1
|
Alizadeh M, Ghasemi H, Bazhan D, Mohammadi Bolbanabad N, Rahdan F, Arianfar N, Vahedi F, Khatami SH, Taheri-Anganeh M, Aiiashi S, Armand N. MicroRNAs in disease States. Clin Chim Acta 2025; 569:120187. [PMID: 39938625 DOI: 10.1016/j.cca.2025.120187] [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] [Received: 11/24/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
This review highlights the role of miRNAs in various diseases affecting major organ systems. miRNAs are small, non-coding RNA molecules that regulate numerous genes. Dysregulation of miRNAs is linked to many pathological conditions due to their involvement in gene silencing and cellular pathways. We discuss miRNA expression patterns, their physiological and pathological roles, and how changes in miRNA levels contribute to disease. Notably, miRNAs like miR-499 and miR-21 are implicated in heart failure and atherosclerosis. miRNA dysregulation is also associated with colorectal and gastric cancers, influencing tumorigenesis and chemoresistance. In neurological diseases, miRNAs exhibit diverse profiles that affect neurodevelopment and degeneration. Additionally, miRNAs modulate cell function in reproductive organs, impacting fertility and cancer progression. miRNAs such as miR-192 and miR-204 serve as biomarkers for nephropathy and acute kidney injury. These miRNAs are involved in skeletal muscle diseases, contributing to conditions like osteoporosis and sarcopenia. miRNAs function as oncogenes or tumor suppressors in cancer, highlighting their potential in diagnostics and therapy. Further research is needed to develop miRNA-based diagnostics and treatments.
Collapse
Affiliation(s)
- Mehdi Alizadeh
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Ghasemi
- Research Center for Environmental Contaminants (RCEC), Abadan University of Medical Sciences, Abadan, Iran
| | - Donya Bazhan
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Arianfar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Vahedi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Saleh Aiiashi
- Abadan University of Medical Sciences, Abadan, Iran.
| | - Nezam Armand
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
2
|
Calewaert A, Palarea-Albaladejo J, Coultous R, Capewell P, Hanks E, Decloedt A, van Loon G. Comparison of serum microRNA in healthy horses and horses with moderate to severe mitral valve regurgitation using a commercially available canine cardiac panel. Equine Vet J 2024. [PMID: 39567225 DOI: 10.1111/evj.14434] [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: 05/03/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND MicroRNA (miRNA) has previously been used as a biomarker for cardiac disease in humans and dogs, however, studies in horses are not yet available. OBJECTIVES To determine if adult horses with moderate or severe mitral valve regurgitation have a different serum miRNA expression profile compared to healthy controls. STUDY DESIGN Retrospective cross-sectional. METHODS Serum samples from 77 adult horses with moderate or severe mitral valve regurgitation and 77 healthy control horses were analysed using a commercial cardiac disease-specific miRNA panel previously used in dogs. RESULTS The commercial canine cardiac miRNA panel had low discriminatory power as a biomarker for mitral valve regurgitation in adult horses. Sensitivity was 0.58 (95% Cl: 0.47-0.69) and specificity 0.57 (95% Cl: 0.46-0.68). MAIN LIMITATIONS Clinical data were extracted retrospectively and currently there is no well-established criteria for grading mitral regurgitation in horses; there were few severe mitral regurgitation cases and the pathogenesis of mitral regurgitation was not considered. Controls were not matched by age, breed or sex. An assay developed for use in dogs was used. CONCLUSION Despite strong miRNA conservation across species, the commercially available canine cardiac miRNA panel failed as biomarker for mitral valve regurgitation in adult horses. Further research is needed to determine if an equine specific panel can be developed that performs better as biomarker for cardiac disease in horses.
Collapse
Affiliation(s)
- Amber Calewaert
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Javier Palarea-Albaladejo
- Department of Computer Sciences, Applied Mathematics and Statistics, University of Girona, Girona, Spain
- MI:RNA Ltd, Edinburgh, UK
| | | | - Paul Capewell
- MI:RNA Ltd, Edinburgh, UK
- School of Molecular Biosciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Annelies Decloedt
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Gunther van Loon
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
3
|
Kepreotis SV, Oh JG, Park M, Yoo J, Lee C, Mercola M, Hajjar RJ, Jeong D. Inhibition of miR-25 ameliorates cardiac and skeletal muscle dysfunction in aged mdx/utrn haploinsufficient (+/-) mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102174. [PMID: 38584818 PMCID: PMC10998245 DOI: 10.1016/j.omtn.2024.102174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
Dystrophic cardiomyopathy is a significant feature of Duchenne muscular dystrophy (DMD). Increased cardiomyocyte cytosolic calcium (Ca2+) and interstitial fibrosis are major pathophysiological hallmarks that ultimately result in cardiac dysfunction. MicroRNA-25 (miR-25) has been identified as a suppressor of both sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) and mothers against decapentaplegic homolog-7 (Smad7) proteins. In this study, we created a gene transfer using an miR-25 tough decoy (TuD) RNA inhibitor delivered via recombinant adeno-associated virus serotype 9 (AAV9) to evaluate the effect of miR-25 inhibition on cardiac and skeletal muscle function in aged dystrophin/utrophin haploinsufficient mice mdx/utrn (+/-), a validated transgenic murine model of DMD. We found that the intravenous delivery of AAV9 miR-25 TuD resulted in strong and stable inhibition of cardiac miR-25 levels, together with the restoration of SERCA2a and Smad7 expression. This was associated with the amelioration of cardiomyocyte interstitial fibrosis as well as recovered cardiac function. Furthermore, the direct quadricep intramuscular injection of AAV9 miR-25 TuD significantly restored skeletal muscle Smad7 expression, reduced tissue fibrosis, and enhanced skeletal muscle performance in mdx/utrn (+/-) mice. These results imply that miR-25 TuD gene transfer may be a novel therapeutic approach to restore cardiomyocyte Ca2+ homeostasis and abrogate tissue fibrosis in DMD.
Collapse
Affiliation(s)
- Sacha V. Kepreotis
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Jae Gyun Oh
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Mina Park
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Cholong Lee
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Roger J. Hajjar
- Mass General Brigham Gene and Cell Therapy Institute, Boston, MA, USA
| | - Dongtak Jeong
- Department of Medicinal and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
- Cardiovascular Research Institute, Icahn School of Medicine, Mount Sinai, NY, USA
| |
Collapse
|
4
|
Liu X, Shao Y, Han L, Zhang R, Chen J. Emerging Evidence Linking the Liver to the Cardiovascular System: Liver-derived Secretory Factors. J Clin Transl Hepatol 2023; 11:1246-1255. [PMID: 37577236 PMCID: PMC10412704 DOI: 10.14218/jcth.2022.00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 07/03/2023] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide. Recently, accumulating evidence has revealed hepatic mediators, termed as liver-derived secretory factors (LDSFs), play an important role in regulating CVDs such as atherosclerosis, coronary artery disease, thrombosis, myocardial infarction, heart failure, metabolic cardiomyopathy, arterial hypertension, and pulmonary hypertension. LDSFs presented here consisted of microbial metabolite, extracellular vesicles, proteins, and microRNA, they are primarily or exclusively synthesized and released by the liver, and have been shown to exert pleiotropic actions on cardiovascular system. LDSFs mainly target vascular endothelial cell, vascular smooth muscle cells, cardiomyocytes, fibroblasts, macrophages and platelets, and further modulate endothelial nitric oxide synthase/nitric oxide, endothelial function, energy metabolism, inflammation, oxidative stress, and dystrophic calcification. Although some LDSFs are known to be detrimental/beneficial, controversial findings were also reported for many. Therefore, more studies are required to further explore the causal relationships between LDSFs and CVDs and uncover the exact mechanisms, which is expected to extend our understanding of the crosstalk between the liver and cardiovascular system and identify potential therapeutic targets. Furthermore, in the case of patients with liver disease, awareness should be given to the implications of these abnormalities in the cardiovascular system. These studies also underline the importance of early recognition and intervention of liver abnormalities in the practice of cardiovascular care, and a multidisciplinary approach combining hepatologists and cardiologists would be more preferable for such patients.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Yijia Shao
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linjiang Han
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Ruyue Zhang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Traxler D, Dannenberg V, Zlabinger K, Gugerell A, Mester-Tonczar J, Lukovic D, Spannbauer A, Hasimbegovic E, Kastrup J, Gyöngyösi M. Plasma Small Extracellular Vesicle Cardiac miRNA Expression in Patients with Ischemic Heart Failure, Randomized to Percutaneous Intramyocardial Treatment of Adipose Derived Stem Cells or Placebo: Subanalysis of the SCIENCE Study. Int J Mol Sci 2023; 24:10647. [PMID: 37445825 DOI: 10.3390/ijms241310647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Small extracellular vesicles (EVs) and their cargo are an important component of cell-to-cell communication in cardiac disease. Allogeneic adipose derived stem cells (ADSCs) are thought to be a potential approach for cardiac regenerative therapy in ischemic heart disease. The SCIENCE study investigated the effect of ADSCs administered via intramyocardial injection on cardiac function in patients with ischemic heart disease. The aim of this substudy, based on samples from 15 patients, was to explore small EV miRNA dynamics after treatment with ADSCs compared to a placebo. Small EVs were isolated at several timepoints after the percutaneous intramyocardial application of ADSCs. No significant effect of ADSC treatment on small EV concentration was detected. After 12 months, the expression of miR-126 decreased significantly in ADSC patients, but not in the placebo-treated group. However, all cardiac miRNAs correlated with plasma cardiac biomarkers. In line with the overall negative results of the SCIENCE study, with the exception of one miR, we did not detect any significant regulation of small EV miRNAs in this patient collective.
Collapse
Affiliation(s)
- Denise Traxler
- Division of Cardiology, Department of Internal Medicine II and Department of Oral and Maxillofacial Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Varius Dannenberg
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Katrin Zlabinger
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Alfred Gugerell
- Division of Cardiology, Department of Internal Medicine II, Department of Thoracic Surgery, Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Medical University of Vienna, 1090 Vienna, Austria
| | - Julia Mester-Tonczar
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Dominika Lukovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Spannbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Ena Hasimbegovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Jens Kastrup
- Cardiology Stem Cell Centre, Department of Cardiology, Centre for Cardiac, Vascular, Pulmonary and Infectious Diseases, Rigshospitalet, University of Copenhagen, Henrik Harpestrengs Vej 4, 2100 Copenhagen, Denmark
| | - Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Pelisek J, Reutersberg B, Greber UF, Zimmermann A. Vascular dysfunction in COVID-19 patients: update on SARS-CoV-2 infection of endothelial cells and the role of long non-coding RNAs. Clin Sci (Lond) 2022; 136:1571-1590. [PMID: 36367091 PMCID: PMC9652506 DOI: 10.1042/cs20220235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 08/16/2023]
Abstract
Although COVID-19 is primarily a respiratory disease, it may affect also the cardiovascular system. COVID-19 patients with cardiovascular disorder (CVD) develop a more severe disease course with a significantly higher mortality rate than non-CVD patients. A common denominator of CVD is the dysfunction of endothelial cells (ECs), increased vascular permeability, endothelial-to-mesenchymal transition, coagulation, and inflammation. It has been assumed that clinical complications in COVID-19 patients suffering from CVD are caused by SARS-CoV-2 infection of ECs through the angiotensin-converting enzyme 2 (ACE2) receptor and the cellular transmembrane protease serine 2 (TMPRSS2) and the consequent dysfunction of the infected vascular cells. Meanwhile, other factors associated with SARS-CoV-2 entry into the host cells have been described, including disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), the C-type lectin CD209L or heparan sulfate proteoglycans (HSPG). Here, we discuss the current data about the putative entry of SARS-CoV-2 into endothelial and smooth muscle cells. Furthermore, we highlight the potential role of long non-coding RNAs (lncRNAs) affecting vascular permeability in CVD, a process that might exacerbate disease in COVID-19 patients.
