastoma cells, but miR-181d overexpression increased both. Furthermore, it was observed that miR-181d has a direct target in dedicator of cytokinesis 4 (DOCK4). The overexpression of DOCK4 partially overcame cell apoptosis and oxidative stress induced by miR-181d upregulation and OGD/R injury. Furthermore, the DOCK4 rs2074130 mutation was related to lower DOCK4 levels in ischemic stroke (IS) peripheral blood and higher susceptibility to IS. These findings suggest that downregulating miR-181d protects neurons from ischemic damage by targeting DOCK4, implying that the miR-181d/DOCK4 axis might be a novel therapeutic target for IS.
Collapse
|
8
|
Baig MS, Deepanshu, Prakash P, Alam P, Krishnan A. In silico analysis reveals hypoxia-induced miR-210-3p specifically targets SARS-CoV-2 RNA. J Biomol Struct Dyn 2023; 41:12305-12327. [PMID: 36752331 DOI: 10.1080/07391102.2023.2175255] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/01/2023] [Indexed: 02/09/2023]
Abstract
Human coronaviruses (HCoVs) until the emergence of SARS in 2003 were associated with mild cold and upper respiratory tract infections. The ongoing pandemic caused by SARS-CoV-2 has enhanced the potential for infection and transmission as compared to other known members of this family. MicroRNAs (miRNA) are 21-25 nucleotides long non-coding RNA that bind to 3' UTR of genes and regulate almost every aspect of cellular function. Several human miRNAs have been known to target viral genomes, mostly to downregulate their expression and sometimes to upregulate also. In some cases, host miRNAs could be sequestered by the viral genome to create a condition for favourable virus existence. The ongoing SARS CoV-2 pandemic is unique based on its transmissibility and severity and we hypothesised that there could be a unique mechanism for its pathogenesis. In this study, we exploited in silico approach to identify human respiratory system-specific miRNAs targeting the viral genome of three highly pathogenic HCoVs (SARS-CoV-2 Wuhan strain, SARS-CoV, and MERS-CoV) and three low pathogenic HCoVs (OC43, NL63, and HKU1). We identified ten common microRNAs that target all HCoVs studied here. In addition, we identified unique miRNAs which targeted specifically one particular HCoV. miR-210-3p was the single unique lung-specific miRNA, which was found to target the NSP3, NSP4, and NSP13 genes of SARS-CoV-2. Further miR-210-NSP3, miR-210-NSP4, and miR-210-NSP13 SARS-CoV-2 duplexes were docked with the hAGO2 protein (PDB ID 4F3T) which showed Z-score values of -1.9, -1.7, and -1.6, respectively. The role of miR-210-3p as master hypoxia regulator and inflammation regulation may be important for SARS-CoV-2 pathogenesis. Overall, this analysis advocates that miR-210-3p be investigated experimentally in SARS-CoV-2 infection.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Deepanshu
- Department of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Prem Prakash
- Department of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Pravej Alam
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Anuja Krishnan
- Department of Molecular Medicine, Jamia Hamdard, New Delhi, India
| |
Collapse
|
9
|
Breulmann FL, Hatt LP, Schmitz B, Wehrle E, Richards RG, Della Bella E, Stoddart MJ. Prognostic and therapeutic potential of microRNAs for fracture healing processes and non-union fractures: A systematic review. Clin Transl Med 2023; 13:e1161. [PMID: 36629031 PMCID: PMC9832434 DOI: 10.1002/ctm2.1161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Approximately 10% of all bone fractures result in delayed fracture healing or non-union; thus, the identification of biomarkers and prognostic factors is of great clinical interest. MicroRNAs (miRNAs) are known to be involved in the regulation of the bone healing process and may serve as functional markers for fracture healing. AIMS AND METHODS This systematic review aimed to identify common miRNAs involved in fracture healing or non-union fractures using a qualitative approach. A systematic literature search was performed with the keywords 'miRNA and fracture healing' and 'miRNA and non-union fracture'. Any original article investigating miRNAs in fracture healing or non-union fractures was screened. Eventually, 82 studies were included in the qualitative analysis for 'miRNA and fracture healing', while 19 were selected for the 'miRNA and fracture non-union' category. RESULTS AND CONCLUSIONS Out of 151 miRNAs, miR-21, miR-140 and miR-214 were the most investigated miRNAs in fracture healing in general. miR-31-5p, miR-221 and miR-451-5p were identified to be regulated specifically in non-union fractures. Large heterogeneity was detected between studies investigating the role of miRNAs in fracture healing or non-union in terms of patient population, sample types and models used. Nonetheless, our approach identified some miRNAs with the potential to serve as biomarkers for non-union fractures, including miR-31-5p, miR-221 and miR-451-5p. We provide a discussion of involved pathways and suggest on alignment of future research in the field.
Collapse
Affiliation(s)
- Franziska Lioba Breulmann
- AO Research Institute DavosDavos PlatzSwitzerland
- Department of Orthopedic Sports MedicineKlinikum Rechts der IsarTechnical University of MunichMunichGermany
| | - Luan Phelipe Hatt
- AO Research Institute DavosDavos PlatzSwitzerland
- Institute for BiomechanicsETH ZürichZurichSwitzerland
| | - Boris Schmitz
- Department of Rehabilitation SciencesFaculty of HealthUniversity of Witten/HerdeckeWittenGermany
- DRV Clinic KönigsfeldCenter for Medical RehabilitationEnnepetalGermany
| | - Esther Wehrle
- AO Research Institute DavosDavos PlatzSwitzerland
- Institute for BiomechanicsETH ZürichZurichSwitzerland
| | - Robert Geoff Richards
- AO Research Institute DavosDavos PlatzSwitzerland
- Faculty of MedicineMedical Center‐Albert‐Ludwigs‐University of FreiburgAlbert‐Ludwigs‐University of FreiburgFreiburgGermany
| | | | - Martin James Stoddart
- AO Research Institute DavosDavos PlatzSwitzerland
- Faculty of MedicineMedical Center‐Albert‐Ludwigs‐University of FreiburgAlbert‐Ludwigs‐University of FreiburgFreiburgGermany
| |
Collapse
|
10
|
Wang G, Wang Z, Wei S, Wang D, Ji A, Zhang W, Sun Q. A new strategy for distinguishing menstrual blood from peripheral blood by the miR-451a/miR-21-5p ratio. Forensic Sci Int Genet 2021; 57:102654. [PMID: 34954475 DOI: 10.1016/j.fsigen.2021.102654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/12/2021] [Accepted: 12/10/2021] [Indexed: 11/04/2022]
Abstract
Distinction between menstrual blood and peripheral blood is vital for forensic casework, as it could provide strong evidence to figure out the nature of some criminal cases. However, to date no single blood-specific gene, including the most variable microRNAs (miRNAs) could work well in identification of blood source. In this study, we developed a new strategy for identification of human blood samples by using the copy number ratios of miR-451a to miR-21-5p based on 133 samples, including 56 menstrual blood and 47 peripheral blood, as well as 30 non-blood samples of saliva (10), semen (10) and vaginal secretion (10). The cut-off value and efficacy of the identification strategy were determined through receiver operating characteristic (ROC) analysis. Our results showed that when the miR-451a/miR-21-5p ratio below 0.929, the sample should be non-blood. In contrast, when the miR-451a/miR-21-5p ratio above 0.929 and below 10.201, the sample should be menstrual blood; and when this ratio above 10.201, the sample should be peripheral blood. External validation using 86 samples (62 menstrual blood and 24 peripheral blood samples) fully supported this strategy with the 100% sensitivity and 100% specificity. We confirmed that this result accuracy was not affected by various potential confounding factors of samples and different experimental platforms. We showed that 0.2 ng of total RNA from menstrual blood and peripheral blood was sufficient for qPCR quantification. In conclusion, our results provide an accurate reference to distinguish menstrual blood from peripheral blood for forensic authentication.
Collapse
Affiliation(s)
- Guoli Wang
- MPS's Key Laboratory of Forensic Genetics, National Engineering Laboratory for Crime Scene Evidence Investigation and Examination, Institute of Forensic Science, Ministry of Public Security (MPS), Beijing 100038, China; Marine College, Shandong University, Weihai 264209, Shandong, China
| | - Zhe Wang
- MPS's Key Laboratory of Forensic Genetics, National Engineering Laboratory for Crime Scene Evidence Investigation and Examination, Institute of Forensic Science, Ministry of Public Security (MPS), Beijing 100038, China
| | - Sunxiang Wei
- MPS's Key Laboratory of Forensic Genetics, National Engineering Laboratory for Crime Scene Evidence Investigation and Examination, Institute of Forensic Science, Ministry of Public Security (MPS), Beijing 100038, China
| | - Di Wang
- National Institute of Metrology, Beijing 100029, China
| | - Anquan Ji
- MPS's Key Laboratory of Forensic Genetics, National Engineering Laboratory for Crime Scene Evidence Investigation and Examination, Institute of Forensic Science, Ministry of Public Security (MPS), Beijing 100038, China
| | - Wei Zhang
- Marine College, Shandong University, Weihai 264209, Shandong, China.
| | - Qifan Sun
- MPS's Key Laboratory of Forensic Genetics, National Engineering Laboratory for Crime Scene Evidence Investigation and Examination, Institute of Forensic Science, Ministry of Public Security (MPS), Beijing 100038, China.
| |
Collapse
|
11
|
An Overview of miRNAs Involved in PASMC Phenotypic Switching in Pulmonary Hypertension. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5765029. [PMID: 34660794 PMCID: PMC8516547 DOI: 10.1155/2021/5765029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022]
Abstract
Pulmonary hypertension (PH) is occult, with no distinctive clinical manifestations and a poor prognosis. Pulmonary vascular remodelling is an important pathological feature in which pulmonary artery smooth muscle cells (PASMCs) phenotypic switching plays a crucial role. MicroRNAs (miRNAs) are a class of evolutionarily highly conserved single-stranded small noncoding RNAs. An increasing number of studies have shown that miRNAs play an important role in the occurrence and development of PH by regulating PASMCs phenotypic switching, which is expected to be a potential target for the prevention and treatment of PH. miRNAs such as miR-221, miR-15b, miR-96, miR-24, miR-23a, miR-9, miR-214, and miR-20a can promote PASMCs phenotypic switching, while such as miR-21, miR-132, miR-449, miR-206, miR-124, miR-30c, miR-140, and the miR-17~92 cluster can inhibit it. The article reviews the research progress on growth factor-related miRNAs and hypoxia-related miRNAs that mediate PASMCs phenotypic switching in PH.
