1
|
Abdel-Wahab BA, El-Shoura EAM, Alqahtani SM, Saad HM, Bakir MB, Zaafar D. Cilostazol alleviates imatinib-induced myocardial injury in rats by modulating the TGF-β1/MAPK, SHC/Grb2/SOS signaling pathways and upregulating miRNA-195-5P. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03946-x. [PMID: 40100370 DOI: 10.1007/s00210-025-03946-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
Tyrosine kinase inhibitors have revolutionized cancer treatment, yet their association with cardiotoxicity remains a challenge. While the pathophysiological consequences of imatinib therapy involve diverse pathways, our understanding of these mechanisms is limited. Cilostazol, a selective phosphodiesterase 3 inhibitor used in intermittent claudication treatment, is known for its antioxidant and anti-inflammatory effects. In this study, we aimed to counteract the cardiotoxic effects of imatinib by administering cilostazol, concurrently investigating the precise mechanisms underlying imatinib-induced myocardial injury. Twenty-eight male Wistar rats were categorized into four groups: control (2 mL/kg normal saline, orally), cilostazol (10 mg/kg, orally), imatinib (40 mg/kg, intraperitonially), and combination (cilostazol and imatinib). Daily treatments were administered for 28 consecutive days. Imatinib therapy induced oxidative stress, pro-inflammatory responses, and cardiotoxicity biomarkers, alongside dysregulation of various protein and gene expressions. The addition of cilostazol to imatinib mitigated these deleterious effects, notably restoring measured biomarkers close to normal values. Histopathological investigations corroborated the biochemical findings. The co-administration of cilostazol with imatinib effectively protects against imatinib-induced myocardial injury in rats, reducing oxidative stress, apoptotic and inflammatory biomarkers, and modulating protein, gene, and miRNA-195-5p expression levels. While promising for alleviating and protecting against myocardial injury in imatinib-treated patients, these findings necessitate further confirmation through clinical studies.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, P.O. Box 1988, Najran, Saudi Arabia
| | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Saad Misfer Alqahtani
- Department of Pathology, College of Medicine, The University Hospital, Najran University, Najran, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt
| | - Marwa B Bakir
- Department of Medical Education, Faculty of Medicine, Najran University, P.O. 1988, Najran, Saudi Arabia
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Zhang F, Geng L, Zhang J, Han S, Guo M, Xu Y, Chen C. miR-486-5p diagnosed atrial fibrillation, predicted the risk of left atrial fibrosis, and regulated angiotensin II-induced cardiac fibrosis via modulating PI3K/Akt signaling through targeting FOXO1. Mol Cell Biochem 2025; 480:1077-1087. [PMID: 38782834 DOI: 10.1007/s11010-024-05027-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
This study focused on miR-486-5p in atrial fibrillation (AF) evaluating its clinical significance and revealing its regulatory mechanism in cardiac fibroblasts, aiming to explore a novel biomarker for AF. The study enrolled 131 AF patients and 77 non-AF individuals. With the help of polymerase chain reaction (PCR), the expression of miR-486-5p was evaluated. The significance of miR-486-5p in the diagnosis of AF and the occurrence of left atrial fibrosis (LAF) was assessed by receiver operating curve (ROC) and logistic analyses. The regulatory effect and mechanism of miR-486-5p on cardiac fibrosis were investigated in human cardiac fibroblasts treated with angiotensin II. miR-486-5p was significantly upregulated in AF patients and discriminated AF patients from non-AF individuals. Increasing miR-486-5p showed a significant association with decreasing left ventricular ejection fraction (LVEF), increasing left atrial diameter (LAD) and left ventricular end-diastolic diameter (LVEDd), and the high incidence of LAF in AF patients. Moreover, miR-486-5p was identified as a risk factor for LAF and could distinguish AF patients with LAF and without LAF. In cardiac fibroblasts, angiotensin II induced the upregulation of miR-486-5p and promoted cell proliferation, migration, and collagen synthesis. miR-486-5p negatively regulated forkhead box O1 (FOXO1) and its knockdown could reverse the promoted effect of angiotensin II. FOXO1 alleviated the effect of miR-486-5p, and the miR-486-5p/FOXO1 could activate PI3K/Akt signaling. The activation of PI3K/Akt signaling alleviated the enhanced proliferation, migration, and collagen synthesis of cardiac fibroblasts induced by angiotensin II, and its inhibition showed opposite effects. Increased miR-486-5p served as a biomarker for the diagnosis and development prediction of AF. miR-486-5p regulated cardiac fibroblast viability and collagen synthesis via modulating the PI3K/Akt signaling through targeting FOXO1.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, No. 212, Yuhua East Road, Baoding, 071000, People's Republic of China
| | - Lu Geng
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People's Republic of China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, No. 212, Yuhua East Road, Baoding, 071000, People's Republic of China
| | - Siliang Han
- Department of Cardiology, Affiliated Hospital of Hebei University, No. 212, Yuhua East Road, Baoding, 071000, People's Republic of China
| | - Mengya Guo
- Department of Cardiology, Affiliated Hospital of Hebei University, No. 212, Yuhua East Road, Baoding, 071000, People's Republic of China
| | - Yaxin Xu
- Department of Cardiology, Affiliated Hospital of Hebei University, No. 212, Yuhua East Road, Baoding, 071000, People's Republic of China
| | - Chunhong Chen
- Department of Cardiology, Affiliated Hospital of Hebei University, No. 212, Yuhua East Road, Baoding, 071000, People's Republic of China.