Collapse
Affiliation(s)
- Jaroslav Pelisek
- Department of Vascular Surgery, University Hospital Zürich, Zürich, Switzerland
| | | | - Urs F Greber
- Department of Molecular Life Sciences, University of Zürich, Switzerland
| | | |
Collapse
|
7
|
Functional Association of miR-133b and miR-21 Through Novel Gene Targets ATG5, LRP6 and SGPP1 in Coronary Artery Disease. Mol Diagn Ther 2022; 26:655-664. [PMID: 36197604 DOI: 10.1007/s40291-022-00615-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Atherosclerosis, a progressive manifestation of coronary artery disease, has been observed to be regulated by microRNAs (miRNAs) targeting various protein-coding genes involved in several pathophysiological events of coronary artery disease. OBJECTIVE In our previous report, we identified differential expression profiles of candidate miRNAs, miR-133b and miR-21, in patients with coronary artery disease as compared with controls, suggesting their possible implication in the pathophysiology of coronary artery disease. To better understand the functional role of these miRNAs, we sought to predict and validate their target genes while assessing the expression pattern of these genes in patients with coronary artery disease, as well as in macrophages. METHODS Potential target genes of miR-133b and miR-21 were predicted bioinformatically followed by validation through the identification of their expression at the protein level in patients with coronary artery disease (n-30), as well as in macrophages treated with respective miRNA inhibitors (antagomiRs), through immunoblotting. RESULTS SGPP1, a gene associated with the sphingolipid pathway, was predicted to be a potential target gene of miR-133b while ATG5 and LRP6 were target genes of miR-21 while being associated with autophagy and Wnt signalling pathways, respectively. SGPP1 was observed to be upregulated significantly (p = 0.019) by 2.07-fold, whereas ATG5 and LRP6 were found to be downregulated (p = 0.026 and 0.007, respectively) by 3.28-fold and 8.46-fold, respectively, in patients with coronary artery disease as compared with controls. Expression patterns of all the genes were also found to be modulated when cells were treated with respective miRNA inhibitors. CONCLUSIONS Results from the present study suggest that SGPP1, ATG5 and LRP6, target genes of miR-133b and miR-21, may serve as potential therapeutic hotspots in the management of coronary artery disease, which undoubtedly merit further experimental confirmation.
Collapse
|
8
|
Rincón LM, Rodríguez-Serrano M, Conde E, Lanza VF, Sanmartín M, González-Portilla P, Paz-García M, Del Rey JM, Menacho M, García Bermejo ML, Zamorano JL. Serum microRNAs are key predictors of long-term heart failure and cardiovascular death after myocardial infarction. ESC Heart Fail 2022; 9:3367-3379. [PMID: 35837763 DOI: 10.1002/ehf2.13919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/09/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Patients with acute myocardial infarction (MI) are at high risk of upcoming events, in particular heart failure (HF), but reliable stratification methods are lacking. Our goal was to evaluate the potential role of circulating miRNAs as prognostic biomarkers in patients presenting with MI. METHODS AND RESULTS We conducted a prospective study among 311 consecutive patients hospitalized with MI (65% ST-segment elevation MI & median age of 55 years) with long-term follow-up. An initial screening was conducted to select candidate miRNAs, with subsequent study of 14 candidate miRNAs. The primary outcome was the composite of hospital admission for HF or cardiovascular death. During a mean follow-up of 2.1 years miR-21-5p, miR-23a-3p, miR27b-3p, miR-122-5p, miR210-3p, and miR-221-3p reliably predicted the primary outcome. Multivariate Cox regression analyses highlighted that miR-210-3p [hazard ratio (HR) 2.65 per 1 SD increase, P < 0.001], miR-23a-3p (HR 2.11 per 1 SD increase, P < 0.001), and miR-221-3p (HR 2.03 per 1 SD increase, P < 0.001) were able to accurately predict the primary outcome, as well as cardiovascular death, HF hospitalizations, and long-term New York Heart Association (NYHA) functional class. These three miRNAs clearly improved the performance of multivariate clinical models: ΔC-statistic = 0.10 [95% confidence interval (CI), 0.03-0.17], continuous net reclassification index = 34.8% (95%CI, 5.8-57.4%), and integrated discrimination improvement (P < 0.001). CONCLUSIONS This is the largest study evaluating the prognostic value of circulating miRNAs for HF-related events among patients with MI. We show that several miRNAs predict HF hospitalizations, cardiovascular mortality, and poor long-term NYHA status and improve current risk prediction methods.
Collapse
Affiliation(s)
- Luis M Rincón
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Macarena Rodríguez-Serrano
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - Elisa Conde
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - Val F Lanza
- Bioinformatics Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marcelo Sanmartín
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
| | - Paz González-Portilla
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
| | - Marta Paz-García
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - José Manuel Del Rey
- Department of Biochemistry, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Miriam Menacho
- Department of Biochemistry, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María-Laura García Bermejo
- Biomarkers and Therapeutic Targets Laboratory and Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spanish Renal Research Network (REDinREN), Ctra. Colmenar Km 9100, Madrid, 28034, Spain
| | - José L Zamorano
- Department of Cardiology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, 28034, Spain
- Hospital La Zarzuela, Madrid, Spain
| |
Collapse
|
9
|
Lam K, Kasavajhala K, Gunasekera S, Simmerling C. Accelerating the Ensemble Convergence of RNA Hairpin Simulations with a Replica Exchange Structure Reservoir. J Chem Theory Comput 2022; 18:3930-3947. [PMID: 35502992 PMCID: PMC10658646 DOI: 10.1021/acs.jctc.2c00065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
RNA is a key participant in many biological processes, but studies of RNA using computer simulations lag behind those of proteins, largely due to less-developed force fields and the slow dynamics of RNA. Generating converged RNA ensembles for force field development and other studies remains a challenge. In this study, we explore the ability of replica exchange molecular dynamics to obtain well-converged conformational ensembles for two RNA hairpin systems in an implicit solvent. Even for these small model systems, standard REMD remains computationally costly, but coupling to a pre-generated structure library using the reservoir REMD approach provides a dramatic acceleration of ensemble convergence for both model systems. Such precise ensembles could facilitate RNA force field development and validation and applications of simulation to more complex RNA systems. The advantages and remaining challenges of applying R-REMD to RNA are investigated in detail.
Collapse
Affiliation(s)
- Kenneth Lam
- Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794, United States
| | - Koushik Kasavajhala
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794, United States
| | - Sarah Gunasekera
- Program in Biology, Stony Brook University, Stony Brook, New York 11794, United States
| | - Carlos Simmerling
- Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York 11794, United States
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
10
|
Zhang MX, Song Y, Xu WL, Zhang LX, Li C, Li YL. Natural Herbal Medicine as a Treatment Strategy for Myocardial Infarction through the Regulation of Angiogenesis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8831750. [PMID: 35600953 PMCID: PMC9119779 DOI: 10.1155/2022/8831750] [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: 01/17/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
Abstract
Methods We conducted a literature search on the bioactive components of medicinal plants and their effects on angiogenesis after MI. We searched for articles in Web of Science, MEDLINE, PubMed, Scopus, Google Scholar, and China National Knowledge Infrastructure databases before April 2021. Results In this article, we summarized the mechanisms by which copper ions, microRNA, Akt1, inflammation, oxidative stress, mitochondria, and pericytes are involved in angiogenesis after myocardial infarction. In addition, we reviewed the angiogenic effects of natural herbal medicines such as Salvia miltiorrhiza Bunge Bunge, Carthamus tinctorius L., Pueraria lobata, Astragalus, Panax ginseng C.A. Mey., Panax notoginseng (Burkill) F.H. Chen, Cinnamomum cassia (L.) J. Presl, Rehmannia glutinosa (Gaertn.) DC., Leonurus japonicus Houtt, Scutellaria baicalensis Georgi., and Geum macrophyllum Willd. Conclusions Some herbs have the effect of promoting angiogenesis. In the future, natural proangiogenic drugs may become candidates for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Mu-xin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yu Song
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wan-li Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ling-xiao Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yun-lun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
11
|
Ball HC, Alejo AL, Kronk T, Alejo AM, Safadi FF. Epigenetic Regulation of Chondrocytes and Subchondral Bone in Osteoarthritis. Life (Basel) 2022; 12:582. [PMID: 35455072 PMCID: PMC9030470 DOI: 10.3390/life12040582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 12/24/2022] Open
Abstract
The aim of this review is to provide an updated review of the epigenetic factors involved in the onset and development of osteoarthritis (OA). OA is a prevalent degenerative joint disease characterized by chronic inflammation, ectopic bone formation within the joint, and physical and proteolytic cartilage degradation which result in chronic pain and loss of mobility. At present, no disease-modifying therapeutics exist for the prevention or treatment of the disease. Research has identified several OA risk factors including mechanical stressors, physical activity, obesity, traumatic joint injury, genetic predisposition, and age. Recently, there has been increased interest in identifying epigenetic factors involved in the pathogenesis of OA. In this review, we detail several of these epigenetic modifications with known functions in the onset and progression of the disease. We also review current therapeutics targeting aberrant epigenetic regulation as potential options for preventive or therapeutic treatment.
Collapse
Affiliation(s)
- Hope C. Ball
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (A.L.A.); (T.K.); (A.M.A.)
- Musculoskeletal Research Group, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Andrew L. Alejo
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (A.L.A.); (T.K.); (A.M.A.)
- Musculoskeletal Research Group, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Trinity Kronk
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (A.L.A.); (T.K.); (A.M.A.)
- Musculoskeletal Research Group, Northeast Ohio Medical University, Rootstown, OH 44272, USA
- GPN Therapeutics, Inc., REDI Zone, Rootstown, OH 44272, USA
| | - Amanda M. Alejo
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (A.L.A.); (T.K.); (A.M.A.)
- Musculoskeletal Research Group, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Fayez F. Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (A.L.A.); (T.K.); (A.M.A.)
- Musculoskeletal Research Group, Northeast Ohio Medical University, Rootstown, OH 44272, USA
- Department of Orthopaedic Surgery, Akron Children’s Hospital, Akron, OH 44308, USA
| |
Collapse
|
12
|
Pan HT, Shi XL, Fang M, Sun XM, Chen PP, Ding JL, Xia GY, Yu B, Zhang T, Zhu HD. Profiling of exosomal microRNAs expression in umbilical cord blood from normal and preeclampsia patients. BMC Pregnancy Childbirth 2022; 22:124. [PMID: 35152894 PMCID: PMC8842963 DOI: 10.1186/s12884-022-04449-w] [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: 09/10/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023] Open
Abstract
Background Epidemiological and experimental studies suggest that preeclampsia has a negative impact on maternity and offspring health. Previous studies report that dysregulation in utero-environment increases risk for elderly disease such as cardiovascular disease. However, the underlying mechanisms remain elusive. Specific microRNAs (miRNAs) are packaged in exosomes may regulate microvascular dysfunction in offspring of mothers with preeclampsia. The present study aimed to identify the differential expression profiles of microRNAs in the serum exosomes between patients with preeclampsia and normal pregnancies. Methods A comprehensive miRNA sequence-based approach was performed to compare exosomes carry miRNAs (Exo-miRNAs) expression levels in umbilical serum between normal and preeclampsia patients. Exosomes were isolated using the ExoQuick precipitation kit. Serum exosomes were then viewed under electron microscopy, and their characteristics determined by western blotting and nanoparticle-tracking analysis. Illumina platform was used to perform sequencing. Bioinformatics analysis was used to explore differentially expressed Exo-miRNAs in umbilical serum. Results Based on sequence similarity, 1733 known miRNAs were retrieved. Furthermore, 157 mature miRNAs in serum exosomes were significantly differential expressed between PE and those control groups (P<0.05, log2|FC| > 1). Out, of the 157 miRNAs, 96 were upregulated miRNAs whereas 61 miRNAs were downregulated. The 157 differentially expressed miRNAs targeted 51,424 differentially expressed genes. Functional analysis through KEGG pathway and Gene Ontology results uncovered that target genes of miRNAs with differential expression were significantly linked to several pathways and biological processes. Conclusion The findings of this study showed differential expression of umbilical serum Exo-miRNAs in normal compared with PE patients, implying that these Exo-miRNAs may associate with microvascular dysfunction in offspring of mothers with preeclampsia. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04449-w.