Collapse
|
12
|
Benson CE, Southgate L. The DOCK protein family in vascular development and disease. Angiogenesis 2021; 24:417-433. [PMID: 33548004 PMCID: PMC8292242 DOI: 10.1007/s10456-021-09768-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/06/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The vascular network is established and maintained through the processes of vasculogenesis and angiogenesis, which are tightly regulated during embryonic and postnatal life. The formation of a functional vasculature requires critical cellular mechanisms, such as cell migration, proliferation and adhesion, which are dependent on the activity of small Rho GTPases, controlled in part by the dedicator of cytokinesis (DOCK) protein family. Whilst the majority of DOCK proteins are associated with neuronal development, a growing body of evidence has indicated that members of the DOCK family may have key functions in the control of vasculogenic and angiogenic processes. This is supported by the involvement of several angiogenic signalling pathways, including chemokine receptor type 4 (CXCR4), vascular endothelial growth factor (VEGF) and phosphatidylinositol 3-kinase (PI3K), in the regulation of specific DOCK proteins. This review summarises recent progress in understanding the respective roles of DOCK family proteins during vascular development. We focus on existing in vivo and in vitro models and known human disease phenotypes and highlight potential mechanisms of DOCK protein dysfunction in the pathogenesis of vascular disease.
Collapse
Affiliation(s)
- Clare E Benson
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St. George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Laura Southgate
- Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St. George's University of London, Cranmer Terrace, London, SW17 0RE, UK. .,Department of Medical & Molecular Genetics, Faculty of Life Sciences & Medicine, King's College London, London, SE1 9RT, UK.
| |
Collapse
|
13
|
Tang Y, Fan W, Zou B, Yan W, Hou Y, Kwabena Agyare O, Jiang Z, Qu S. TGF-β signaling and microRNA cross-talk regulates abdominal aortic aneurysm progression. Clin Chim Acta 2020; 515:90-95. [PMID: 33388307 DOI: 10.1016/j.cca.2020.12.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
Abdominal aortic aneurysms (AAA) are permanent and irreversible local dilatations of the abdominal aortic wall. Recent data indicate that the transforming growth factor-beta (TGF-β) signaling pathway exerts a protective effect on the development of AAA. Some dysregulated microRNAs (miRNA) also appear involved in the expansion of AAA and miRNA-based therapeutics have been shown to effectively inhibit this process. New evidence has revealed that TGF-β signaling and miRNA interaction may of physiologic and pathophysiologic significance including the progression of AAA. As such, miRNA that regulate TGF-β signaling may hold promise as potential therapeutic targets. This review explores potential crosstalk between TGF-β signaling and miRNA in AAA in order improve our understanding of this pathology and explore development of potential therapeutic targets.
Collapse
Affiliation(s)
- Ying Tang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Wenjing Fan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Emergency Department, The Second Affiliated Hospital, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Bu Zou
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Wei Yan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Yangfeng Hou
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Oware Kwabena Agyare
- International College, Hengyang Medical School, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhisheng Jiang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
14
|
Rubattu S, Stanzione R, Cotugno M, Bianchi F, Marchitti S, Forte M. Epigenetic control of natriuretic peptides: implications for health and disease. Cell Mol Life Sci 2020; 77:5121-5130. [PMID: 32556416 PMCID: PMC11105024 DOI: 10.1007/s00018-020-03573-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/27/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022]
Abstract
The natriuretic peptides (NPs) family, including a class of hormones and their receptors, is largely known for its beneficial effects within the cardiovascular system to preserve regular functions and health. The concentration level of each component of the family is of crucial importance to guarantee a proper control of both systemic and local cardiovascular functions. A fine equilibrium between gene expression, protein secretion and clearance is needed to achieve the final optimal level of NPs. To this aim, the regulation of gene expression and translation plays a key role. In this regard, we know the existence of fine regulatory mechanisms, the so-called epigenetic mechanisms, which target many genes at either the promoter or the 3'UTR region to inhibit or activate their expression. The gene encoding ANP (NPPA) is regulated by histone modifications, DNA methylation, distinct microRNAs and a natural antisense transcript (NPPA-AS1) with consequent implications for both health and disease conditions. Notably, ANP modulates microRNAs on its own. Histone modifications of BNP gene (NPPB) are associated with several cardiomyopathies. The proBNP processing is regulated by miR30-GALNT1/2 axis. Among other components of the NPs family, CORIN, NPRA, NPRC and NEP may undergo epigenetic regulation. A better understanding of the epigenetic control of the NPs family will allow to gain more insights on the pathological basis of common cardiovascular diseases and to identify novel therapeutic targets. The present review article aims to discuss the major achievements obtained so far with studies on the epigenetic modulation of the NPs family.
Collapse
Affiliation(s)
- Speranza Rubattu
- IRCCS Neuromed, Pozzilli, Isernia, Italy.
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
15
|
Usui-Kawanishi F, Takahashi M, Sakai H, Suto W, Kai Y, Chiba Y, Hiraishi K, Kurahara LH, Hori M, Inoue R. Implications of immune-inflammatory responses in smooth muscle dysfunction and disease. J Smooth Muscle Res 2020; 55:81-107. [PMID: 32023567 PMCID: PMC6997890 DOI: 10.1540/jsmr.55.81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In the past few decades, solid evidence has been accumulated for the pivotal significance
of immunoinflammatory processes in the initiation, progression, and exacerbation of many
diseases and disorders. This groundbreaking view came from original works by Ross who
first described that excessive inflammatory-fibroproliferative response to various forms
of insult to the endothelium and smooth muscle of the artery wall is essential for the
pathogenesis of atherosclerosis (Ross, Nature 1993; 362(6423): 801–9). It is now widely
recognized that both innate and adaptive immune reactions are avidly involved in the
inflammation-related remodeling of many tissues and organs. When this state persists,
irreversible fibrogenic changes would occur often culminating in fatal insufficiencies of
many vital parenchymal organs such as liver, lung, heart, kidney and intestines. Thus,
inflammatory diseases are becoming the common life-threatening risk for and urgent concern
about the public health in developed countries (Wynn et al., Nature Medicine 2012; 18(7):
1028–40). Considering this timeliness, we organized a special symposium entitled
“Implications of immune/inflammatory responses in smooth muscle dysfunction and disease”
in the 58th annual meeting of the Japan Society of Smooth Muscle Research. This symposium
report will provide detailed synopses of topics presented in this symposium; (1) the role
of inflammasome in atherosclerosis and abdominal aortic aneurysms by Fumitake
Usui-Kawanishi and Masafumi Takahashi; (2) Mechanisms underlying the pathogenesis of
hyper-contractility of bronchial smooth muscle in allergic asthma by Hiroyasu Sakai,
Wataru Suto, Yuki Kai and Yoshihiko Chiba; (3) Vascular remodeling in pulmonary arterial
hypertension by Keizo Hiraishi, Lin Hai Kurahara and Ryuji Inoue.
Collapse
Affiliation(s)
- Fumitake Usui-Kawanishi
- Division of Biopharmaceutical Engineering, Department of Pharmaceutical Engineering, Toyoma Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan.,Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, 3311-159 Yakushiji, Shimono-shi, Tochigi 329-0498, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, 3311-159 Yakushiji, Shimono-shi, Tochigi 329-0498, Japan
| | - Hiroyasu Sakai
- Department of Analytical Pathophysiology, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Wataru Suto
- Department of Physiology and Molecular Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yuki Kai
- Department of Analytical Pathophysiology, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Keizo Hiraishi
- Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Lin Hai Kurahara
- Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.,Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, 1750-1 Ido, Miki-machi, Kida-gun, Kagawa 761-0793, Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
16
|
Park N, Kang H. BMP-Induced MicroRNA-101 Expression Regulates Vascular Smooth Muscle Cell Migration. Int J Mol Sci 2020; 21:ijms21134764. [PMID: 32635504 PMCID: PMC7369869 DOI: 10.3390/ijms21134764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022] Open
Abstract
Proliferation and migration of vascular smooth muscle cells (VSMCs) are implicated in blood vessel development, maintenance of vascular homeostasis, and pathogenesis of vascular disorders. MicroRNAs (miRNAs) mediate the regulation of VSMC functions in response to microenvironmental signals. Because a previous study reported that miR-101, a tumor-suppressive miRNA, is a critical regulator of cell proliferation in vascular disease, we hypothesized that miR-101 controls important cellular processes in VSMCs. The present study aimed to elucidate the effects of miR-101 on VSMC function and its molecular mechanisms. We revealed that miR-101 regulates VSMC proliferation and migration. We showed that miR-101 expression is induced by bone morphogenetic protein (BMP) signaling, and we identified dedicator of cytokinesis 4 (DOCK4) as a novel target of miR-101. Our results suggest that the BMP–miR-101–DOCK4 axis mediates the regulation of VSMC function. Our findings help further the understanding of vascular physiology and pathology.