| |
Collapse
|
3
|
Sulastomo H, Dinarti LK, Hariawan H, Haryana SM. MicroRNA expression alteration in chronic thromboembolic pulmonary hypertension: A systematic review. Pulm Circ 2024; 14:e12443. [PMID: 39308943 PMCID: PMC11413763 DOI: 10.1002/pul2.12443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is marked by persistent blood clots in pulmonary arteries, leading to significant morbidity and mortality. Emerging evidence highlights the role of microRNAs (miRNAs) in pulmonary hypertension, though findings on miRNA expression in CTEPH remain limited and inconsistent. This systematic review evaluates miRNA expression changes in CTEPH and their direction. Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, we registered our protocol in International Prospective Register of Systematic Reviews (CRD42024524469). We included studies on miRNA expression in CTEPH with comparative or analytical designs, excluding nonhuman studies, interventions, non-English texts, conference abstracts, and editorials. Databases searched included PubMed, EMBASE, Scopus, CENTRAL, and ProQuest. The Quality Assessment of Diagnostic Accuracy Studies-2 tool assessed bias risk, and results were synthesized narratively. Of 313 unique studies, 39 full texts were reviewed, and 9 met inclusion criteria, totaling 235 participants. Blood samples were analysed using quantitative real time polymerase chain reaction. Seven miRNAs (miR-665, miR-3202, miR-382, miR-127, miR-664, miR-376c, miR-30) were uniformly upregulated, while nine (miR-20a-5p13, miR-17-5p, miR-93-5p, miR-22, let-7b, miR-106b-5p, miR-3148, miR-320-a, miR-320b) were downregulated in CTEPH patients. Two upregulated miRNAs (miR-127 and miR-30a) were consistently associated with previous evidence in the mechanism inducing the development of CTEPH, and five downregulated miRNAs (miR-20-a, miR-17-5p, miR-93-5p, let-7b, miR-106b-5p) were associated with a protective effect against CTEPH. We also identified gaps in the literature where the evidence for five upregulated miRNAs (miR-665, miR-3202, miR-382, miR-664 and miR-376c) and four downregulated miRNAs (miR-22, miR-3148, miR-320-a, and miR-320b) in CTEPH is conflicting. Our findings offer insights into the role of miRNAs in CTEPH and underscore the need for further research to validate these miRNAs as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Heru Sulastomo
- Department of Cardiology and Vascular Medicine, Faculty of MedicineUniversitas Sebelas MaretSurakartaIndonesia
| | - Lucia Kris Dinarti
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and NursingUniversitas Gadjah MadaYogyakartaIndonesia
| | - Hariadi Hariawan
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and NursingUniversitas Gadjah MadaYogyakartaIndonesia
| | - Sofia Mubarika Haryana
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and NursingUniversitas Gadjah MadaYogyakartaIndonesia
| |
Collapse
|
4
|
Kaymaz S, Aydın D, Uğur K, Çobankara V, Tan S. Expression Levels and Clinical Values of miR-195, miR-424, miR-10b, miR-103a-3p, and miR-542-3p in Vasculo-Behçet's Disease. Mediterr J Rheumatol 2024; 35:255-262. [PMID: 39211027 PMCID: PMC11350416 DOI: 10.31138/mjr.030623.elc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/30/2023] [Accepted: 09/05/2023] [Indexed: 09/04/2024] Open
Abstract
Objective MicroRNAs (miRNAs) are involved in a range of pathological and biological processes. Vascular involvement is an important complication associated with morbidity and mortality in Behçet's disease (BD). In this study, we aimed to evaluate the expression levels of miR-195, miR-424, miR-10b, miR-103a-3p, and miR-542-3p in Turkish patients with BD, and their possible association with vascular involvement and clinical activity. Methods This cross-sectional study included 61 BD patients and 25 age- and sex-matched healthy individuals. The patients were categorised into two groups based on the presence or absence of vascular involvement. Demographic data, disease duration, disease activity, and medical treatments were recorded. Disease activity