Collapse
Affiliation(s)
- Hai-Tao Pan
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.,The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiao-Liang Shi
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Min Fang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Xiang-Mei Sun
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Pan-Pan Chen
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Jin-Long Ding
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Gui-Yu Xia
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Bin Yu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Tao Zhang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China. .,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.
| | - Hong-Dan Zhu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China. .,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.
| |
Collapse
|
13
|
Yan N, Xiao C, Wang X, Xu Z, Yang J. Tanshinone IIA from Salvia miltiorrhiza exerts anti-fibrotic effects on cardiac fibroblasts and rat heart tissues by suppressing the levels of pro-fibrotic factors: The key role of miR-618. J Food Biochem 2022; 46:e14078. [PMID: 35014054 DOI: 10.1111/jfbc.14078] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/11/2021] [Accepted: 12/24/2021] [Indexed: 11/27/2022]
Abstract
Tanshinone IIA (TAN) is widely employed for handling cardiovascular disorders. The current study explored the potential role of miRs in the antifibrotic effect of TAN on heart. Fibrotic features were induced in cardiac fibroblasts (CFs) and in rat hearts, and then handled with TAN. MicroRNAs (miRs) responding to TAN were determined using a microarray assay. The selected miR was modulated to verify its role in antifibrotic effects of TAN. TAN suppressed the viability and the production of α-SMA in CFs, which was associated with 101 miR being upregulated and 223 miR being downregulated. MiR-618 was selected as the potential target of TAN. Ang II inhibited miR-618 level and resulted in the upregulation of pro-fibrosis factors, which was reversed by TAN. The antifibrotic effect of TAN was weakened by miR-618 inhibition. TAN inhibits hypertrophy and collagen deposition in heart tissues, which is associated with the increased level of miR-618. PRACTICAL APPLICATIONS: The findings outlined in the current study show that the antifibrotic function of TAN is closely related to the function of miRs: the induction of miR-618 is indispensable for the function of TAN against the fibrotic process after heart injury, which will promote the application of TAN as an adjuvant therapy for improving heart function.
Collapse
Affiliation(s)
- Na Yan
- Second Department of Cardiology, Ganzhou People's Hospital, Ganzhou, China
| | - Chunqing Xiao
- Second Department of Cardiology, Ganzhou People's Hospital, Ganzhou, China
| | - Xianggui Wang
- Second Department of Cardiology, Ganzhou People's Hospital, Ganzhou, China
| | - Zufang Xu
- Second Department of Cardiology, Ganzhou People's Hospital, Ganzhou, China
| | - Jiangyong Yang
- Department of Cardiology, Ganzhou Municipal Hospital, Ganzhou, China
| |
Collapse
|
14
|
Yin X, Yao D, Lam MHW, Liang H. A facile biosynthesis strategy of plasmid DNA-derived nanowires for readable microRNA logic operations. J Mater Chem B 2022; 10:3055-3063. [DOI: 10.1039/d1tb02699b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multiple microRNAs (miRNAs) logical assays have attracted wide attention recently, which can be applied to mimic and reveal cellular events at the molecular level. However, it remains challenging to develop...
Collapse
|
15
|
Tsantilas P, Lao S, Wu Z, Eberhard A, Winski G, Vaerst M, Nanda V, Wang Y, Kojima Y, Ye J, Flores A, Jarr KU, Pelisek J, Eckstein HH, Matic L, Hedin U, Tsao PS, Paloschi V, Maegdefessel L, Leeper NJ. Chitinase 3 like 1 is a regulator of smooth muscle cell physiology and atherosclerotic lesion stability. Cardiovasc Res 2021; 117:2767-2780. [PMID: 33471078 PMCID: PMC8848327 DOI: 10.1093/cvr/cvab014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/17/2020] [Accepted: 02/07/2021] [Indexed: 12/13/2022] Open
Abstract
AIMS Atherosclerotic cerebrovascular disease underlies the majority of ischaemic strokes and is a major cause of death and disability. While plaque burden is a predictor of adverse outcomes, plaque vulnerability is increasingly recognized as a driver of lesion rupture and risk for clinical events. Defining the molecular regulators of carotid instability could inform the development of new biomarkers and/or translational targets for at-risk individuals. METHODS AND RESULTS Using two independent human endarterectomy biobanks, we found that the understudied glycoprotein, chitinase 3 like 1 (CHI3L1), is up-regulated in patients with carotid disease compared to healthy controls. Further, CHI3L1 levels were found to stratify individuals based on symptomatology and histopathological evidence of an unstable fibrous cap. Gain- and loss-of-function studies in cultured human carotid artery smooth muscle cells (SMCs) showed that CHI3L1 prevents a number of maladaptive changes in that cell type, including phenotype switching towards a synthetic and hyperproliferative state. Using two murine models of carotid remodelling and lesion vulnerability, we found that knockdown of Chil1 resulted in larger neointimal lesions comprised by de-differentiated SMCs that failed to invest within and stabilize the fibrous cap. Exploratory mechanistic studies identified alterations in potential downstream regulatory genes, including large tumour suppressor kinase 2 (LATS2), which mediates macrophage marker and inflammatory cytokine expression on SMCs, and may explain how CHI3L1 modulates cellular plasticity. CONCLUSION CHI3L1 is up-regulated in humans with carotid artery disease and appears to be a strong mediator of plaque vulnerability. Mechanistic studies suggest this change may be a context-dependent adaptive response meant to maintain vascular SMCs in a differentiated state and to prevent rupture of the fibrous cap. Part of this effect may be mediated through downstream suppression of LATS2. Future studies should determine how these changes occur at the molecular level, and whether this gene can be targeted as a novel translational therapy for subjects at risk of stroke.
Collapse
MESH Headings
- Animals
- Carotid Arteries/enzymology
- Carotid Arteries/pathology
- Carotid Arteries/physiopathology
- Carotid Artery Diseases/enzymology
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/pathology
- Carotid Artery Diseases/physiopathology
- Cell Differentiation
- Cells, Cultured
- Chitinase-3-Like Protein 1/genetics
- Chitinase-3-Like Protein 1/metabolism
- Disease Models, Animal
- Fibrosis
- Humans
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- Plaque, Atherosclerotic
- Rupture, Spontaneous
- Vascular Remodeling
- Mice
Collapse
Affiliation(s)
- Pavlos Tsantilas
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
| | - Shen Lao
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou 510120, China
| | - Zhiyuan Wu
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
| | - Anne Eberhard
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Greg Winski
- Department of Medicine, Karolinska Institute, Stockholm, Solnavägen 1, 171 77 Solna, Sweden
| | - Monika Vaerst
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Vivek Nanda
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Ying Wang
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Alyssa Flores
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| | - Jaroslav Pelisek
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
- Department for Vascular Surgery, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Potsdamer Str. 58, 10785 Berlin, Germany, partner site Munich Heart Alliance
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Solnavägen 1, 171 77 Solna, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Solnavägen 1, 171 77 Solna, Sweden
| | - Philip S Tsao
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, 870 Quarry Road, Stanford, CA 94305, USA
- Veterans Affairs (VA) Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, USA
| | - Valentina Paloschi
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Potsdamer Str. 58, 10785 Berlin, Germany, partner site Munich Heart Alliance
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaningerstr. 22, 81675 Munich, Germany
- Department of Medicine, Karolinska Institute, Stockholm, Solnavägen 1, 171 77 Solna, Sweden
- German Center for Cardiovascular Research (DZHK), Potsdamer Str. 58, 10785 Berlin, Germany, partner site Munich Heart Alliance
| | - Nicholas J Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Alway Bldg., M121 Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, 265 Campus Drive Stanford, CA 94305, USA
| |
Collapse
|
16
|
Ali F, Shen A, Islam W, Saleem MZ, Muthu R, Xie Q, Wu M, Cheng Y, Chu J, Lin W, Peng J. Role of MicroRNAs and their corresponding ACE2/Apelin signaling pathways in hypertension. Microb Pathog 2021; 162:105361. [PMID: 34919993 DOI: 10.1016/j.micpath.2021.105361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/12/2021] [Accepted: 12/12/2021] [Indexed: 11/28/2022]
Abstract
Hypertension is controlled via the alteration of microRNAs (miRNAs), their therapeutic targets angiotensin II type I receptor (AT1R) and cross talk of signaling pathways. The stimulation of the Ang II/AT1R pathway by deregulation of miRNAs, has also been linked to cardiac remodeling as well as the pathophysiology of high blood pressure. As miRNAs have been associated to ACE2/Apelin and Mitogen-activated protein kinases (MAPK) signaling, it has revealed an utmost protective impact over hypertension and cardiovascular system. The ACE2-coupled intermodulation between RAAS, Apelin system, MAPK signaling pathways, and miRNAs reveal the practicalities of high blood pressure. The research of miRNAs may ultimately lead to the expansion of an innovative treatment strategy for hypertension, which indicates the need to explore them further at the molecular level. Therefore, here we have focused on the mechanistic importance of miRNAs in hypertension, ACE2/Apelin signaling as well as their biological functions, with a focus on interplay and crosstalk between ACE2/Apelin signaling, miRNAs, and hypertension, and the progress in miRNA-based diagnostic techniques with the goal of facilitating the development of new hypertension-controlling therapeutics.
Collapse
Affiliation(s)
- Farman Ali
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Waqar Islam
- Xinjiang Key Laboratory of Desert Plant Roots Ecology and Vegetation Restoration, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi, 830011, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Ragunath Muthu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Jiangfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Wei Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Chen Keji Academic Thought Inheritance Studio, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
17
|
Emmi G, Bagni G, Lastraioli E, Di Patti F, Bettiol A, Fiorillo C, Becatti M, Silvestri E, Urban ML, Emmi L, Prisco D, Arcangeli A. A unique circulating miRNA profile highlights thrombo-inflammation in Behçet's syndrome. Ann Rheum Dis 2021; 81:386-397. [PMID: 34844932 PMCID: PMC8862064 DOI: 10.1136/annrheumdis-2021-220859] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Objectives Behçet’s syndrome (BS) is a rare systemic vasculitis often complicated by thrombotic events. Given the lack of validated biomarkers, BS diagnosis relies on clinical criteria. In search of novel biomarkers for BS diagnosis, we determined the profile of plasmatic circulating microRNAs (ci-miRNAs) in patients with BS compared with healthy controls (HCs). Methods ci-miRNA profile was evaluated by microarray in a screening cohort (16 patients with BS and 18 HCs) and then validated by poly(T) adaptor PCR (PTA-PCR) in a validation cohort (30 patients with BS and 30 HCs). Two disease control groups (30 patients with systemic lupus erythematosus (SLE) and 30 patients with giant cell arteritis (GCA) were also analysed. Results From the microarray screening, 29 deregulated (differentially expressed (DE)) human ci-miRNAs emerged. A hierarchical cluster analysis indicated that DE ci-miRNAs clearly segregated patients from controls, independently of clinical features. PTA-PCR analysis on the validation cohort confirmed the deregulation of miR-224-5p, miR-206 and miR-653-5p. The combined receiver operating characteristic (ROC) curve analyses showed that such ci-miRNAs discriminate BS from HCs (and BS with active vs inactive disease), as well as BS from patients with SLE and GCA. The functional annotation analyses (FAAs) showed that the most enriched pathways affected by DE ci-miRNAs (ie, cell–matrix interaction, oxidative stress and blood coagulation) are related to thrombo-inflammatory mechanisms. Accordingly, the expression of the three ci-miRNAs from the validation cohort significantly correlated with leucocyte reactive oxygen species production and plasma lipid peroxidation. Conclusions The ci-miRNA profile identified in this study may represent a novel, poorly invasive BS biomarker, while suggesting an epigenetic control of BS-related thrombo-inflammation.