Collapse
Affiliation(s)
- Nanju Park
- Department of Life Sciences, Incheon National University, Incheon 22012, Korea;
| | - Hara Kang
- Department of Life Sciences, Incheon National University, Incheon 22012, Korea;
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea
- Correspondence: ; Tel.: +82-32-835-8238; Fax: +82-32-835-0763
| |
Collapse
|
17
|
Abstract
Vascular smooth muscle cells (VSMCs) are a unique cell type that has unusual plasticity controlled by environmental stimuli. As an abnormal increase of VSMC proliferation is associated with various vascular diseases, tight regulation of VSMC phenotypes is essential for maintaining vascular homeostasis. Hypoxia is one environmental stress that stimulates VSMC proliferation. Emerging evidence has indicated that microRNAs (miRNAs) are critical regulators in the hypoxic responses of VSMCs. Therefore, we previously investigated miRNAs modulated by hypoxia in VSMCs and found that miR-1260b is one of the most upregulated miRNAs under hypoxia. However, the mechanism that underlies the regulation of VSMCs via miR-1260b in response to hypoxia has not been explored. Here we demonstrated that hypoxia-induced miR-1260b promotes VSMC proliferation. We also identified growth differentiation factor 11 (GDF11), a member of the TGF-β superfamily, as a novel target of miR-1260b. miR-1260b directly targets the 3’UTR of GDF11. Downregulation of GDF11 inhibited Smad signaling and consequently enhanced the proliferation of VSMCs. Our findings suggest that miR-1260b-mediated GDF11-Smad-dependent signaling is an essential regulatory mechanism in the proliferation of VSMCs, and this axis is modulated by hypoxia to promote abnormal VSMC proliferation. Therefore, our study unveils a novel function of miR-1260b in the pathological proliferation of VSMCs under hypoxia.
Collapse
Affiliation(s)
- Minhyeong Seong
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea
| |
Collapse
|
18
|
Ciebiera M, Włodarczyk M, Zgliczyński S, Łoziński T, Walczak K, Czekierdowski A. The Role of miRNA and Related Pathways in Pathophysiology of Uterine Fibroids-From Bench to Bedside. Int J Mol Sci 2020; 21:ijms21083016. [PMID: 32344726 PMCID: PMC7216240 DOI: 10.3390/ijms21083016] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Uterine fibroids (UFs) are the most common benign tumors of the female genital tract. Their prevalence usually is estimated at 30-40%, but may reach up to 70-80% in predisposed groups of women. UFs may cause various clinical issues which might constitute the major reason of the overall deterioration of the quality of life. The mechanisms leading to UFs formation and growth still remain poorly understood. The transformation of smooth muscle cells of the uterus into abnormal, immortal cells, capable of clonal division, is thought to be a starting point of all pathways leading to UF formation. Micro-ribonucleic acids (miRNAs) are non-coding single-stranded RNAs about 22 nucleotides in length, that regulate gene expression. One of recent advances in this field is the comprehension of the role of miRNAs in tumorigenesis. Alterations in the levels of miRNAs are related to the formation and growth of several tumors which show a distinct miRNA signature. The aim of this review is to summarize the current data about the role of miRNAs in the pathophysiology of UFs. We also discuss future directions in the miRNA research area with an emphasis on novel diagnostic opportunities or patient-tailored therapies. In our opinion data concerning the regulation of miRNA and its gene targets in the UFs are still insufficient in comparison with gynecological malignancies. The potential translational use of miRNA and derived technologies in the clinical care is at the early phase and needs far more evidence. However, it is one of the main areas of interest for the future as the use of miRNAs in the diagnostics and treatment of UFs is a new and exciting opportunity.
Collapse
Affiliation(s)
- Michał Ciebiera
- Second Department of Obstetrics and Gynecology, The Center of Postgraduate Medical Education, 01-809 Warsaw, Poland
- Correspondence: ; Tel.: +48-607-155-177
| | - Marta Włodarczyk
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Stanisław Zgliczyński
- Department of Internal Diseases and Endocrinology, Central Teaching Clinical Hospital, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Tomasz Łoziński
- Department of Obstetrics and Gynecology, Pro-Familia Hospital, 35-302 Rzeszów, Poland;
| | - Klaudia Walczak
- Students’ Scientific Association at the Department of Endocrinology, The Center of Postgraduate Medical Education, 01-809 Warsaw, Poland;
| | - Artur Czekierdowski
- Department of Gynecological Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland;
| |
Collapse
|
19
|
Malhotra R, Mauer AC, Lino Cardenas CL, Guo X, Yao J, Zhang X, Wunderer F, Smith AV, Wong Q, Pechlivanis S, Hwang SJ, Wang J, Lu L, Nicholson CJ, Shelton G, Buswell MD, Barnes HJ, Sigurslid HH, Slocum C, Rourke CO, Rhee DK, Bagchi A, Nigwekar SU, Buys ES, Campbell CY, Harris T, Budoff M, Criqui MH, Rotter JI, Johnson AD, Song C, Franceschini N, Debette S, Hoffmann U, Kälsch H, Nöthen MM, Sigurdsson S, Freedman BI, Bowden DW, Jöckel KH, Moebus S, Erbel R, Feitosa MF, Gudnason V, Thanassoulis G, Zapol WM, Lindsay ME, Bloch DB, Post WS, O'Donnell CJ. HDAC9 is implicated in atherosclerotic aortic calcification and affects vascular smooth muscle cell phenotype. Nat Genet 2019; 51:1580-1587. [PMID: 31659325 PMCID: PMC6858575 DOI: 10.1038/s41588-019-0514-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/16/2019] [Indexed: 01/16/2023]
Abstract
Aortic calcification is an important independent predictor of future cardiovascular events. We performed a genome-wide association meta-analysis to determine SNPs associated with the extent of abdominal aortic calcification (n = 9,417) or descending thoracic aortic calcification (n = 8,422). Two genetic loci, HDAC9 and RAP1GAP, were associated with abdominal aortic calcification at a genome-wide level (P < 5.0 × 10-8). No SNPs were associated with thoracic aortic calcification at the genome-wide threshold. Increased expression of HDAC9 in human aortic smooth muscle cells promoted calcification and reduced contractility, while inhibition of HDAC9 in human aortic smooth muscle cells inhibited calcification and enhanced cell contractility. In matrix Gla protein-deficient mice, a model of human vascular calcification, mice lacking HDAC9 had a 40% reduction in aortic calcification and improved survival. This translational genomic study identifies the first genetic risk locus associated with calcification of the abdominal aorta and describes a previously unknown role for HDAC9 in the development of vascular calcification.
Collapse
Affiliation(s)
- Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Andreas C Mauer
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Christian L Lino Cardenas
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiaoling Zhang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Department of Medicine (Biomedical Genetics Section), Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Florian Wunderer
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Albert V Smith
- Icelandic Heart Association, Kópavogur, Iceland
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Quenna Wong
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Sonali Pechlivanis
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Division of Intramural Research, Bethesda, MD, USA
| | - Judy Wang
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lingyi Lu
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher J Nicholson
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Georgia Shelton
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary D Buswell
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hanna J Barnes
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Haakon H Sigurslid
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Charles Slocum
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Caitlin O' Rourke
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - David K Rhee
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aranya Bagchi
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sagar U Nigwekar
- Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Emmanuel S Buys
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Matthew Budoff
- Division of Cardiology, Department of Medicine and Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Michael H Criqui
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Division of Intramural Research, Bethesda, MD, USA
| | - Ci Song
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Division of Intramural Research, Bethesda, MD, USA
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Stephanie Debette
- Inserm U1219, University of Bordeaux, Bordeaux, France
- Department of Neurology, University Hospital of Bordeaux, Bordeaux, France
| | - Udo Hoffmann
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Hagen Kälsch
- Department of Cardiology, Alfried Krupp Hospital, Essen, Germany
- Witten/Herdecke University, Witten, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain GmbH, University of Bonn, Bonn, Germany
| | | | | | | | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
- Centre for Urban Epidemiology, University Hospital Essen, Essen, Germany
| | - Raimund Erbel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
| | - Mary F Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - George Thanassoulis
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Warren M Zapol
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mark E Lindsay
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Donald B Bloch
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology; Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher J O'Donnell
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA.