was evaluated using the Behçet's Disease Current Activity Form (BDCAF) and the Behçet's Syndrome Activity Scale (BSAS). The expression levels of miRNAs were measured using real-time quantitative polymerase chain reaction (RT-qPCR). Results The comparison of the clinical features of BD patients with and without vascular involvement revealed no significant difference. However, the expression levels of miR-195, miR-424, miR-10b, miR-103a-3p, and miR-542-3p were significantly higher in BD patients than in healthy controls (p<0.001, p<0.001, p=0.010, p<0.01, p=0.039, respectively). Moreover, the expression level of miR-195 was significantly higher in vasculo-Behçet patients than in the other groups (p=0.0318). However, no significant association was found between the expression levels of miR-195 and clinical activity. Conclusion Our study results indicated elevated serum levels of miR-195 in BD patients, which may be associated with vascular involvement. Therefore, miR-195 could potentially serve as a biomarker for the diagnosis and monitoring of vasculo-Behçet's disease.
Collapse
Affiliation(s)
| | | | | | | | - Seçil Tan
- Department of Cancer Molecular Biology, Pamukkale University, Denizli, Turkey
| |
Collapse
|
5
|
Huang X, Bai S, Luo Y. Advances in research on biomarkers associated with acute myocardial infarction: A review. Medicine (Baltimore) 2024; 103:e37793. [PMID: 38608048 PMCID: PMC11018244 DOI: 10.1097/md.0000000000037793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
Acute myocardial infarction (AMI), the most severe cardiovascular event in clinical settings, imposes a significant burden with its annual increase in morbidity and mortality rates. However, it is noteworthy that mortality due to AMI in developed countries has experienced a decline, largely attributable to the advancements in medical interventions such as percutaneous coronary intervention. This trend highlights the importance of accurate diagnosis and effective treatment to preserve the myocardium at risk and improve patient outcomes. Conventional biomarkers such as myoglobin, creatine kinase isoenzymes, and troponin have been instrumental in the diagnosis of AMI. However, recent years have witnessed the emergence of new biomarkers demonstrating the potential to further enhance the accuracy of AMI diagnosis. This literature review focuses on the recent advancements in biomarker research in the context of AMI diagnosis.
Collapse
Affiliation(s)
| | - Suwen Bai
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People’s Hospital of Shenzhen, Shenzhen, China
| | - Yumei Luo
- Guangdong Medical University, Zhanjiang, China
- Cardiology Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People’s Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
6
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
7
|
Nougué H, Picard F, Cohen-Solal A, Logeart D, Launay JM, Vodovar N. Impact of sacubitril/valsartan on cardiac and systemic hypoxia in chronic heart failure. iScience 2024; 27:108520. [PMID: 38161412 PMCID: PMC10755360 DOI: 10.1016/j.isci.2023.108520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
In heart failure patients with reduced ejection fraction, Sacubitril/valsartan (S/V) increased proBNP T71 glycosylation, which is regulated negatively by hypoxia via miR-30a in vitro. Using a cohort of 73 HFrEF patients who were transitioned from standard HF medication to S/V, we found that the increase in proBNP T71 glycosylation after S/V was associated with a decrease in cardiac hypoxia. We further found that plasma levels of K709-acteylated HIF1α, HIF-regulated and HIF-independent biomarkers also evolved consistently with a decrease in hypoxia. We further confirmed that biomarker changes were related to hypoxia, in a rat model subjected to isobaric hypoxia. We measured them in rats subjected to isobaric hypoxia. Overall, these data strongly suggest that optimally treated HFrEF patients exhibited subclinical hypoxia that is improved by S/V. The data also posit proBNP T71 glycosylation as a biomarker of cardiac hypoxia.