Collapse
Affiliation(s)
- Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.,SOD Interdisciplinary Internal Medicine-Behçet Center and Lupus Clinic-Azienda Ospedaliero Universitaria Careggi, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Giacomo Bagni
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Elena Lastraioli
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Francesca Di Patti
- Department of Mathematics and Informatics, University of Perugia, Perugia, Italy.,Center for the Study of Complex Dynamics, University of Florence, Firenze, Italy.,Department of Physics, University of Florence, Florence, Italy
| | - Alessandra Bettiol
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.,SOD Interdisciplinary Internal Medicine-Behçet Center and Lupus Clinic-Azienda Ospedaliero Universitaria Careggi, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Maria Letizia Urban
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.,SOD Interdisciplinary Internal Medicine-Behçet Center and Lupus Clinic-Azienda Ospedaliero Universitaria Careggi, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Lorenzo Emmi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Firenze, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.,SOD Interdisciplinary Internal Medicine-Behçet Center and Lupus Clinic-Azienda Ospedaliero Universitaria Careggi, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy .,Center for the Study of Complex Dynamics, University of Florence, Firenze, Italy
| |
Collapse
|
18
|
Lamsisi M, Wakrim L, Bouziyane A, Benhessou M, Oudghiri M, Laraqui A, Elkarroumi M, Ennachit M, El Mzibri M, Ennaji MM. The Biological Significance of Long noncoding RNAs Dysregulation and their Mechanism of Regulating Signaling Pathways in Cervical Cancer. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:75-101. [PMID: 34703793 PMCID: PMC8496250 DOI: 10.22088/ijmcm.bums.10.2.75] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/01/2021] [Indexed: 12/19/2022]
Abstract
Despite the remarkable decrease in cervical cancer incidence due to the availability of the HPV vaccine and implementation of screening programs for early detection in developed countries, this cancer remains a major health problem globally, especially in developing countries where most of the cases and mortality occur. Therefore, more understanding of molecular mechanisms of cervical cancer development might lead to the discovery of more effective diagnosis and treatment options. Research on long noncoding RNAs (lncRNAs) demonstrates the important roles of these molecules in many physiological processes and diseases, especially cancer. In the present review, we discussed the significance of lncRNAs altered expression in cervical cancer, highlighting their roles in regulating highly conserved signaling pathways, such as mitogen-activated protein kinase (MAPK), Wnt/β-catenin, Notch, and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathways and their association with the progression of cervical cancer in order to bring more insight and understanding of this disease and their potential implications in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Maryame Lamsisi
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
| | - Lahcen Wakrim
- Laboratory of Virology, Pasteur Institute of Morocco. Casablanca, Morocco.
| | - Amal Bouziyane
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
- University Mohammed VI of Health Science, Casablanca, Morocco.
| | - Mustapha Benhessou
- Team of Virology, Oncology and Medical Biotechnologies, Laboratory of Virology, Microbiology, Quality, and Biotechnologies/ ETB. Faculty of Science and Techniques Mohammedia, Hassan II University of Casablanca, Morocco.
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | - Mounia Oudghiri
- Immunology and Biodiversity laboratory, Faculty of Sciences Ain chock, Hassan II University of Casablanca, Morocco.
| | - Abdelilah Laraqui
- Research and Biosafety Laboratory, Mohammed V Military Hospital, University Mohammed V of Rabat, Morocco.
| | - Mohamed Elkarroumi
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | - Mohammed Ennachit
- School of Medicine and Pharmacy, University Hassan II of Casablanca, Morocco.
| | | | - Moulay Mustapha Ennaji
- Corresponding author: Faculty of Science and Techniques Mohammedia, University Hassan II of Casablanca, Morocco. E-mail:
| |
Collapse
|
19
|
Li C, Wang N, Rao P, Wang L, Lu D, Sun L. Role of the microRNA-29 family in myocardial fibrosis. J Physiol Biochem 2021; 77:365-376. [PMID: 34047925 DOI: 10.1007/s13105-021-00814-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 04/01/2021] [Indexed: 12/11/2022]
Abstract
Myocardial fibrosis (MF) is an inevitable pathological process in the terminal stage of many cardiovascular diseases, often leading to serious cardiac dysfunction and even death. Currently, microRNA-29 (miR-29) is thought to be a novel diagnostic and therapeutic target of MF. Understanding the underlying mechanisms of miR-29 that regulate MF will provide a new direction for MF therapy. In the present review, we concentrate on the underlying signaling pathway of miR-29 affecting MF and the crosstalk regulatory relationship among these pathways to illustrate the complex regulatory network of miR-29 in MF. Additionally, based on our mechanistic understanding, we summarize opportunities and challenges of miR-29-based MF diagnosis and therapy.
Collapse
Affiliation(s)
- Changyan Li
- Science and Technology Achievement Incubation Center, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Nan Wang
- Science and Technology Achievement Incubation Center, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Peng Rao
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Limeiting Wang
- Science and Technology Achievement Incubation Center, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China
| | - Di Lu
- Science and Technology Achievement Incubation Center, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan, China.
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China.
| |
Collapse
|
20
|
Ramzan F, Vickers MH, Mithen RF. Epigenetics, microRNA and Metabolic Syndrome: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22095047. [PMID: 34068765 PMCID: PMC8126218 DOI: 10.3390/ijms22095047] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Epigenetics refers to the DNA chemistry changes that result in the modification of gene transcription and translation independently of the underlying DNA coding sequence. Epigenetic modifications are reported to involve various molecular mechanisms, including classical epigenetic changes affecting DNA methylation and histone modifications and small RNA-mediated processes, particularly that of microRNAs. Epigenetic changes are reversible and are closely interconnected. They are recognised to play a critical role as mediators of gene regulation, and any alteration in these mechanisms has been identified to mediate various pathophysiological conditions. Moreover, genetic predisposition and environmental factors, including dietary alterations, lifestyle or metabolic status, are identified to interact with the human epigenome, highlighting the importance of epigenetic factors as underlying processes in the aetiology of various diseases such as MetS. This review will reflect on how both the classical and microRNA-regulated epigenetic changes are associated with the pathophysiology of metabolic syndrome. We will then focus on the various aspects of epigenetic-based strategies used to modify MetS outcomes, including epigenetic diet, epigenetic drugs, epigenome editing tools and miRNA-based therapies.
Collapse
|
21
|
MicroRNA-15a modulates lens epithelial cells apoptosis and proliferation through targeting B-cell lymphoma-2 and E2F transcription factor 3 in age-related cataracts. Biosci Rep 2020; 39:221172. [PMID: 31737898 PMCID: PMC6900469 DOI: 10.1042/bsr20191773] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/25/2019] [Accepted: 11/07/2019] [Indexed: 12/17/2022] Open
Abstract
Age-related cataract remains a serious problem in the aged over the world. MicroRNAs are abnormally expressed in various diseases including age-related cataract. MicroRNA-15a (MicroRNA-15a) has been involved in various diseases and plays crucial roles in many cellular processes. However, the mechanism of microRNA-15a in the genesis of cataract remains barely known. We therefore aimed to investigate the role of microRNA-15a in the cataract. Herein, human lens epithelial B3 cells, HLE-B3 cells were treated with 200 μmol/l H2O2 for 24 h. H2O2 was utilized in our study to induce HLE-B3 cells injury. We observed that cell apoptosis was induced by the treatment of H2O2 and meanwhile, cell proliferation was repressed by 200 μmol/l H2O2. Then, it was found that microRNA-15a was significantly increased with the H2O2 exposure in vitro. Importantly, B-cell lymphoma-2 (BCL2) and E2F transcription factor 3 (E2F3) exert crucial roles in cell apoptosis and cell proliferation. We found that BCL2 and E2F3 were greatly reduced by 200 μmol/l H2O2 in human lens epithelial cells. In addition, microRNA-15a overexpression induced cell apoptosis and repressed cell proliferation through suppressing BCL2 and E2F3. Subsequently, BCL2 and E2F3 were predicted as a direct target of microRNA-15a. The direct correlation between microRNA-15a and BCL2/E2F3 was confirmed by dual luciferase reporter assay. In conclusion, we demonstrated that microRNA-15a triggered apoptosis and repressed the proliferation of HLE-B3 cells by modulating BCL2 and E2F3.
Collapse
|
22
|
Yan Y, Song D, Zhang X, Hui G, Wang J. GEO Data Sets Analysis Identifies COX-2 and Its Related Micro RNAs as Biomarkers for Non-Ischemic Heart Failure. Front Pharmacol 2020; 11:1155. [PMID: 32848764 PMCID: PMC7419645 DOI: 10.3389/fphar.2020.01155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/15/2020] [Indexed: 01/06/2023] Open
Abstract
Heart failure (HF) is a heterogeneous clinical syndrome with a variety of causes, risk factors, and pathology. Clinically, only brain natriuretic peptide (BNP) or its precursor N-terminus proBNP (NTproBNP) has been validated for HF diagnosis, but they are also affected by other conditions, such as female gender, renal disease, and acute coronary syndromes, and false low levels in the setting of obesity or flash pulmonary edema. In addition, there is no one biomarker which could encompass all heart failure phenotypes. Advances in bioinformatics have provided us with large databases that characterize the complex genetic and epigenetic changes associated with human diseases. The use of data mining strategies on public access databases to identify previously unknown disease markers is an innovative approach to identify potential biomarkers or even new therapeutic targets in complex diseases such as heart failure (HF). In this study, we analyzed the genomic and transcription data of HF peripheral blood mononuclear cell (PBMC) samples obtained from the Gene Expression Omnibus data sets using Omicsbean online database (http://www.omicsbean.cn/) and found that the prostaglandin-endoperoxide synthase 2 (PTGS2), also named as cyclooxygenase-2 (COX-2), as well as its related micro RNAs including miR-1297 and miR-4649-3p might be used as potential biomarkers for non-ischemic heart failure. Our result showed that plasma COX-2 and miR-4649-3p were significantly up-regulated, whereas the plasma miR-1297 was significantly decreased, and miR-4649-3p displayed high predictive power for non-ischemic heart failure.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
| | - Gang Hui
- The Department of 31656 Troops of Chinese People's Liberation Army, Leshan, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Sabre L, Punga T, Punga AR. Circulating miRNAs as Potential Biomarkers in Myasthenia Gravis: Tools for Personalized Medicine. Front Immunol 2020; 11:213. [PMID: 32194544 PMCID: PMC7065262 DOI: 10.3389/fimmu.2020.00213] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease caused by antibodies which attack receptors at the neuromuscular junction. One of the main difficulties in predicting the clinical course of MG is the heterogeneity of the disease, where disease progression differs greatly depending on the subgroup that the patient is classified into. MG subgroups are classified according to: age of onset [early-onset MG (EOMG; onset ≤ 50 years) versus late-onset MG (LOMG; onset > 50 years]; the presence of a thymoma (thymoma-associated MG); antibody subtype [acetylcholine receptor antibody seropositive (AChR+) and muscle-specific tyrosine kinase antibody seropositive (MuSK+)]; as well as clinical subtypes (ocular versus generalized MG). The diagnostic tests for MG, such as antibody titers, neurophysiological tests, and objective clinical fatigue score, do not necessarily reflect disease progression. Hence, there is a great need for reliable objective biomarkers in MG to follow the disease course as well as the individualized response to therapy toward personalized medicine. In this regard, circulating microRNAs (miRNAs) have emerged as promising potential biomarkers due to their accessibility in body fluids and unique profiles in different diseases, including autoimmune disorders. Several studies on circulating miRNAs in MG subtypes have revealed specific miRNA profiles in patients’ sera. In generalized AChR+ EOMG, miR-150-5p and miR-21-5p are the most elevated miRNAs, with lower levels observed upon treatment with immunosuppression and thymectomy. In AChR+ generalized LOMG, the miR-150-5p, miR-21-5p, and miR-30e-5p levels are elevated and decrease in accordance with the clinical response after immunosuppression. In ocular MG, higher levels of miR-30e-5p discriminate patients who will later generalize from those remaining ocular. In contrast, in MuSK+ MG, the levels of the let-7 miRNA family members are elevated. Studies of circulating miRNA profiles in Lrp4 or agrin antibody-seropositive MG are still lacking. This review summarizes the present knowledge of circulating miRNAs in different subgroups of MG.