- U.S. Department of Veterans Affairs, Boston, MA, USA.
| |
Collapse
|
20
|
Pravoverov K, Whiting K, Thapa S, Bushong T, Trang K, Lein PJ, Chandrasekaran V. MicroRNAs are Necessary for BMP-7-induced Dendritic Growth in Cultured Rat Sympathetic Neurons. Cell Mol Neurobiol 2019; 39:917-934. [PMID: 31104181 PMCID: PMC6713596 DOI: 10.1007/s10571-019-00688-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/14/2019] [Indexed: 01/28/2023]
Abstract
Neuronal connectivity is dependent on size and shape of the dendritic arbor. However, mechanisms controlling dendritic arborization, especially in the peripheral nervous system, are not completely understood. Previous studies have shown that bone morphogenetic proteins (BMPs) are important initiators of dendritic growth in peripheral neurons. In this study, we examined the hypothesis that post-transcriptional regulation mediated by microRNAs (miRNAs) is necessary for BMP-7-induced dendritic growth in these neurons. To examine the role of miRNAs in BMP-7-induced dendritic growth, microarray analyses was used to profile miRNA expression in cultured sympathetic neurons from the superior cervical ganglia of embryonic day 21 rat pups at 6 and 24 h after treatment with BMP-7 (50 ng/mL). Our data showed that BMP-7 significantly regulated the expression of 43 of the 762 miRNAs. Of the 43 miRNAs, 22 showed robust gene expression; 14 were upregulated by BMP-7 and 8 were downregulated by BMP-7. The expression profile for miR-335, miR-664-1*, miR-21, and miR-23b was confirmed using qPCR analyses. Functional studies using morphometric analyses of dendritic growth in cultured sympathetic neurons transfected with miRNA mimics and inhibitors indicated that miR-664-1*, miR-23b, and miR-21 regulated early stages of BMP-7-induced dendritic growth. In summary, our data provide evidence for miRNA-mediated post-transcriptional regulation as important downstream component of BMP-7 signaling during early stages of dendritic growth in sympathetic neurons.
Collapse
Affiliation(s)
- Kristina Pravoverov
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Katherine Whiting
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Slesha Thapa
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Trevor Bushong
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Karen Trang
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, 1089 Veterinary Medicine Drive, Davis, Davis, CA 95616
| | - Vidya Chandrasekaran
- Department of Biology, Saint Mary’s College of California, 1928 Saint Mary’s Road, Moraga, CA 94556.,Corresponding author: Vidya Chandrasekaran, Department of Biology, Saint Mary’s College of California, Moraga, CA 94556.
| |
Collapse
|
21
|
Silvério de Barros R, Dias GS, Paula do Rosario A, Paladino FV, Lopes GH, Campos AH. Gremlin-1 potentiates the dedifferentiation of VSMC in early stages of atherosclerosis. Differentiation 2019; 109:28-33. [PMID: 31494396 DOI: 10.1016/j.diff.2019.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/12/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMC) are highly specialized, and exhibit a contractile phenotype when mature and fully differentiated, being responsible for vessel homeostasis and blood pressure control. In response to pro-atherogenic stimuli VSMC alter their state of differentiation, increase proliferation and migration, resulting in SMC phenotypes ranging from contractile to synthetic. This variability is observed in cell morphology and expression level of marker genes for differentiation status. There is growing evidence that bone morphogenetic protein (BMP) signaling is involved in vascular diseases, including atherosclerosis. Here, we evaluated in vitro the role of specific agonists/antagonists belonging to the BMP pathway on dedifferentiation of VSMC harvested during early stages of atherosclerosis. RESULTS: Comparing primary VSMC isolated from aortas of susceptible ApoE-/- animals fed 8 weeks of western diet with their littermate controls fed usual diet, we observed that recombinant BMP4 was able to reduce SM22-alpha and alpha actin gene expression indicating dedifferentiation was under way. Unexpectedly, treatment with recombinant Gremlin-1, a known BMP antagonist, also reduced 4-6.5 folds gene expression of SM22-alpha, alpha-actin and, calponin, exclusively in VSMC from ApoE-/- animals, independently on the diet consumed. CONCLUSION: Our data show that BMP4 is capable of modulating of SM22-alpha and alpha actin gene expression, indicative of cell dedifferentiation in VSMC. Additionally, we report for first time that Gremlin-1 acts independently of the BMP pathway and selectively on VSMC from susceptible animals, reducing the expression of all genes evaluated.
Collapse
Affiliation(s)
- Renata Silvério de Barros
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Grazielle Suhett Dias
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Ana Paula do Rosario
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Fernanda Vieira Paladino
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | | | - Alexandre Holthausen Campos
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil.
| |
Collapse
|
22
|
Cordella M, Tabolacci C, Senatore C, Rossi S, Mueller S, Lintas C, Eramo A, D'Arcangelo D, Valitutti S, Facchiano A, Facchiano F. Theophylline induces differentiation and modulates cytoskeleton dynamics and cytokines secretion in human melanoma-initiating cells. Life Sci 2019; 230:121-131. [PMID: 31125565 DOI: 10.1016/j.lfs.2019.05.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/22/2022]
Abstract
AIMS Cutaneous melanoma is the most aggressive skin cancer, derived from neoplastic transformation of melanocytes. Since several evidences highlighted the importance of a hierarchical model of differentiation among cancer cells, closely related to resistance mechanisms and tumor relapse, we investigated the effects of theophylline (Theo), a methylxanthine commonly used in treatment of respiratory diseases, on melanoma cells with different degree of differentiation, including patient-derived melanoma-initiating cells. MATERIALS AND METHODS The antiproliferative and antimetastatic effects of Theo was demonstrated by cell counting, adhesion and migration assays on A375 and SK-MEL-30 cells. Further, Theo ability to reduce cell growth was highly significant in A375-derived spheroids and in two patient-derived melanoma-initiating cells (MICs). In order to identify pathways potentially involved in the antineoplastic properties of Theo, a comparative mass spectrometry proteomic analysis was used. Then, melanin content, tyrosinase and tissue transglutaminase activities as differentiation markers and actin re-organization through confocal microscopy were evaluated. Furthermore, a secretome profile of MICs after Theo treatments was performed by multiplex immunoassay. KEY FINDINGS Obtained results demonstrate inhibitory effects of Theo on melanoma cell proliferation and migration, mainly in MICs, together with the induction of differentiation parameters. Moreover, our data indicate that the known anti-melanoma effect of Theo is due also to its ability to interfere with cytoskeleton dynamics and to induce the secretion of inflammatory molecules involved in recruitment of immunosuppressive cells in tumor microenvironment. SIGNIFICANCE Data strongly suggest that Theo supplement, either as drug or as dietary supply, may represent a potent additional weapon against melanoma.
Collapse
Affiliation(s)
- Martina Cordella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Cinzia Senatore
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Carla Lintas
- Center for Neurodevelopmental Disorders, Laboratory of Molecular Psychiatry and Neurogenetics, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Adriana Eramo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Salvatore Valitutti
- Cancer Research Center of Toulouse, Toulouse, France; Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| | | | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
23
|
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: 140] [Impact Index Per Article: 28.0] [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
|
24
|
Sundaravel S, Kuo WL, Jeong JJ, Choudhary GS, Gordon-Mitchell S, Liu H, Bhagat TD, McGraw KL, Gurbuxani S, List AF, Verma A, Wickrema A. Loss of Function of DOCK4 in Myelodysplastic Syndromes Stem Cells is Restored by Inhibitors of DOCK4 Signaling Networks. Clin Cancer Res 2019; 25:5638-5649. [PMID: 31308061 DOI: 10.1158/1078-0432.ccr-19-0924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Myelodysplastic syndromes (MDS) with deletion of chromosome 7q/7 [-7/(del)7q MDS] is associated with worse outcomes and needs novel insights into pathogenesis. Reduced expression of signaling protein dedicator of cytokinesis 4 (DOCK4) in patients with -7/(del)7q MDS leads to a block in hematopoietic stem cell (HSC) differentiation. Identification of targetable signaling networks downstream of DOCK4 will provide means to restore hematopoietic differentiation in MDS.Experimental Design: We utilized phosphoproteomics approaches to identify signaling proteins perturbed as a result of reduced expression of DOCK4 in human HSCs and tested their functional significance in primary model systems. RESULTS We demonstrate that reduced levels of DOCK4 lead to increased global tyrosine phosphorylation of proteins in primary human HSCs. LYN kinase and phosphatases INPP5D (SHIP1) and PTPN6 (SHP1) displayed greatest levels of tyrosine phosphorylation when DOCK4 expression levels were reduced using DOCK4-specific siRNA. Our data also found that increased phosphorylation of SHIP1 and SHP1 phosphatases were due to LYN kinase targeting these phosphatases as substrates. Increased migration and impediment of HSC differentiation were consequences of these signaling alterations. Pharmacologic inhibition of SHP1 reversed these functional aberrations in HSCs expressing low DOCK4 levels. In addition, differentiation block seen in DOCK4 haplo-insufficient [-7/(del)7q] MDS was rescued by inhibition of SHP1 phosphatase. CONCLUSIONS LYN kinase and phosphatases SHP1 and SHIP1 are perturbed when DOCK4 expression levels are low. Inhibition of SHP1 promotes erythroid differentiation in healthy HSCs and in -7/(del)7q MDS samples with low DOCK4 expression. Inhibitors of LYN, SHP1 and SHIP1 also abrogated increased migratory properties in HSCs expressing reduced levels of DOCK4.
Collapse
Affiliation(s)
- Sriram Sundaravel
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Wen-Liang Kuo
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Jong Jin Jeong
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gaurav S Choudhary
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Hui Liu
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Tushar D Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Sandeep Gurbuxani
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | | | - Amit Verma
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Amittha Wickrema
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
25
|
Jiang C, Guo Y, Yu H, Lu S, Meng L. Pleiotropic microRNA-21 in pulmonary remodeling: novel insights for molecular mechanism and present advancements. Allergy Asthma Clin Immunol 2019; 15:33. [PMID: 31139230 PMCID: PMC6528201 DOI: 10.1186/s13223-019-0345-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/08/2019] [Indexed: 12/29/2022] Open
Abstract
MicroRNA-21 (miR-21), probably one of the most studied miRNAs to date, is found pleiotropic in various biological events. Its emerging role in pulmonary remodeling has attracted extensive attention. This review summarizes the genomic information of its primary transcript and various transcriptional regulations on its promoter. In addition, the role of miR-21 in pulmonary remodeling related signaling such as transforming growth factor β (TGF-β), bone morphogenetic protein (BMP), epidermal growth factor receptor (EGFR) and Notch signaling is discussed. Various validated miR-21 target genes participate in controlling of the overactive cell accumulation, smooth muscle contraction, inflammatory stress (trigger for lung epithelium damage), extracellular matrix deposition and hypoxia-induced disorders. Moreover, we focus on its particular implication in events including inflammatory stress-driven epithelium damage, epithelial-to-mesenchymal transition (EMT), transdifferentiation of fibroblasts into myofibroblasts, hypoxia stimuli and ROS response, as well as some other pulmonary remodeling related events such as overactive fibroblast (myofibroblast) accumulation, extracellular matrix deposition, and angiogenesis. Here, we summarize the strong potential of miR-21 in pulmonary remodeling and provide novel clues for further research in this area.
Collapse
Affiliation(s)
- Congshan Jiang
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| | - Yuanxu Guo
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| | - Hongchuan Yu
- Department of Respiratory Medicine, Xi'an Children Hospital, Xi'an, Shaanxi People's Republic of China
| | - Shemin Lu
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| | - Liesu Meng
- 1Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, West Yanta Road No.76, Xi'an, Shaanxi People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi People's Republic of China
| |
Collapse
|
26
|
Huang X, Liu Z, Shen L, Jin Y, Xu G, Zhang Z, Fang C, Guan W, Liu C. Augmentation of miR-202 in varicose veins modulates phenotypic transition of vascular smooth muscle cells by targeting proliferator-activated receptor-γ coactivator-1α. J Cell Biochem 2018; 120:10031-10042. [PMID: 30556158 DOI: 10.1002/jcb.28287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/19/2018] [Indexed: 02/04/2023]
Abstract
In varicose veins, vascular smooth muscle cells (VSMCs) often show abnormal proliferative and migratory rates and phenotypic transition. This study aimed to investigate whether microRNA (miR)-202 and its potential target, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), were involved in VSMC phenotypic transition. miR-202 expression was analyzed in varicose veins and in VSMCs conditioned with platelet-derived growth factor. The effect of miR-202 on cell proliferation and migration was assessed. Furthermore, contractile marker SM-22α, synthetic markers vimentin and collagen I, and PGC-1α were analyzed by Western blot analysis. The modulation of PGC-1α expression by miR-202 was also evaluated. In varicose veins and proliferative VSMCs, miR-202 expression was upregulated, with decreased SM-22α expression and increased vimentin and collagen I expression. Transfection with a miR-202 mimic induced VSMC proliferation and migration, whereas a miR-202 inhibitor reduced cell proliferation and migration. miR-202 mimic constrained luciferase activity in HEK293 cells that were cotransfected with the PGC-1α 3'-untranslated region (3'-UTR) but not those with mutated 3'-UTR. miR-202 suppressed PGC-1α protein expression, with no influence on its messenger RNA expression. PGC-1α mediated VSMC phenotypic transition and was correlated with reactive oxygen species production. In conclusion, miR-202 affects VSMC phenotypic transition by targeting PGC-1α expression, providing a novel target for varicose vein therapy.
Collapse
Affiliation(s)
- Xianchen Huang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhao Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liming Shen
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yiqi Jin
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Guoxiong Xu
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhixuan Zhang
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Changwen Fang
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Wenxian Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Changjian Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Lee J, Heo J, Kang H. miR-92b-3p-TSC1 axis is critical for mTOR signaling-mediated vascular smooth muscle cell proliferation induced by hypoxia. Cell Death Differ 2018; 26:1782-1795. [PMID: 30518907 PMCID: PMC6748132 DOI: 10.1038/s41418-018-0243-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 11/11/2018] [Accepted: 11/15/2018] [Indexed: 01/05/2023] Open
Abstract
Pulmonary artery smooth muscle cells (PASMCs) undergo proliferation by the mammalian target of rapamycin (mTOR) signaling pathway under hypoxia. Hypoxia induces expression of a specific set of microRNAs (miRNAs) in a variety of cell types. We integrated genomic analyses of both small non-coding RNA and coding transcripts using next-generation sequencing (NGS)-based RNA sequencing with the molecular mechanism of the mTOR signaling pathway in hypoxic PASMCs. These analyses revealed hypoxia-induced miR-92b-3p as a potent regulator of the mTOR signaling pathway. We demonstrated that miR-92b-3p directly targets the 3′-UTR of a negative regulator in the mTOR signaling pathway, TSC1. mTOR signaling and consequent cell proliferation were promoted by enforced expression of miR-92b-3p but inhibited by knocking down endogenous miR-92b-3p. Furthermore, inhibition of miR-92b-3p attenuated hypoxia-induced proliferation of vascular smooth muscle cells (VSMCs). Therefore, this study elucidates a novel role of miR-92b-3p as a hypoxamir in the regulation of the mTOR signaling pathway and the pathological VSMC proliferative response under hypoxia. These findings will help us better understand the miRNA-mediated molecular mechanism of the proliferative response of hypoxic VSMCs through the mTOR signaling pathway.
Collapse
Affiliation(s)
- Jihui Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea
| | - Jeongyeon Heo
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea.
| |
Collapse
|
28
|
Tang N, Jiang S, Yang Y, Liu S, Ponnusamy M, Xin H, Yu T. Noncoding RNAs as therapeutic targets in atherosclerosis with diabetes mellitus. Cardiovasc Ther 2018; 36:e12436. [PMID: 29797660 DOI: 10.1111/1755-5922.12436] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/08/2018] [Accepted: 05/20/2018] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the major macrovascular complications of diabetes mellitus (DM), and it is the main cause of death from clinical observation. Among various cell types involved in this disorder, endothelial cells, vascular smooth muscle cells (VSMCs), and macrophages play a crucial role in the occurrence and development of this disease. The regulation and stabilization of these cells are a key therapeutic strategy for DM-associated atherosclerosis. An increasing number of evidences implicate that various types of noncoding RNAs (ncRNAs) play a vital role in many cellular responses as well as in physiological and pathological processes of atherosclerosis and DM that drive atherogenic/antiatherogenic processes in those cells. Encouragingly, many ncRNAs have already been tested in animal experiments or clinical trials showing good performance. In this review, we summarize recent progresses in research on functional regulatory role of ncRNAs in atherosclerosis with DM. More importantly, we illustrate new thoughts and findings relevant to ncRNAs as potential therapeutic targets or biomarkers for atherosclerosis with DM.
Collapse
Affiliation(s)
- Ningning Tang
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Shaoyan Jiang
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Yanyan Yang
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Shaoyan Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Yu
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Canfrán-Duque A, Rotllan N, Zhang X, Fernández-Fuertes M, Ramírez-Hidalgo C, Araldi E, Daimiel L, Busto R, Fernández-Hernando C, Suárez Y. Macrophage deficiency of miR-21 promotes apoptosis, plaque necrosis, and vascular inflammation during atherogenesis. EMBO Mol Med 2018; 9:1244-1262. [PMID: 28674080 PMCID: PMC5582411 DOI: 10.15252/emmm.201607492] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis, the major cause of cardiovascular disease, is a chronic inflammatory disease characterized by the accumulation of lipids and inflammatory cells in the artery wall. Aberrant expression of microRNAs has been implicated in the pathophysiological processes underlying the progression of atherosclerosis. Here, we define the contribution of miR‐21 in hematopoietic cells during atherogenesis. Interestingly, we found that miR‐21 is the most abundant miRNA in macrophages and its absence results in accelerated atherosclerosis, plaque necrosis, and vascular inflammation. miR‐21 expression influences foam cell formation, sensitivity to ER‐stress‐induced apoptosis, and phagocytic clearance capacity. Mechanistically, we discovered that the absence of miR‐21 in macrophages increases the expression of the miR‐21 target gene, MKK3, promoting the induction of p38‐CHOP and JNK signaling. Both pathways enhance macrophage apoptosis and promote the post‐translational degradation of ABCG1, a transporter that regulates cholesterol efflux in macrophages. Altogether, these findings reveal a major role for hematopoietic miR‐21 in atherogenesis.