Collapse
Affiliation(s)
- Liis Sabre
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia.,Department of Neuroscience, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Tanel Punga
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anna Rostedt Punga
- Department of Neuroscience, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
24
|
Abstract
Cardiovascular diseases are one of the most common causes of death in both developing and developed countries worldwide. Even though there have been improvements in primary prevention, the prevalence of cardiovascular diseases continues to increase in recent years. Hence, it is crucial to both investigate the molecular pathophysiology of cardiovascular diseases in-depth and find novel biomarkers regarding the early and proper prevention and diagnosis of these diseases. MicroRNAs, or miRNAs, are endogenous, conserved, single-stranded non-coding RNAs of 21-25 nucleotides in length. miRNAs have important roles in various cellular events such as embryogenesis, proliferation, vasculogenesis, apoptosis, cell growth, differentiation, and tumorigenesis. They also have potential roles in the cardiovascular system, including angiogenesis, cardiac cell contractility, control of lipid metabolism, plaque formation, the arrangement of cardiac rhythm, and cardiac cell growth. Circulating miRNAs are promising novel biomarkers for purposes of the diagnosis and prognosis of cardiovascular diseases. Cell or tissue specificity, stability in serum or plasma, resistance to degradative factors such as freeze-thaw cycles or enzymes in the blood, and fast-release kinetics, provide the potential for miRNAs to be surrogate markers for the early and accurate diagnosis of disease and for predicting middle- or long-term prognosis. Moreover, it may be a logical approach to combine miRNAs with traditional biomarkers to improve risk stratification and long-term prognosis. In addition to their efficacy in both diagnosis and prognosis, miRNA-based therapeutics may be beneficial for treating cardiovascular diseases using novel platforms and computational tools and in combination with traditional methods of analysis. microRNAs are promising, novel therapeutic agents, which can affect multiple genes using different signaling pathways. miRNAs therapeutic modulation techniques have been used in the settings of atherosclerosis, acute myocardial infarction, restenosis, vascular remodeling, arrhythmias, hypertrophy and fibrosis, angiogenesis and cardiogenesis, aortic aneurysm, pulmonary hypertension, and ischemic injury. This review presents detailed information about miRNAs regarding structure and biogenesis, stages of synthesis and functions, expression profiles in serum/plasma of living organisms, diagnostic and prognostic potential as novel biomarkers, and therapeutic applications in various diseases.
Collapse
Affiliation(s)
| | - Mehmet Demir
- Department of Cardiology, University of Health Sciences, Bursa Yüksek İhtisas Research and Training Hospital, Bursa, Turkey
| |
Collapse
|
25
|
Kasprzyk-Pawelec A, Wojciechowska A, Kuc M, Zielinski J, Parulski A, Kusmierczyk M, Lutynska A, Kozar-Kaminska K. microRNA expression profile in Smooth Muscle Cells isolated from thoracic aortic aneurysm samples. Adv Med Sci 2019; 64:331-337. [PMID: 31022558 DOI: 10.1016/j.advms.2019.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/31/2019] [Accepted: 04/10/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE Thoracic aortic aneurysm (TAA) is a cardiovascular disease characterized by increased aortic diameter, treated with surgery and endovascular therapy in order to avoid aortic dissection or rupture. The mechanism of TAA formation has not been thoroughly studied and many factors have been proposed to drive its progression; however strong focus is attributed to modification of smooth muscle cells (SMCs). Latest research indicates, that microRNAs (miRNAs) may play a significant role in TAA development - these are multifunctional molecules consisting of 19-24 nucleotides involved in regulation of the gene expression level related to many biological processes, i.e. cardiovascular disease pathophysiology, immunity or inflammation. MATERIALS AND METHODS Primary SMCs were isolated from aortic scraps of TAA patients and age- and sex-matched healthy controls. Purity of isolated SMCs was determined by flow cytometry using specific markers: α-SMA, CALP, MHC and VIM. Real-time polymerase chain reaction (RT-PCR) was conducted for miRNA analysis. RESULTS We established an isolation protocol and investigated the miRNA expression level in SMCs isolated from aneurysmal and non-aneurysmal aortic samples. We identified that let-7 g (0.71-fold, p = 0.01), miR-130a (0.40-fold, p = 0.04), and miR-221 (0.49-fold, p = 0.05) significantly differed between TAA patients and healthy controls. CONCLUSIONS Further studies are required to improve our understanding of the pathophysiology underlying TAA, which may aid the development of novel, targeted therapies. The pivotal role of miRNAs in the cardiovascular system provides a new perspective on the pathophysiology of thoracic aortic aneurysms.
Collapse
Affiliation(s)
- Anna Kasprzyk-Pawelec
- Department of Medical Biology, Immunology Laboratory, Institute of Cardiology, Warsaw, Poland
| | - Anna Wojciechowska
- Department of Medical Biology, Immunology Laboratory, Institute of Cardiology, Warsaw, Poland
| | - Mateusz Kuc
- Department of Cardiac Surgery and Transplantology, Institute of Cardiology, Warsaw, Poland
| | - Jakub Zielinski
- Department of Cardiac Surgery and Transplantology, Institute of Cardiology, Warsaw, Poland
| | - Adam Parulski
- Department of Cardiac Surgery and Transplantology, Institute of Cardiology, Warsaw, Poland
| | - Mariusz Kusmierczyk
- Department of Cardiac Surgery and Transplantology, Institute of Cardiology, Warsaw, Poland
| | - Anna Lutynska
- Department of Medical Biology, Institute of Cardiology, Warsaw, Poland
| | | |
Collapse
|
26
|
Abdullah OA, El Gazzar WB, Salem TI, Elmohamady MN, Nasif SN, Eltaher SM. miR-15a: a potential diagnostic biomarker and a candidate for non-operative therapeutic modality for age-related cataract. Br J Biomed Sci 2019; 76:184-189. [PMID: 31264507 DOI: 10.1080/09674845.2019.1639337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: In order to better understand the role of hsa-miR-15a in the pathogenesis of age-related cataracts, we hypothesised altered expression, and of target anti-apoptotic genes, BCL-2 and MCL-1, in lens epithelial cells amongst age-related cataract patients.Material and methods: Reverse transcription quantitative polymerase chain reaction (RT-qPCR) quantified the expression of hsa-miR-15a and the target genes BCL-2 and MCL-1 in lens epithelial cells of 120 age-related cataract patients (40 patients with cortical cataracts, 40 patients with nuclear cataracts and 40 patients with posterior subcapsular cataracts) and 40 controls. Sixty specimens (15 normal and 45 cataracts) were stained immunohistochemically with BCL-2 and MCL-1 markers.Results: The expression of hsa-miR-15a was significantly increased (p = 0.003) in lens epithelial cells of cataract patients compared to the control group. BCL-2 and MCL-1 expression levels were significantly decreased in cataract patients (p < 0.001). A significant increase in hsa-miR-15a expression in the cortical subtype compared to the posterior subcapsular subtype (p = 0.003) and a significant decrease in BCL-2 and MCL-1 expressions in the cortical subtype compared to the nuclear and the posterior subcapsular subtype was detected.Conclusions: The increased expression of hsa-miR-15a in lens epithelial cells of cataract patients may repress the expression of BCL-2 and MCL-1. The expression of hsa-miR-15a and the subsequent apoptosis of lens epithelial cells are part of the pathogenesis of age-related cataracts.
Collapse
Affiliation(s)
- O A Abdullah
- Departments of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - W B El Gazzar
- Departments of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - T I Salem
- Ophthalmology, Faculty of Medicine, Benha University, Benha, Egypt
| | - M N Elmohamady
- Ophthalmology, Faculty of Medicine, Benha University, Benha, Egypt
| | - S N Nasif
- Pathology, Faculty of Medicine, Benha University, Benha, Egypt
| | - S M Eltaher
- Community Medicine and Public Health, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
27
|
Leeper NJ, Maegdefessel L. Non-coding RNAs: key regulators of smooth muscle cell fate in vascular disease. Cardiovasc Res 2019; 114:611-621. [PMID: 29300828 DOI: 10.1093/cvr/cvx249] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/28/2017] [Indexed: 01/02/2023] Open
Abstract
The vascular smooth muscle cell (SMC) is one of the most plastic cells in the body. Understanding how non-coding RNAs (ncRNAs) regulate SMC cell-fate decision making in the vasculature has significantly enhanced our understanding of disease development, and opened up exciting new avenues for potential therapeutic applications. Recent studies on SMC physiology have in addition challenged our traditional view on their role and contribution to vascular disease, mainly in the setting of atherosclerosis as well as aneurysm disease, and restenosis after angioplasties. The impact of SMC behaviour on vascular disease is now recognized to be context dependent; SMC proliferation and migration can be harmful or beneficial, whereas their apoptosis, senescence, and switching into a more macrophage-like phenotype can promote inflammation and disease progression. This is in particular true for atherosclerosis-related diseases, where proliferation of SMCs was believed to promote lesion formation, but may also prevent plaque rupture by stabilizing the fibrous cap. Based on newer findings of genetic lineage tracing studies, it was revealed that SMC phenotypic switching can result in less-differentiated forms that lack classical SMC markers while exhibiting functions more related to macrophage-like cells. This switching can directly promote atherogenesis. The aim of this current review is to summarize and discuss how ncRNAs (mainly microRNAs and long ncRNAs) are involved in SMC plasticity, and how they directly affect vascular disease development and progression. Finally, we want to critically assess where potential future therapies could be useful to influence the burden of vascular diseases.
Collapse
Affiliation(s)
- Nicholas J Leeper
- Division of Vascular Surgery, Stanford University, Stanford, CA, USA
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, and German Center for Cardiovascular Research Center (DZHK) Partner Site Munich, 81675 Munich, Germany.,Karolinska Institute, Center for Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
28
|
Fu L, Jiang G, Weng H, Dick GM, Chang Y, Kassab GS. Cerebrovascular miRNAs correlate with the clearance of Aβ through perivascular route in younger 3xTg-AD mice. Brain Pathol 2019; 30:92-105. [PMID: 31206909 DOI: 10.1111/bpa.12759] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
The "two-hit vascular hypothesis for Alzheimer's disease (AD)" and amyloid-β (Aβ) oligomer hypothesis suggest that impaired soluble Aβ oligomers clearance through the cerebral vasculature may be an initial step of the AD process. Soluble Aβ oligomers are driven into perivascular spaces from the brain parenchyma and toward peripheral blood flow. The underlying vascular-based mechanism, however, has not been defined. Given that microRNAs (miRNAs), emerging as novel modulators, are involved in numerous physiological and pathological processes, we hypothesized that cerebrovascular miRNAs may regulate the activities of brain blood vessels, which further affects the concentration of Aβ in the AD brain. In this study, perivascular Aβ deposits, higher vascular activation, increased pericyte coverage and up-regulated capillaries miRNAs at 6 months old (6 mo) were found to correlate with the lower Aβ levels of middle AD stage (9 mo) in 3xTg-AD (3xTg) mice. It is implicated that at the early stage of AD when intracellular Aβ appeared, higher expression of vessel-specific miRNAs, elevated pericyte coverage, and activated endothelium facilitate Aβ oligomer clearance through the perivascular route, resulting in a transient reduction of Aβ oligomers at 9 mo. Additionally, ghrelin-induced upregulation of capillary miRNAs and increased pericyte coverage attenuated Aβ burden at 9 mo, in further support of the relationship between vascular miRNAs and Aβ clearance. This work suggests a cerebral microvessel miRNA may boost endothelial highly activated phenotypes to promote elimination of Aβ oligomers through the perivascular drainage pathway and contribute to AD progression. The targeting of brain vessel-specific miRNAs may provide a new rationale for the development of innovative therapeutic strategies for AD treatment.
Collapse
Affiliation(s)
- Lijuan Fu
- California Medical Innovations Institute, San Diego, USA
| | - Ge Jiang
- California Medical Innovations Institute, San Diego, USA
| | - Hope Weng
- California Medical Innovations Institute, San Diego, USA
| | - Gregory M Dick
- California Medical Innovations Institute, San Diego, USA
| | - Yanzhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | | |
Collapse
|
29
|
Abstract
Calcific aortic valve disease (CAVD) is the most common heart valve disorder in human populations. Nevertheless, there are presently no effective means for its prevention and treatment. It is therefore critical to comprehensively define key mechanisms of the disease. A major focus of cardiovascular research has been characterization of how regulation of gene expression maintains healthy physiologic status of the component tissues of the system and how derangements of gene regulation may become pathological. Recently, substantial evidence has emerged that noncoding RNAs, which are an enormous and versatile class of regulatory elements, such as microRNAs and long noncoding RNAs, have roles in onset and prognosis of CAVD. Authors of the present report have therefore here provided a summary of the current understanding of contributions made by noncoding RNAs major features of CAVD. It is anticipated that this article will serve as a valuable guide to research strategy in this field and may additionally provide both researchers and clinicians with an expanded range of CAVD-associated biomarkers.