Collapse
Affiliation(s)
- Alberto Canfrán-Duque
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Marta Fernández-Fuertes
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Cristina Ramírez-Hidalgo
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Elisa Araldi
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Lidia Daimiel
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Rebeca Busto
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal de Investigación Sanitaria (IRyCIS), Madrid, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program and the Departments of Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
30
|
Huang X, Yue Z, Wu J, Chen J, Wang S, Wu J, Ren L, Zhang A, Deng P, Wang K, Wu C, Ding X, Ye P, Xia J. MicroRNA-21 Knockout Exacerbates Angiotensin II–Induced Thoracic Aortic Aneurysm and Dissection in Mice With Abnormal Transforming Growth Factor-β–SMAD3 Signaling. Arterioscler Thromb Vasc Biol 2018. [DOI: 10.1161/atvbaha.117.310694] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective—
Thoracic aortic aneurysm and dissection (TAAD) are severe vascular conditions. Dysfunctional transforming growth factor-β (TGF-β) signaling in vascular smooth muscle cells and elevated angiotensin II (AngII) levels are implicated in the development of TAAD. In this study, we investigated whether these 2 factors lead to TAAD in a mouse model and explored the possibility of using microRNA-21 (
miR-21
) for the treatment of TAAD.
Approach and Results—
TAAD was developed in
Smad3
(mothers against decapentaplegic homolog 3) heterozygous (S3
+/−
) mice infused with AngII. We found that p-ERK (phosphorylated extracellular regulated protein kinases)– and p-JNK (phosphorylated c-Jun N-terminal kinase)–associated
miR-21
was higher in TAAD lesions. We hypothesize that downregulation of
miR-21
mitigate TAAD formation. However,
Smad3
+/−
:miR-21
−/−
(S3
+/−
21
−/−
) mice exhibited conspicuous TAAD formation after AngII infusion. The vascular wall was dilated, and aortic rupture occurred within 23 days during AngII infusion. We then examined canonical and noncanonical TGF-β signaling and found that
miR-21
knockout in S3
+/−
mice increased SMAD7 and suppressed canonical TGF-β signaling. Vascular smooth muscle cells lacking TGF-β signals tended to switch from a contractile to a synthetic phenotype. The silencing of
Smad7
with lentivirus prevented AngII-induced TAAD formation in S3
+/−
21
−/−
mice.
Conclusions—
Our study demonstrated that
miR-21
knockout exacerbated AngII-induced TAAD formation in mice, which was associated with TGF-β signaling dysfunction. Therapeutic strategies targeting TAAD should consider unexpected side effects associated with alterations in TGF-β signaling.
Collapse
Affiliation(s)
- Xiaofan Huang
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Zhang Yue
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Jia Wu
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
- Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan (J.W.)
| | - Jiuling Chen
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Sihua Wang
- Department of Thoracic Surgery, Union Hospital (S.W.)
| | - Jie Wu
- Central Laboratory, Central Hospital of Wuhan (J.W.)
| | - Linyun Ren
- Department of Anesthesia, Central Hospital of Wuhan (L.R.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anchen Zhang
- Department of Cardiovascular Medicine, Central Hospital of Wuhan (A.Z., P.Y.)
| | - Peng Deng
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Ke Wang
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Chuangyan Wu
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Xiangchao Ding
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Ping Ye
- Department of Cardiovascular Medicine, Central Hospital of Wuhan (A.Z., P.Y.)
| | - Jiahong Xia
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
- Department of Cardiovascular Surgery, Central Hospital of Wuhan (J.X.)
| |
Collapse
|
31
|
Sakai H, Suto W, Kai Y, Chiba Y. Mechanisms underlying the pathogenesis of hyper-contractility of bronchial smooth muscle in allergic asthma. J Smooth Muscle Res 2018; 53:37-47. [PMID: 28484126 PMCID: PMC5411784 DOI: 10.1540/jsmr.53.37] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Airway hyperresponsiveness (AHR) and inflammation are key pathophysiological
features of asthma. Enhanced contraction of bronchial smooth muscle (BSM) is one
of the causes of the AHR. It is thus important for development of asthma therapy
to understand the change in the contractile signaling of airway smooth muscle
cells associated with the AHR. In addition to the Ca2+-mediated
phosphorylation of myosin light chain (MLC), contractile agonists also enhance
MLC phosphorylation level, Ca2+-independently, by inactivating MLC
phosphatase (MLCP), called Ca2+ sensitization of contraction, in
smooth muscle cells including airways. To date, involvements of RhoA/ROCKs and
PKC/Ppp1r14a (also called as CPI-17) pathways in the Ca2+
sensitization have been identified. Our previous studies revealed that the
agonist-induced Ca2+ sensitization of contraction is markedly
augmented in BSMs of animal models of allergen-induced AHR. In BSMs of these
animal models, the expression of RhoA and CPI-17 proteins were significantly
increased, indicating that both the Ca2+ sensitizing pathways are
augmented. Interestingly, incubation of BSM cells with asthma-associated
cytokines, such as interleukin-13 (IL-13), IL-17, and tumor necrosis factor-α
(TNF-α), caused up-regulations of RhoA and CPI-17 in BSM cells of naive animals
and cultured human BSM cells. In addition to the transcription factors such as
STAT6 and NF-κB activated by these inflammatory cytokines, an involvement of
down-regulation of miR-133a, a microRNA that negatively regulates RhoA
translation, has also been suggested in the IL-13- and IL-17-induced
up-regulation of RhoA. Thus, the Ca2+ sensitizing pathways and the
cytokine-mediated signaling including microRNAs in BSMs might be potential
targets for treatment of allergic asthma, especially the AHR.
Collapse
Affiliation(s)
- Hiroyasu Sakai
- Department of Analytical Pathophysiology, Hoshi University
| | - Wataru Suto
- Department of Physiology and Molecular Sciences, Hoshi University
| | - Yuki Kai
- Department of Analytical Pathophysiology, Hoshi University
| | - Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, Hoshi University
| |
Collapse
|
32
|
Poenitzsch Strong AM, Berry SM, Beebe DJ, Li JL, Spiegelman VS. miFAST: A novel and rapid microRNA target capture method. Mol Carcinog 2018; 57:559-566. [PMID: 29350431 DOI: 10.1002/mc.22780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRNAs), small 22-25 nucleotide non-coding RNAs, play important roles in cellular and tumor biology. However, characterizing miRNA function remains challenging due to an abundance of predicted targets and an experimental bottleneck in identifying biologically relevant direct targets. Here, we developed a novel technique (miFAST) to identify direct miRNA target genes. Using miFAST, we confirmed several previously reported miR-340 target genes and identified five additional novel direct miR-340 targets in melanoma cells. This methodology can also be efficiently applied for the global characterization of miRNA targets. Utilizing miFAST to characterize direct miRNA targetomes will further our understanding of miRNA biology and function.
Collapse
Affiliation(s)
| | - Scott M Berry
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jian-Liang Li
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida
| | - Vladimir S Spiegelman
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
33
|
Role of MicroRNAs in TGF-β Signaling Pathway-Mediated Pulmonary Fibrosis. Int J Mol Sci 2017; 18:ijms18122527. [PMID: 29186838 PMCID: PMC5751130 DOI: 10.3390/ijms18122527] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/19/2017] [Accepted: 11/22/2017] [Indexed: 12/31/2022] Open
Abstract
Pulmonary fibrosis is the most common form of interstitial lung disease. The transforming growth factor-β (TGF-β) signaling pathway is extensively involved in the development of pulmonary fibrosis by inducing cell differentiation, migration, invasion, or hyperplastic changes. Accumulating evidence indicates that microRNAs (miRNAs) are dysregulated during the initiation of pulmonary fibrosis. miRNAs are small noncoding RNAs functioning as negative regulators of gene expression at the post-transcriptional level. A number of miRNAs have been reported to regulate the TGF-β signaling pathway and consequently affect the process of pulmonary fibrosis. A better understanding of the pro-fibrotic role of the TGF-β signaling pathway and relevant miRNA regulation will shed light on biomedical research of pulmonary fibrosis. This review summarizes the current knowledge of miRNAs regulating the TGF-β signaling pathway with relevance to pulmonary fibrosis.
Collapse
|
34
|
miR-21-5p is associated with the regulation of estradiol benzoate and oxytocin induced primary dysmenorrhea in rat uterus: a bioinformatic study. Genes Genomics 2017. [DOI: 10.1007/s13258-017-0591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Li Y, Zhang J, Lei Y, Lyu L, Zuo R, Chen T. MicroRNA-21 in Skin Fibrosis: Potential for Diagnosis and Treatment. Mol Diagn Ther 2017; 21:633-642. [DOI: 10.1007/s40291-017-0294-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
36
|
Zhang L, Yao J, Li W, Zhang C. Micro-RNA-21 Regulates Cancer-Associated Fibroblast-Mediated Drug Resistance in Pancreatic Cancer. Oncol Res 2017; 26:827-835. [PMID: 28477403 PMCID: PMC7844724 DOI: 10.3727/096504017x14934840662335] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer deaths due to its highly aggressive biological nature and resistance to chemotherapy. Previous studies indicate that miR-21 is an important regulator in the activation of cancer-associated fibroblasts (CAFs). However, whether miR-21 in CAFs would regulate PDAC’s tumor microenvironment and lead to drug resistance remain unknown. In this study, we evaluated the relationship between CAF activation, miR-21 expression, and drug resistance using tumor samples from PDAC patients. We changed the miR-21 expression level in CAFs and tested its roles in regulating the function of CAFs. In addition, we explored the roles of miR-21 in CAFs in the development of PDAC using an animal model. We found that PDAC patients who were resistant to gemcitabine treatment tended to have higher miR-21 expression and more activated CAFs. An in vitro study showed that CAFs with high miR-21 expression had elevated MMP-3, MMP-9, PDGF, and CCL-7 expression and promoted the invasion of PDAC cell lines. miR-21 overexpression also contributed to the activation of CAFs by regulating the PDCD4 gene. The in vivo study showed that upregulating miR-21 in CAFs promoted PDAC desmoplasia and increased its drug resistance to gemcitabine treatment, but downregulating miR-21 in CAFs suppressed desmoplasia and enhanced the effect of gemcitabine. We concluded that miR-21 promoted the activation of CAFs and contributed to the drug resistance of PDAC.
Collapse
Affiliation(s)
- Lulin Zhang
- Department of Oncology, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, P.R. China
| | - Jun Yao
- Department of Oncology, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, P.R. China
| | - Wenyao Li
- Department of Oncology, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, P.R. China
| | - Ce Zhang
- Department of Oncology, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, P.R. China
| |
Collapse
|
37
|
Abstract
Pulmonary hypertension (PH) is a multifaceted vascular disease where development and severity are determined by both genetic and environmental factors. Over the past decade, there has been an acceleration of the discovery of molecular effectors that mediate PH pathogenesis, including large numbers of microRNA molecules that are expressed in pulmonary vascular cell types and exert system-wide regulatory functions in all aspects of vascular health and disease. Due to the inherent pleiotropy, overlap, and redundancy of these molecules, it has been challenging to define their integrated effects on overall disease manifestation. In this review, we summarize our current understanding of the roles of microRNAs in PH with an emphasis on potential methods to discern the hierarchical motifs governing their multifunctional and interconnected activities. Deciphering this higher order of regulatory structure will be crucial for overcoming the challenges of developing these molecules as biomarkers or therapeutic targets, in isolation or combination.
Collapse
|
38
|
Elçin AE, Parmaksiz M, Dogan A, Seker S, Durkut S, Dalva K, Elçin YM. Differential gene expression profiling of human adipose stem cells differentiating into smooth muscle-like cells by TGFβ1/BMP4. Exp Cell Res 2017; 352:207-217. [DOI: 10.1016/j.yexcr.2017.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 12/18/2022]
|
39
|
Abstract
PURPOSE OF REVIEW Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are becoming fundamentally important in the pathophysiology relating to injury-induced vascular remodelling. We highlight recent studies that demonstrate the involvement of ncRNAs in vein graft disease, in in-stent restenosis and in pulmonary arterial hypertension, with a particular focus on endothelial cell and vascular smooth muscle cell function. We also briefly discuss the emerging role of exosomal-derived ncRNAs and how this mechanism impacts on vascular function. RECENT FINDINGS ncRNAs have been described as novel regulators in the pathophysiology of vascular injury, inflammation, and vessel wall remodelling. In particular, several studies have demonstrated that manipulation of miRNAs can reduce the burden of pathological vascular remodelling. Such studies have also shown that exosomal miRNA-mediated, cell-to-cell communication between endothelial cells and vascular smooth muscle cells is critical in the disease process. In addition to miRNAs, lncRNAs are emerging as regulators of vascular function in health and disease. Although lncRNAs are complex in both their sheer numbers and mechanisms of action, identifying their contribution to vascular disease is essential. SUMMARY Given the important roles of ncRNAs in vascular injury and remodelling together will their capacity for cell-to-cell communication, manipulating ncRNA might provide novel therapeutic interventions.
Collapse
Affiliation(s)
- Lin Deng
- aBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow bCentre for Cardiovascular Science, Queen's Medical Research Institute, BHF/University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
40
|
Wahlang B, Petriello MC, Perkins JT, Shen S, Hennig B. Polychlorinated biphenyl exposure alters the expression profile of microRNAs associated with vascular diseases. Toxicol In Vitro 2016; 35:180-7. [PMID: 27288564 PMCID: PMC4949395 DOI: 10.1016/j.tiv.2016.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/05/2016] [Accepted: 06/07/2016] [Indexed: 01/17/2023]
Abstract
Exposure to persistent organic pollutants, including polychlorinated biphenyls (PCBs) is correlated with multiple vascular complications including endothelial cell dysfunction and atherosclerosis. PCB-induced activation of the vasculature subsequently leads to oxidative stress and induction of pro-inflammatory cytokines and adhesion proteins. Gene expression of these cytokines/proteins is known to be regulated by small, endogenous oligonucleotides known as microRNAs that interact with messenger RNA. MicroRNAs are an acknowledged component of the epigenome, but the role of environmentally-driven epigenetic changes such as toxicant-induced changes in microRNA profiles is currently understudied. The objective of this study was to determine the effects of PCB exposure on microRNA expression profile in primary human endothelial cells using the commercial PCB mixture Aroclor 1260. Samples were analyzed using Affymetrix GeneChip® miRNA 4.0 arrays for high throughput detection and selected microRNA gene expression was validated (RT-PCR). Microarray analysis identified 557 out of 6658 microRNAs that were changed with PCB exposure (p<0.05). In-silico analysis using MetaCore database identified 21 of these microRNAs to be associated with vascular diseases. Further validation showed that Aroclor 1260 increased miR-21, miR-31, miR-126, miR-221 and miR-222 expression levels. Upregulated miR-21 has been reported in cardiac injury while miR-126 and miR-31 modulate inflammation. Our results demonstrated evidence of altered microRNA expression with PCB exposure, thus providing novel insights into mechanisms of PCB toxicity.
Collapse
Affiliation(s)
- Banrida Wahlang
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40536, USA
| | - Michael C Petriello
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40536, USA
| | - Jordan T Perkins
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Shu Shen
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Animal and Food Sciences, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40536, USA; Graduate Center for Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
41
|
Kim K, Yang DK, Kim S, Kang H. miR-142-3p Is a Regulator of the TGFβ-Mediated Vascular Smooth Muscle Cell Phenotype. J Cell Biochem 2016; 116:2325-33. [PMID: 25832008 DOI: 10.1002/jcb.25183] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/31/2015] [Indexed: 11/06/2022]
Abstract
The transforming growth factor β (TGFβ) signaling pathway is critical for the promotion and maintenance of the contractile phenotype of vascular smooth muscle cells (VSMCs). Though multiple microRNAs (miRNAs) implicated in the regulation of the VSMC phenotype have been identified, the modulation of miRNAs in the VSMCs by TGFβ signaling has not been fully described. In this study, we identified microRNA-142-3p (miR-142-3p) as a modulator of the VSMC phenotype in response to TGFβ signaling. We show that miR-142-3p is induced upon TGFβ signaling, leading to the repression of a novel target, dedicator of cytokinesis 6 (DOCK6). The downregulation of DOCK6 by miR-142-3p is critical for cell migration. Thus, this study demonstrates that miR-142-3p is a key regulator of the TGFβ-mediated contractile phenotype of VSMCs that acts through inhibiting cell migration through targeting DOCK6.
Collapse
Affiliation(s)
- Kwangho Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea
| | | | - Sunghwan Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 701-310, Republic of Korea
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea
| |
Collapse
|
42
|
Kang H, Hata A. Quantitative Real-Time PCR Analysis of MicroRNAs and Their Precursors Regulated by TGF-β Signaling. Methods Mol Biol 2016; 1344:313-23. [PMID: 26520134 DOI: 10.1007/978-1-4939-2966-5_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The signaling pathway of TGF-β and its family member BMP has been implicated in vascular development and maintenance of homeostasis by modulating expression of small noncoding microRNAs (miRNAs). MiRNAs repress target genes, which play a critical role in regulating vascular smooth muscle cell (VSMC) growth, phenotype, and function. To understand the mechanisms by which specific miRNAs control the TGF-β and BMP signaling pathway in VSMC, it is essential to quantitate levels of specific miRNAs and their precursors whose expression are controlled by TGF-β/BMP signaling. Here, we describe a real-time quantization method for accurate and sensitive detection of miRNAs and their precursors, such as primary transcripts of miRNAs (pri-miRNAs) and precursor miRNAs (pre-miRNAs). This method requires two steps; synthesis of single-stranded complementary DNAs (cDNAs) from total RNA samples and quantization of specific pri-, pre-, or mature miRNAs by quantitative polymerase chain reaction (PCR) using a real-time PCR machine.