Collapse
|
30
|
Cardioprotective microRNAs: Lessons from stem cell-derived exosomal microRNAs to treat cardiovascular disease. Atherosclerosis 2019; 285:1-9. [PMID: 30939341 DOI: 10.1016/j.atherosclerosis.2019.03.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/28/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
The stem cell-based therapy has emerged as a promising therapeutic strategy for treating cardiovascular ischemic diseases (CVIDs), such as myocardial infarction (MI). However, some important functional shortcomings of stem cell transplantation, such as immune rejection, tumorigenicity and infusional toxicity, have overshadowed stem cell therapy in the setting of cardiovascular diseases (CVDs). Accumulating evidence suggests that the therapeutic effects of transplanted stem cells are predominately mediated by secreting paracrine factors, importantly, microRNAs (miRs) present in the secreted exosomes. Therefore, novel cell-free therapy based on the stem cell-secreted exosomal miRs can be considered as a safe and effective alternative tool to stem cell therapy for the treatment of CVDs. Stem cell-derived miRs have recently been found to transfer, via exosomes, from a transplanted stem cell into a recipient cardiac cell, where they regulate various cellular process, such as proliferation, apoptosis, stress responses, as well as differentiation and angiogenesis. The present review aimed to summarize cardioprotective exosomal miRs secreted by transplanted stem cells from various sources, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and cardiac stem/progenitor cells, which showed beneficial modulatory effects on the myocardial infracted heart. In summary, stem cell-exosomal miRs, including miR-19a, mirR-21, miR-21-5p, miR-21-a5p, miR-22 miR-24, miR-26a, miR-29, miR-125b-5p, miR-126, miR-201, miR-210, and miR-294, have been shown to have cardioprotective effects by enhancing cardiomyocyte survival and function and attenuating cardiac fibrosis. Additionally, MCS-exosomal miRs, including miR-126, miR-210, miR-21, miR-23a-3p and miR-130a-3p, are found to exert cardioprotective effects through induction of angiogenesis in ischemic heart after MI.
Collapse
|
31
|
Wang Z, Zhang J, Zhang S, Yan S, Wang Z, Wang C, Zhang X. MiR‑30e and miR‑92a are related to atherosclerosis by targeting ABCA1. Mol Med Rep 2019; 19:3298-3304. [PMID: 30816508 DOI: 10.3892/mmr.2019.9983] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic disease characterized by the accumulation of lipids and fibrous elements in the large arteries, which is the principal cause of coronary artery disease. Dysregulated exosomal microRNA (miRNA) levels in serum have been identified in patients with various diseases, including CAD. In the present study, nine candidate miRNAs were detected in the plasma exosome from 42 patients with coronary atherosclerosis, and a higher expression of miR‑30e and miR‑92a was identified in patients. Following bioinformatics analysis and confirmation through immunoblotting, it was demonstrated that ATP binding cassette (ABC)A1 is a direct target of miR‑30e, and miR‑92a. Furthermore, a negative correlation was identified between plasma miR‑30e and ABCA1, or miR‑30e and cholesterol. Thus, the results of the present study suggest that the miR‑30e level in exosomes from serum may have the potential to be a novel diagnostic biomarker for coronary atherosclerosis.
Collapse
Affiliation(s)
- Zhisheng Wang
- Medical Department of Shandong Medical College, Shandong 250002, P.R. China
| | - Jiayun Zhang
- Department of Cardiology, Staff Hospital of Hebei GEO University, Hebei 050031, P.R. China
| | - Songlan Zhang
- Second Department of Internal Medicine, People's Hospital of Shizhong District, Shandong 250002, P.R. China
| | - Shifang Yan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| | - Zihui Wang
- Department of Cardiology, People's Hospital of Mancheng District, Hebei 071000, P.R. China
| | - Chao Wang
- Department of Cardiology, People's Hospital of Zouping County, Shandong 256200, P.R. China
| | - Xiaojiang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, P.R. China
| |
Collapse
|
32
|
Abstract
High-throughput profiling/sensing of nucleic acids has recently emerged as a highly promising strategy for the early diagnosis and improved prognosis of a broad range of pathologies, most notably cancer. Among the potential biomarker candidates, microRNAs (miRNAs), a class of non-coding RNAs of 19-25 nucleotides in length, are of particular interest due to their role in the post-transcriptional regulation of gene expression. Developing miRNA sensing technologies that are quantitative, ultrasensitive and highly specific has proven very challenging because of their small size, low natural abundance and the high degree of sequence similarity among family members. When compared to optical based methods, electrochemical sensors offer many advantages in terms of sensitivity and scalability. This non-comprehensive review aims to break-down and highlight some of the most promising strategies for electrochemical sensing of microRNA biomarkers.
Collapse
Affiliation(s)
- Philip Gillespie
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW72AZ, UK.
| | - Sylvain Ladame
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW72AZ, UK.
| | - Danny O'Hare
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW72AZ, UK.
| |
Collapse
|
33
|
MicroRNAs and immunity in periodontal health and disease. Int J Oral Sci 2018; 10:24. [PMID: 30078842 PMCID: PMC6080405 DOI: 10.1038/s41368-018-0025-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are critical regulators of the host immune and inflammatory response against bacterial pathogens. In the present review, we discuss target genes, target gene functions, the potential regulatory role of miRNAs in periodontal tissues, and the potential role of miRNAs as biomarkers and therapeutics. In periodontal disease, miRNAs exert control over all aspects of innate and adaptive immunity, including the functions of neutrophils, macrophages, dendritic cells and T and B cells. Previous human studies have highlighted some key miRNAs that are dysregulated in periodontitis patients. In the present study, we mapped the major miRNAs that were altered in our reproducible periodontitis mouse model relative to control animals. The miRNAs that were upregulated as a result of periodontal disease in both human and mouse studies included miR-15a, miR-29b, miR-125a, miR-146a, miR-148/148a and miR-223, whereas miR-92 was downregulated. The association of individual miRNAs with unique aspects of periodontal disease and their stability in gingival crevicular fluid underscores their potential as markers for periodontal disease progression or healthy restitution. Moreover, miRNA therapeutics hold great promise for the future of periodontal therapy because of their ability to modulate the immune response to infection when applied in conjunction with synthetic antagomirs and/or relatively straightforward delivery strategies.
Collapse
|
34
|
Bayoumi AS, Teoh JP, Aonuma T, Yuan Z, Ruan X, Tang Y, Su H, Weintraub NL, Kim IM. MicroRNA-532 protects the heart in acute myocardial infarction, and represses prss23, a positive regulator of endothelial-to-mesenchymal transition. Cardiovasc Res 2018; 113:1603-1614. [PMID: 29016706 DOI: 10.1093/cvr/cvx132] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/07/2017] [Indexed: 01/13/2023] Open
Abstract
Aims Acute myocardial infarction (MI) leads to cardiac remodelling and development of heart failure. Insufficient myocardial capillary density after MI is considered a critical determinant of this process. MicroRNAs (miRs), negative regulators of gene expression, have emerged as important players in MI. We previously showed that miR-532-5p (miR-532) is up-regulated by the β-arrestin-biased β-adrenergic receptor antagonist (β-blocker) carvedilol, which activates protective pathways in the heart independent of G protein-mediated second messenger signalling. Here, we hypothesize that β2-adrenergic receptor/β-arrestin-responsive miR-532 confers cardioprotection against MI. Methods and results Using cultured cardiac endothelial cell (CEC) and in vivo approaches, we show that CECs lacking miR-532 exhibit increased transition to a fibroblast-like phenotype via endothelial-to-mesenchymal transition (EndMT), while CECs over-expressing miR-532 display decreased EndMT. We also demonstrate that knockdown of miR-532 in mice causes abnormalities in cardiac structure and function as well as reduces CEC proliferation and cardiac vascularization after MI. Mechanistically, cardioprotection elicited by miR-532 is in part attributed to direct repression of a positive regulator of maladaptive EndMT, prss23 (a protease serine 23) in CECs. Conclusions In conclusion, these findings reveal a pivotal role for miR-532-prss23 axis in regulating CEC function after MI, and this novel axis could be suitable for therapeutic intervention in ischemic heart disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huabo Su
- Vascular Biology Center.,Department of Pharmacology and Toxicology
| | | | - Il-Man Kim
- Vascular Biology Center.,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, CB-3717, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
35
|
Spin JM, Li DY, Maegdefessel L, Tsao PS. Non-coding RNAs in aneurysmal aortopathy. Vascul Pharmacol 2018; 114:110-121. [PMID: 29909014 DOI: 10.1016/j.vph.2018.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/21/2018] [Accepted: 06/09/2018] [Indexed: 02/07/2023]
Abstract
Aortic aneurysms represent a major public health burden, and currently have no medical treatment options. The pathophysiology behind these aneurysms is complex and variable, depending on location and underlying cause, and generally involves progressive dysfunction of all elements of the aortic wall. Changes in smooth muscle behavior, endothelial signaling, extracellular matrix remodeling, and to a variable extent inflammatory signaling and cells, all contribute to the dilation of the aorta, ultimately resulting in high mortality and morbidity events including dissection and rupture. A large number of researchers have identified non-coding RNAs as crucial regulators of aortic aneurysm development, both in humans and in animal models. While most work to-date has focused on microRNAs, intriguing information has also begun to emerge regarding the role of long-non-coding RNAs. This review summarizes the currently available data regarding the involvement of non-coding RNAs in aneurysmal aortopathies. Going forward, these represent key potential therapeutic targets that might be leveraged in the future to slow or prevent aortic aneurysm formation, progression and rupture.
Collapse
Affiliation(s)
- Joshua M Spin
- Cardiovascular Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, USA
| | - Daniel Y Li
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Lars Maegdefessel
- Vascular Biology Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technical University of Munich, Munich, Germany; Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Philip S Tsao
- Cardiovascular Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, USA.
| |
Collapse
|
36
|
Liu G, Niu X, Meng X, Zhang Z. Sensitive miRNA markers for the detection and management of NSTEMI acute myocardial infarction patients. J Thorac Dis 2018; 10:3206-3215. [PMID: 30069316 DOI: 10.21037/jtd.2018.05.141] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background NSTEMI patients will benefit greatly with better biomarker screening to detect and prognose the disease. Using miRNAs, we evaluated the clinical utility in acute myocardial infarction (AMI) patients during disease onset and therapy. Methods A total of 145 NSTEMI patients and 30 healthy volunteers with no history of cardiovascular disease (CVD) were recruited. miRNA levels in plasma were measured during disease manifestation and serially during treatment phase. Levels of multiple candidates (miR-1, miR-133, miR-208, miR-499) were analysed. The miRNA levels were directly compared between NSTEMI and healthy volunteers. Results Cardiac related miRNAs levels demonstrated significant increase compared with healthy controls. miR-499 exhibited the highest elevation with more than 6.03-fold change compared with healthy participants. Conventional cTnT measurements were in good agreement to miRNA relative expressions. In serial measurements, miR-499 demonstrated large fluctuations and could be linked to the secondary complications. In contrast, miR-133 showed insignificant variations in mean levels during serial sampling. Conclusions miRNA is a potentially sensitive biomarker for NSTEMI AMI patients for disease detection and treatment monitoring. The sensitivities were comparable to cTnT for diagnostic accuracy and patients with sustained or higher levels were correlated to secondary complications.