Collapse
Affiliation(s)
- Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea.,Cardiovascular Research Institute, University of California, 555 Mission Bay Blvd. South, Rm. 252T, San Francisco, CA, 94158-9001, USA
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, 555 Mission Bay Blvd. South, Rm. 252T, San Francisco, CA, 94158-9001, USA.
| |
Collapse
|
43
|
Martin E, Qureshi A, Dasa V, Freitas M, Gimble J, Davis T. MicroRNA regulation of stem cell differentiation and diseases of the bone and adipose tissue: Perspectives on miRNA biogenesis and cellular transcriptome. Biochimie 2016; 124:98-111. [DOI: 10.1016/j.biochi.2015.02.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/17/2015] [Indexed: 12/19/2022]
|
44
|
Coll-Bonfill N, de la Cruz-Thea B, Pisano MV, Musri MM. Noncoding RNAs in smooth muscle cell homeostasis: implications in phenotypic switch and vascular disorders. Pflugers Arch 2016; 468:1071-87. [PMID: 27109570 DOI: 10.1007/s00424-016-1821-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/04/2016] [Indexed: 12/16/2022]
Abstract
Vascular smooth muscle cells (SMC) are a highly specialized cell type that exhibit extraordinary plasticity in adult animals in response to a number of environmental cues. Upon vascular injury, SMC undergo phenotypic switch from a contractile-differentiated to a proliferative/migratory-dedifferentiated phenotype. This process plays a major role in vascular lesion formation and during the development of vascular remodeling. Vascular remodeling comprises the accumulation of dedifferentiated SMC in the intima of arteries and is central to a number of vascular diseases such as arteriosclerosis, chronic obstructive pulmonary disease or pulmonary hypertension. Therefore, it is critical to understand the molecular mechanisms that govern SMC phenotype. In the last decade, a number of new classes of noncoding RNAs have been described. These molecules have emerged as key factors controlling tissue homeostasis during physiological and pathological conditions. In this review, we will discuss the role of noncoding RNAs, including microRNAs and long noncoding RNAs, in the regulation of SMC plasticity.
Collapse
Affiliation(s)
- N Coll-Bonfill
- Department of Pulmonary Medicine Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - B de la Cruz-Thea
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - M V Pisano
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - M M Musri
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.
| |
Collapse
|
45
|
Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacol Rev 2016; 68:476-532. [PMID: 27037223 PMCID: PMC4819215 DOI: 10.1124/pr.115.010652] [Citation(s) in RCA: 298] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.
Collapse
Affiliation(s)
- F V Brozovich
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C J Nicholson
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C V Degen
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - Yuan Z Gao
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - M Aggarwal
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - K G Morgan
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| |
Collapse
|
46
|
Morrell NW, Bloch DB, ten Dijke P, Goumans MJTH, Hata A, Smith J, Yu PB, Bloch KD. Targeting BMP signalling in cardiovascular disease and anaemia. Nat Rev Cardiol 2016; 13:106-20. [PMID: 26461965 PMCID: PMC4886232 DOI: 10.1038/nrcardio.2015.156] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone morphogenetic proteins (BMPs) and their receptors, known to be essential regulators of embryonic patterning and organogenesis, are also critical for the regulation of cardiovascular structure and function. In addition to their contributions to syndromic disorders including heart and vascular development, BMP signalling is increasingly recognized for its influence on endocrine-like functions in postnatal cardiovascular and metabolic homeostasis. In this Review, we discuss several critical and novel aspects of BMP signalling in cardiovascular health and disease, which highlight the cell-specific and context-specific nature of BMP signalling. Based on advancing knowledge of the physiological roles and regulation of BMP signalling, we indicate opportunities for therapeutic intervention in a range of cardiovascular conditions including atherosclerosis and pulmonary arterial hypertension, as well as for anaemia of inflammation. Depending on the context and the repertoire of ligands and receptors involved in specific disease processes, the selective inhibition or enhancement of signalling via particular BMP ligands (such as in atherosclerosis and pulmonary arterial hypertension, respectively) might be beneficial. The development of selective small molecule antagonists of BMP receptors, and the identification of ligands selective for BMP receptor complexes expressed in the vasculature provide the most immediate opportunities for new therapies.
Collapse
Affiliation(s)
- Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Donald B Bloch
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Peter ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, 500 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jim Smith
- MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Paul B Yu
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Kenneth D Bloch
- Anaesthesia Centre for Critical Care Research, Department of Anaesthesia, Critical Care and Pain Medicine, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
47
|
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate the stability and expression of target RNAs in a sequence-dependent manner. Identifying miRNA-regulated genes is key to understanding miRNA function. Here, we describe an unbiased biochemical pulldown method to identify with high-specificity miRNA targets. Regulated transcripts are enriched in streptavidin-captured mRNAs that bind to a transfected biotinylated miRNA mimic. The method is relatively simple, does not involve cross-linking and can be performed with only a million cells. Addition of an on-bead RNase digestion step also identifies miRNA recognition elements (MRE).
Collapse
Affiliation(s)
- Shen Mynn Tan
- Cellular and Molecular Medicine Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, 200 Longwood Ave, Boston, MA, 02115, USA
| | - Judy Lieberman
- Cellular and Molecular Medicine Program, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, 200 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
48
|
Effects of the Pinggan Qianyang Recipe on MicroRNA Gene Expression in the Aortic Tissue of Spontaneously Hypertensive Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:154691. [PMID: 26379738 PMCID: PMC4561872 DOI: 10.1155/2015/154691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 01/24/2015] [Accepted: 01/28/2015] [Indexed: 01/17/2023]
Abstract
The present study aimed to investigate the relationship between miRNAs and in spontaneously hypertensive rats (SHR) vascular remodeling and analyze the impact of the Pinggan Qianyang recipe (PQR) on miRNAs. Mammalian miRNA microarrays containing 509 miRNA genes were employed to analyze the differentially expressed miRNAs in the three groups. MiRNAs were considered to be up- or downregulated when the fluorescent intensity ratio between the two groups was over 4-fold. Validation of those miRNAs changed in SHR after PQR treatment was used by real-time quantitative RT-PCR (qRT-PCR). Compared with the normal group, a total of 32 miRNAs were differentially expressed by more than twofold; among these, 18 were upregulated and 14 were downregulated in the model group. Compared with the normal group, there were a number of 17 miRNAs which were significantly expressed by more than twofold in the different expressions of 32 miRNAs; among these, 10 were downregulated and 7 were upregulated in the PQR group. qRT-PCR verified that miR-20a, miR-145, miR-30, and miR-98 were significantly expressed in the three groups. These data show that PQR could exert its antihypertensive effect through deterioration of the vascular remodeling process. The mechanism might be associated with regulating differentially expressed miRNAs in aorta tissue.
Collapse
|
49
|
Kurakula K, Goumans MJ, Ten Dijke P. Regulatory RNAs controlling vascular (dys)function by affecting TGF-ß family signalling. EXCLI JOURNAL 2015; 14:832-50. [PMID: 26862319 PMCID: PMC4743484 DOI: 10.17179/excli2015-423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/15/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Over the last few years, microRNAs (miRNAs) have emerged as master regulators of gene expression in cardiovascular biology and disease. miRNAs are small endogenous non-coding RNAs that usually bind to 3′ untranslated region (UTR) of their target mRNAs and inhibit mRNA stability or translation of their target genes. miRNAs play a dynamic role in the pathophysiology of many CVDs through their effects on target mRNAs in vascular cells. Recently, numerous miRNAs have been implicated in the regulation of the transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signalling pathway which plays crucial roles in diverse biological processes, and is involved in pathogenesis of many diseases including CVD. This review gives an overview of current literature on the role of miRNAs targeting TGF-β/BMP signalling in vascular cells, including endothelial cells and smooth muscle cells. We also provide insight into how this miRNA-mediated regulation of TGF-β/BMP signalling might be used to harness CVD.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-Jose Goumans
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
50
|
Xu D, Gu JT, Yi B, Chen L, Wang GS, Qian GS, Lu KZ. Requirement of miR-9-dependent regulation of Myocd in PASMCs phenotypic modulation and proliferation induced by hepatopulmonary syndrome rat serum. J Cell Mol Med 2015; 19:2453-61. [PMID: 26147104 PMCID: PMC4594686 DOI: 10.1111/jcmm.12631] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/13/2015] [Indexed: 12/18/2022] Open
Abstract
Hepatopulmonary syndrome (HPS) is characterized by a triad of severe liver disease, intrapulmonary vascular dilation and hypoxaemia. Pulmonary vascular remodelling (PVR) is a key feature of HPS pathology. Our previous studies have established the role of the pulmonary artery smooth muscle cell (PASMC) phenotypic modulation and proliferation in HPS-associated PVR. Myocardin, a robust transcriptional coactivator of serum response factor, plays a critical role in the vascular smooth muscle cell phenotypic switch. However, the mechanism regulating myocardin upstream signalling remains unclear. In this study, treatment of rat PASMCs with serum drawn from common bile duct ligation rats, which model symptoms of HPS, resulted in a significant increase in miR-9 expression correlated with a decrease in expression of myocardin and the phenotypic markers SM-α-actin and smooth muscle-specific myosin heavy chain (SM-MHC). Furthermore, miRNA functional analysis and luciferase reporter assay demonstrated that miR-9 effectively regulated myocardin expression by directly binding to its 3′-untranslated region. Both the knockdown of miR-9 and overexpression of myocardin effectively attenuated the HPS rat serum-induced phenotype switch and proliferation of PASMCs. Taken together, the findings of our present study demonstrate that miR-9 is required in HPS rat serum-induced phenotypic modulation and proliferation of PASMCs for targeting of myocardin and that miR-9 may serve as a potential therapeutic target in HPS.
Collapse
Affiliation(s)
- Duo Xu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jian-teng Gu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bin Yi
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lin Chen
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Guan-song Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Gui-sheng Qian
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Kai-zhi Lu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|