Collapse
Affiliation(s)
- Guoyong Liu
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China.,The Cardiovascular Department, Qinghai Provincial People's Hospital, Xining 810007, China
| | - Xiaowei Niu
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaoxue Meng
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China
| | - Zheng Zhang
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
37
|
Inhibition of miR-155 attenuates abdominal aortic aneurysm in mice by regulating macrophage-mediated inflammation. Biosci Rep 2018; 38:BSR20171432. [PMID: 29459426 PMCID: PMC5938419 DOI: 10.1042/bsr20171432] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 02/07/2018] [Accepted: 02/19/2018] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to identify abdominal aortic aneurysms (AAA)-associated miR-155 contributing to AAA pathology by regulating macrophage-mediated inflammation. Angiotensin II (AngII)-infused apolipoprotein E-deficient (ApoE-/-) mice and THP-1 cells model of miR-155 overexpression and deficiency were used in the experiments. The expression of miR-155 was detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Cytokines were evaluated using enzyme-linked immunoabsorbent assay (ELISA). Western blotting was used to measure the levels of MMP-2, MMP-9, iNOS, and monocyte chemoattractant protein (MCP)-1 proteins. Immunostaining and transwell were used to determine CD68, elastic collagen, proliferation, and migration of vascular smooth muscle cells (VSMCs). The results showed that miR-155 and cytokines were up-regulated in AAA patients or ApoE-/- mice. Overexpression of miR-155 enhanced MMP-2, MMP-9, iNOS, and MCP-1 levels, and stimulated the proliferation and migration of VSMCs. Meanwhile, inhibition of miR-155 had the opposite effect. In addition, histology demonstrated accumulation of CD68 and elastic collagen-positive areas significantly decreased in miR-155 antagomir injection group. In conclusion, the results of the present study suggest that inhibiting miR-155 is crucial to prevent the development of AAA by regulating macrophage inflammation.
Collapse
|
38
|
Roberts JT, Patterson DG, King VM, Amin SV, Polska CJ, Houserova D, Crucello A, Barnhill EC, Miller MM, Sherman TD, Borchert GM. ADAR Mediated RNA Editing Modulates MicroRNA Targeting in Human Breast Cancer. Processes (Basel) 2018; 6. [PMID: 30197877 PMCID: PMC6128407 DOI: 10.3390/pr6050042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RNA editing by RNA specific adenosine deaminase acting on RNA (ADAR) is increasingly being found to alter microRNA (miRNA) regulation. Editing of miRNA transcripts can affect their processing, as well as which messenger RNAs (mRNAs) they target. Further, editing of target mRNAs can also affect their complementarity to miRNAs. Notably, ADAR editing is often increased in malignancy with the effect of these RNA changes being largely unclear. In addition, numerous reports have now identified an array of miRNAs that directly contribute to various malignancies although the majority of their targets remain largely undefined. Here we propose that modulating the targets of miRNAs via mRNA editing is a frequent occurrence in cancer and an underappreciated participant in pathology. In order to more accurately characterize the relationship between these two regulatory processes, this study examined RNA editing events within mRNA sequences of two breast cancer cell lines (MCF-7 and MDA-MB-231) and determined whether or not these edits could modulate miRNA associations. Computational analyses of RNA-Seq data from these two cell lines identified over 50,000 recurrent editing sites within human mRNAs, and many of these were located in 3’ untranslated regions (UTRs). When these locations were screened against the list of currently-annotated miRNAs we discovered that editing caused a subset (~9%) to have significant alterations to mRNA complementarity. One miRNA in particular, miR-140–3p, is known to be misexpressed in many breast cancers, and we found that mRNA editing allowed this miRNA to directly target the apoptosis inducing gene DFFA in MCF-7, but not in MDA-MB-231 cells. As these two cell lines are known to have distinct characteristics in terms of morphology, invasiveness and physiological responses, we hypothesized that the differential RNA editing of DFFA in these two cell lines could contribute to their phenotypic differences. Indeed, we confirmed through western blotting that inhibiting miR-140–3p increases expression of the DFFA protein product in MCF-7, but not MDA-MB-231, and further that inhibition of miR-140–3p also increases cellular growth in MCF-7, but not MDA-MB-231. Broadly, these results suggest that the creation of miRNA targets may be an underappreciated function of ADAR and may help further elucidate the role of RNA editing in tumor pathogenicity.
Collapse
Affiliation(s)
- Justin T Roberts
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Dillon G Patterson
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Valeria M King
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Shivam V Amin
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Caroline J Polska
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Dominika Houserova
- Department of Pharmacology, USA College of Medicine, Mobile, AL 36688-0002, USA;
| | - Aline Crucello
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Emmaline C Barnhill
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Molly M Miller
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Timothy D Sherman
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
| | - Glen M Borchert
- Department of Biology, University of South Alabama, Mobile, AL 36688-0002, USA
- Department of Pharmacology, USA College of Medicine, Mobile, AL 36688-0002, USA;
| |
Collapse
|
39
|
Jin H, Li DY, Chernogubova E, Sun C, Busch A, Eken SM, Saliba-Gustafsson P, Winter H, Winski G, Raaz U, Schellinger IN, Simon N, Hegenloh R, Matic LP, Jagodic M, Ehrenborg E, Pelisek J, Eckstein HH, Hedin U, Backlund A, Maegdefessel L. Local Delivery of miR-21 Stabilizes Fibrous Caps in Vulnerable Atherosclerotic Lesions. Mol Ther 2018; 26:1040-1055. [PMID: 29503197 PMCID: PMC6080193 DOI: 10.1016/j.ymthe.2018.01.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 01/22/2023] Open
Abstract
miRNAs are potential regulators of carotid artery stenosis and concordant vulnerable atherosclerotic plaques. Hence, we analyzed miRNA expression in laser captured micro-dissected fibrous caps of either ruptured or stable plaques (n = 10 each), discovering that miR-21 was significantly downregulated in unstable lesions. To functionally evaluate miR-21 in plaque vulnerability, miR-21 and miR-21/apolipoprotein-E double-deficient mice (Apoe-/-miR-21-/-) were assessed. miR-21-/- mice lacked sufficient smooth muscle cell proliferation in response to carotid ligation injury. When exposing Apoe-/-miR-21-/- mice to an inducible plaque rupture model, they presented with more atherothrombotic events (93%) compared with miR-21+/+Apoe-/- mice (57%). We discovered that smooth muscle cell fate in experimentally induced advanced lesions is steered via a REST-miR-21-REST feedback signaling pathway. Furthermore, Apoe-/-miR-21-/- mice presented with more pronounced atherosclerotic lesions, greater foam cell formation, and substantially higher levels of arterial macrophage infiltration. Local delivery of a miR-21 mimic using ultrasound-targeted microbubbles into carotid plaques rescued the vulnerable plaque rupture phenotype. In the present study, we identify miR-21 as a key modulator of pathologic processes in advanced atherosclerosis. Targeted, lesion site-specific overexpression of miR-21 can stabilize vulnerable plaques.
Collapse
Affiliation(s)
- Hong Jin
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Daniel Y Li
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | | | - Changyan Sun
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Albert Busch
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Suzanne M Eken
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Hanna Winter
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Greg Winski
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Uwe Raaz
- University Heart Center, Göttingen, Germany
| | | | - Nancy Simon
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Renate Hegenloh
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Ljubica Perisic Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ewa Ehrenborg
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Hans-Henning Eckstein
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | | | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden; Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany.
| |
Collapse
|
40
|
Thanikachalam PV, Ramamurthy S, Wong ZW, Koo BJ, Wong JY, Abdullah MF, Chin YH, Chia CH, Tan JY, Neo WT, Tan BS, Khan WF, Kesharwani P. Current attempts to implement microRNA-based diagnostics and therapy in cardiovascular and metabolic disease: a promising future. Drug Discov Today 2018; 23:460-480. [DOI: 10.1016/j.drudis.2017.10.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/09/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
|
41
|
Liu K, Xuekelati S, Zhou K, Yan Z, Yang X, Inayat A, Wu J, Guo X. Expression Profiles of Six Atherosclerosis-Associated microRNAs That Cluster in Patients with Hyperhomocysteinemia: A Clinical Study. DNA Cell Biol 2018; 37:189-198. [PMID: 29461880 DOI: 10.1089/dna.2017.3845] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of this study is to discuss the hypothesis that expression of plasma atherosclerosis-associated microRNAs (miRNAs) in hyperhomocysteinemia (Hhcy) patients could predict the presence of atherosclerosis from different channels. Six plasma miRNAs (miR-145, miR-155, miR-222, miR-133, miR-217, and miR-30) selected for our study have been confirmed as critical gene regulators involved in atherosclerosis and can be steadily determined in plasma. Expression of the above six plasma circulating miRNAs revealed significant upregulation of two miRNAs (miR-133 and miR-217) and downregulation of three miRNAs (miR-145, miR-155, and miR-222). Six candidate miRNAs showed a significant correlation with homocysteine (Hcy) or lipid parameters. The results of this study indicated that miR-217 was further significantly upregulated in Hhcy + ATH groups than in normal control, Hhcy-, and atherosclerosis-alone (ATH) groups and it showed a significant negative correlation with Hcy and triglycerides. More specifically, miR-217 showed the most specific expression patterns in all patients with atherosclerosis (ATH and Hhcy + ATH groups), which may have been a diagnostic value for Hhcy complicated with atherosclerosis, and predicted the progress of atherosclerosis in Hhcy patients effectively.
Collapse
Affiliation(s)
- Kejian Liu
- 1 Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China .,2 Department of Cardiology, The First Affiliated Hospital, Shihezi University School of Medicine , Shihezi, China
| | - Saiyare Xuekelati
- 3 The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University , Shihezi, China
| | - Kang Zhou
- 2 Department of Cardiology, The First Affiliated Hospital, Shihezi University School of Medicine , Shihezi, China
| | - Zhitao Yan
- 2 Department of Cardiology, The First Affiliated Hospital, Shihezi University School of Medicine , Shihezi, China
| | - Xu Yang
- 2 Department of Cardiology, The First Affiliated Hospital, Shihezi University School of Medicine , Shihezi, China
| | - Azeem Inayat
- 3 The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University , Shihezi, China
| | - Jiangdong Wu
- 3 The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University , Shihezi, China
| | - Xiaomei Guo
- 1 Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| |
Collapse
|
42
|
Chen R, Li H, Cai J, Wang C, Lin Z, Liu C, Niu Y, Zhao Z, Li W, Kan H. Fine Particulate Air Pollution and the Expression of microRNAs and Circulating Cytokines Relevant to Inflammation, Coagulation, and Vasoconstriction. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:017007. [PMID: 29342453 PMCID: PMC6014692 DOI: 10.1289/ehp1447] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 05/18/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) are a key factor in epigenetic regulation of gene expression, but miRNA responses to fine particulate matter (PM2.5) air pollution and their potential contribution to cardiovascular effects of PM2.5 are unknown. OBJECTIVE We explored the potential influence of PM2.5 on the expression of selected cytokines relevant to systemic inflammation, coagulation, and vasoconstriction, and on miRNAs that may regulate their expression. METHODS We designed a double-blind, randomized crossover study in which true and sham air purifiers were used to expose 55 healthy young adult students in Shanghai, China, to reduced or ambient levels of indoor PM2.5 during two-week periods, and we measured the expression (mRNA and protein) of 10 serum cytokines, and miRNAs that target them, after each intervention period. We used linear mixed-effect models to estimate associations of the intervention, and time-weighted personal PM2.5 exposures, with the cytokines, mRNA, and miRNAs; we also explored potential mediation by miRNAs. RESULTS The findings were generally consistent for associations with the intervention and for associations with an interquartile range increase in time-weighted PM2.5. Specifically, higher PM2.5 exposure was positively associated with the expression (mRNA, protein, or both) of interleukin-1 (encoded by IL1), IL6, tumor necrosis factor (encoded by TNF), toll-like receptor 2 (encoded by TLR2), coagulation factor 3 (encoded by F3), and endothelin 1 (encoded by EDN1), and was negatively associated with miRNAs (miR-21-5p, miR-187-3p, miR-146a-5p, miR-1-3p, and miR-199a-5p) predicted to target mRNAs of IL1, TNF, TLR2, and EDN1. CONCLUSIONS Our findings require confirmation but suggest that effects of PM2.5 on cardiovascular diseases may be related to acute effects on cytokine expression, which may be partly mediated through effects of PM2.5 on miRNAs that regulate cytokine expression. https://doi.org/10.1289/EHP1447.
Collapse
Affiliation(s)
- Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
- Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Huichu Li
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
| | - Jing Cai
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
- Shanghai Key Laboratory of Meteorology and Health, Shanghai, China
| | - Cuicui Wang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
| | - Zhijing Lin
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
| | - Cong Liu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
| | - Yue Niu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
| | - Zhuohui Zhao
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
| | - Weihua Li
- Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, Fudan University , Shanghai, China
- School of Public Health, Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University , Shanghai, China
- Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Jeong D, Yoo J, Lee P, Kepreotis SV, Lee A, Wahlquist C, Brown BD, Kho C, Mercola M, Hajjar RJ. miR-25 Tough Decoy Enhances Cardiac Function in Heart Failure. Mol Ther 2017; 26:718-729. [PMID: 29273502 DOI: 10.1016/j.ymthe.2017.11.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/17/2017] [Accepted: 11/22/2017] [Indexed: 01/14/2023] Open
Abstract
MicroRNAs are promising therapeutic targets, because their inhibition has the potential to normalize gene expression in diseased states. Recently, our group found that miR-25 is a key SERCA2a regulating microRNA, and we showed that multiple injections of antagomirs against miR-25 enhance cardiac contractility and function through SERCA2a restoration in a murine heart failure model. However, for clinical application, a more stable suppressor of miR-25 would be desirable. Tough Decoy (TuD) inhibitors are emerging as a highly effective method for microRNA inhibition due to their resistance to endonucleolytic degradation, high miRNA binding affinity, and efficient delivery. We generated a miR-25 TuD inhibitor and subcloned it into a cardiotropic AAV9 vector to evaluate its efficacy. The AAV9 TuD showed selective inhibition of miR-25 in vitro cardiomyoblast culture. In vivo, AAV9-miR-25 TuD delivered to the murine pressure-overload heart failure model selectively decreased expression of miR-25, increased levels of SERCA2a protein, and ameliorated cardiac dysfunction and fibrosis. Our data indicate that miR-25 TuD is an effective long-term suppressor of miR-25 and a promising therapeutic candidate to treat heart failure.
Collapse
Affiliation(s)
- Dongtak Jeong
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jimeen Yoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha V Kepreotis
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ahyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christine Wahlquist
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Brian D Brown
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Changwon Kho
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark Mercola
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
44
|
Punga AR, Punga T. Circulating microRNAs as potential biomarkers in myasthenia gravis patients. Ann N Y Acad Sci 2017; 1412:33-40. [PMID: 29125182 DOI: 10.1111/nyas.13510] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/05/2017] [Accepted: 09/09/2017] [Indexed: 12/16/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNA molecules that bind to specific mRNA targets and regulate a wide range of important biological processes within cells. Circulating miRNAs are released into the extracellular space and can be measured in most biofluids, including blood serum and plasma. Recently, circulating miRNAs have emerged as easily accessible markers in various body fluids with different profiles and quantities specific for different human disorders, including autoimmune diseases. In myasthenia gravis (MG), diagnostic tests such as titers of serum autoantibodies specific for either the acetylcholine receptor (AChR+ ) or muscle-specific tyrosine kinase (MuSK+ ) do not necessarily reflect disease progression, and there is a great need for reliable objective biomarkers to monitor the disease course and therapeutic response. Recent studies in AChR+ MG revealed elevated levels of the immuno-miRNAs miR-150-5p and miR-21-5p. Of particular importance, levels of miR-150-5p were lower in immunosuppressed patients and in patients with clinical improvement following thymectomy. In MuSK+ MG, another profile of circulating miRNAs was found, including upregulation of the let-7 family of miRNAs. Here, we summarize the potential role of circulating miRNAs as biomarkers in general and in MG, and highlight important considerations for the analysis of circulating miRNA.
Collapse
Affiliation(s)
- Anna Rostedt Punga
- Department of Neuroscience, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Tanel Punga
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
45
|
MicroRNA-1906, a Novel Regulator of Toll-Like Receptor 4, Ameliorates Ischemic Injury after Experimental Stroke in Mice. J Neurosci 2017; 37:10498-10515. [PMID: 28924010 DOI: 10.1523/jneurosci.1139-17.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 02/03/2023] Open
Abstract
Toll-like receptor 4 (TLR4) is a proinflammatory cascade initiator in poststroke inflammation. In this study, miR-1906, a novel regulator of TLR4, was identified via in silico analysis and microRNA profiling in male adult mice and its expression was then quantitated in the ischemic hemisphere. We found miR-1906 to be significantly brain enriched in the ischemic hemisphere and even more drastically enriched in the peri-infarct regions. Furthermore, in vitro experiments demonstrated that, during oxygen-glucose deprivation, miR-1906 expression was increased in glial cells but decreased in neurons. Surprisingly, despite the augmentation of intracellular abundance, miR-1906 expression in extracellular vesicles was decreased in astrocyte cell culture supernatants, suggesting reduced sources of miR-1906 from glia to neurons. When exogenous miR-1906 was administered, decreased TLR4 protein expression was observed both in vitro and in vivo Using Cy3 labeling, exogenous miR-1906 uptake by astrocytes, microglia, and neurons was visualized directly in vivo Reduced infarct volumes and improved functional outcomes were observed in middle cerebral artery occlusion mice receiving miR-1906. However, the protective effects of miR-1906 disappeared with the genetic knock-out of TLR4, suggesting that TLR4 is a major target of miR-1906 through which the microRNA exerts its therapeutic effects.SIGNIFICANCE STATEMENT The current study identified miR-1906 as a novel specific regulator of Toll-like receptor 4 (TLR4) and depicted its distinct expression patterns in different cerebral regions and cell types during ischemic attack. Therefore, the therapeutic supplementation of miR-1906 can be beneficial in the modulation of poststroke inflammation. Using Cy3 labeling, exogenous miR-1906 expression was visualized and shown to enter astrocytes, microglia, and neurons successfully in vivo Supplemental therapeutic miR-1906 resulted in reduced TLR4 expression and improved outcomes after middle cerebral artery occlusion in a mouse model, but its neuroprotective function was TLR4 dependent, suggesting that TLR4 is a major target of miR-1906.
Collapse
|
46
|
Paloschi V, Maegdefessel L. Towards Point-of-Care Measurements Using Noncoding RNAs. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.117.001859. [DOI: 10.1161/circgenetics.117.001859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Valentina Paloschi
- From the Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (V.P., L.M.); and Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden (L.M.)
| | - Lars Maegdefessel
- From the Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (V.P., L.M.); and Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden (L.M.)
| |
Collapse
|
47
|
Current epigenetic aspects the clinical kidney researcher should embrace. Clin Sci (Lond) 2017; 131:1649-1667. [DOI: 10.1042/cs20160596] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD), affecting 10–12% of the world’s adult population, is associated with a considerably elevated risk of serious comorbidities, in particular, premature vascular disease and death. Although a wide spectrum of causative factors has been identified and/or suggested, there is still a large gap of knowledge regarding the underlying mechanisms and the complexity of the CKD phenotype. Epigenetic factors, which calibrate the genetic code, are emerging as important players in the CKD-associated pathophysiology. In this article, we review some of the current knowledge on epigenetic modifications and aspects on their role in the perturbed uraemic milieu, as well as the prospect of applying epigenotype-based diagnostics and preventive and therapeutic tools of clinical relevance to CKD patients. The practical realization of such a paradigm will require that researchers apply a holistic approach, including the full spectrum of the epigenetic landscape as well as the variability between and within tissues in the uraemic milieu.
Collapse
|
48
|
Liu K, Xuekelati S, Zhang Y, Yin Y, Li Y, Chai R, Li X, Peng Y, Wu J, Guo X. Expression levels of atherosclerosis-associated miR-143 and miR-145 in the plasma of patients with hyperhomocysteinaemia. BMC Cardiovasc Disord 2017. [PMID: 28633641 PMCID: PMC5477732 DOI: 10.1186/s12872-017-0596-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background An elevated level of homocysteine (Hcy) in the blood is designated hyperhomocysteinaemia (Hhcy) and is regarded as a strong risk factor for the development of atherosclerosis (ATH), although the association remains controversial. Considered to be essential gene expression regulators, micro-RNAs (miRNAs) modulate cardiovascular disease development and thus can be regarded as potential biomarkers and therapeutic targets in atherosclerosis. The aim of the current study is to investigate the expression levels of atherosclerosis-associated miR-143 and miR-145 in Hhcy patients and predict the progress of atherosclerosis in Hhcy patients. Methods A total of 100 participants were enrolled and included normal control subjects (NC = 20), hyperhomocysteinaemia alone subjects (Hhcy = 25), hyperhomocysteinaemia and carotid artery atherosclerosis combined subjects (Hhcy + ATH = 30) and patients with standalone carotid artery atherosclerosis (ATH = 25). Plasma Hcy, supplementary biochemical parameters and carotid artery ultrasonography (USG) were measured in all participants. MicroRNA expression levels in the peripheral blood were calculated by real-time reverse transcription-polymerase chain reaction (qRT-PCR). The correlations of miR-143 and miR-145 with Hcy, blood lipid parameters and carotid artery atherosclerotic plaques were evaluated using Pearson’s correlation coefficients. Receiver operating characteristic (ROC) curve analyses were performed to evaluate the capacities of miR-143 and miR-145 for the detection of Hhcy and atherosclerosis patients. Results MiR-143 and miR-145 exhibited trends towards significance with stepwise decreases from the NC to Hhcy groups and then to the Hhcy + ATH and ATH groups. Similar results were observed in the carotid artery plaque group (Hhcy + ATH and ATH grups) compared with the no-plaque group (NC and Hhcy groups). The miR-143 expression level exhibited significant negative correlations with Hcy, total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-c). The miR-145 expression level exhibited significant negative correlations with Hcy, TC, triglyceride (TG) and LDL-c. MiR-143 and miR-145 exhibited the greatest area under the curves (AUCs) (0.775 and 0.681, respectively) for the detection of every Hhcy patient, including those in the Hhcy and Hhcy + ATH groups, from among all subjects. Conclusion The results indicated that the levels of atherosclerosis-associated circulating miR-143 and miR-145 are linked to Hhcy. MiR-143 may be used as a potential non-invasive biomarkers of Hhcy and thus may be helpful in predicting the progress of atherosclerosis in Hhcy patients. Electronic supplementary material The online version of this article (doi:10.1186/s12872-017-0596-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kejian Liu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Saiyare Xuekelati
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Yue Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Yin Yin
- Department of Cardiology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Rui Chai
- Department of Cardiology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xinwei Li
- Department of Cardiology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yi Peng
- Department of Cardiology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jiangdong Wu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China.
| | - Xiaomei Guo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
49
|
Li Y, Maegdefessel L. Non-coding RNA Contribution to Thoracic and Abdominal Aortic Aneurysm Disease Development and Progression. Front Physiol 2017; 8:429. [PMID: 28670289 PMCID: PMC5472729 DOI: 10.3389/fphys.2017.00429] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022] Open
Abstract
Multiple research groups have started to uncover the complex genetic and epigenetic machinery necessary to maintain cardiovascular homeostasis. In particular, the key contribution of non-coding RNAs (ncRNAs) in regulating gene expression has recently received great attention. Aneurysms in varying locations of the aorta are defined as permanent dilations, predisposing to the fatal consequence of rupture. The characteristic pathology of an aneurysm is characterized by progressive vessel wall dilation, promoted by dying vascular smooth muscle cells and limited proliferation, as well as impaired synthesis and degradation of extracellular matrix components, which at least partially is the result of transmural inflammation and its disruptive effect on vessel wall homeostasis. Currently no conservative pharmacological approach exists that could slow down aneurysm progression and protect from the risk of acute rupture. In the recent past, several non-coding RNAs (mainly microRNAs) have been discovered as being involved in aneurysm progression throughout varying locations of the aorta. Exploring ncRNAs as key regulators and potential therapeutic targets by using antisense oligonucleotide strategies could open up promising opportunities for patients in the near future. Purpose of this current review is to summarize current findings and novel concepts of perspectivly utilizing ncRNAs for future therapeutic and biomarker applications.
Collapse
Affiliation(s)
- Yuhuang Li
- Vascular Biology Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technical University of MunichMunich, Germany
| | - Lars Maegdefessel
- Vascular Biology Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technical University of MunichMunich, Germany.,Department of Medicine, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
50
|
Iyer V, Rowbotham S, Biros E, Bingley J, Golledge J. A systematic review investigating the association of microRNAs with human abdominal aortic aneurysms. Atherosclerosis 2017; 261:78-89. [DOI: 10.1016/j.atherosclerosis.2017.03.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 12/24/2022]
|