Collapse
Affiliation(s)
- Hélène Nougué
- Université de Paris and Inserm UMR-S 942, Paris, France
- Department of Anaesthesiology and Intensive Care, Saint Louis – Lariboisière – Fernand Vidal University Hospital, Paris, France
| | - François Picard
- Service d’insuffisance cardiaque, Hôpital Cardiologique du Haut-Lévêque, Pessac, France
| | - Alain Cohen-Solal
- Université de Paris and Inserm UMR-S 942, Paris, France
- Department of Cardiology, Lariboisière Hospital, Paris, France
| | - Damien Logeart
- Université de Paris and Inserm UMR-S 942, Paris, France
- Department of Cardiology, Lariboisière Hospital, Paris, France
| | | | | |
Collapse
|
8
|
Wagh V, Nguemo F, Kiseleva Z, Mader RM, Hescheler J, Mohl W. Circulating microRNAs and cardiomyocyte proliferation in heart failure patients related to 10 years survival. ESC Heart Fail 2023; 10:3559-3572. [PMID: 37752740 PMCID: PMC10682869 DOI: 10.1002/ehf2.14516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023] Open
Abstract
AIMS Mechanochemical signalling drives organogenesis and is highly conserved in mammal evolution. Regaining recovery in myocardial jeopardy by inducing principles linking cardiovascular therapy and clinical outcome has been the dream of scientists for decades. Concepts involving embryonic pathways to regenerate adult failing hearts became popular in the early millennium. Since then, abundant data on stem cell research have been published, never reaching widespread application in heart failure therapy. Another conceptual access, using mechanotransduction in cardiac veins to limit myocardial decay, is pressure-controlled intermittent coronary sinus occlusion (PICSO). Recently, we reported acute molecular signs and signals of PICSO activating regulatory miRNA and inducing cell proliferation mimicking cardiac development in adult failing hearts. According to a previously formulated hypothesis, 'embryonic recall', this study aimed to define molecular signals involved in endogenous heart repair during PICSO and study their relation to patient survival. METHODS AND RESULTS We previously reported a study on the acute molecular effects of PICSO in an observational non-randomized study. Eight out of the thirty-two patients with advanced heart failure undergoing cardiac resynchronization therapy (CRT) were treated with PICSO. Survival was monitored over 10 years, and coronary sinus blood samples were collected during intervention before and after 20 min and tested for miRNA signalling and proliferation when co-cultured with cardiomyocytes. A numerically lower death rate post-CRT and PICSO as compared with control CRT only, and a non-significant reduction in all-cause mortality risk of 42% was observed (37.5% vs. 54.0%, relative risk = 0.58, 95% confidence interval: 0.17-2.05; P = 0.402). Four miRNAs involved in cell cycle, proliferation, morphogenesis, embryonic development, and apoptosis significantly increased concomitantly in survivors and PICSO compared with a decrease in non-survivors (hsa-miR Let7b, P < 0.01; hsa-miR- 421, P < 0.006; hsa-miR 363-3p, P < 0.03 and hsa-miR 19b-3p P < 0.01). In contrast, three miRNAs involved in proliferation and survival, determining cell fate, and recycling endosomes decreased in survivors and PICSO (hsa miR 101-3p, P < 0.03; hsa-miR 25-3p, P < 002; hsa-miR 30d-5p P < 0.04). In vitro cellular proliferation increased in survivors and lowered in non-survivors showing a pattern distinction, discriminating longevity according to up to 10-year survival in heart failure patients. CONCLUSIONS This study proposes that generating regenerative signals observed during PICSO intervention relate to patient outcomes. Morphogenetic pathways induced by periods of flow reversal in cardiac veins in a domino-like pattern transform embryonic into regenerative signals. Studies supporting the conversion of mechanochemical signals into regenerative molecules during PICSO are warranted to substantiate predictive power on patient longevity, opening new therapeutic avenues in otherwise untreatable heart failure.
Collapse
Affiliation(s)
- Vilas Wagh
- Merck Research LabsBostonMAUSA
- Center of Physiology and Pathophysiology, Institute of NeurophysiologyUniversity of CologneCologneGermany
| | - Filomain Nguemo
- Center of Physiology and Pathophysiology, Institute of NeurophysiologyUniversity of CologneCologneGermany
| | - Zlata Kiseleva
- Department of Cardiac Surgery emeritusMedical University ViennaViennaAustria
| | - Robert M. Mader
- Department of Medicine IMedical University ViennaViennaAustria
| | - Juergen Hescheler
- Center of Physiology and Pathophysiology, Institute of NeurophysiologyUniversity of CologneCologneGermany
| | - Werner Mohl
- Department of Cardiac Surgery emeritusMedical University ViennaViennaAustria
| |
Collapse
|
9
|
Gao Y, Wang Y, Li M, Gao C. Bioinformatics analysis of potential common pathogenic mechanisms for systemic lupus erythematosus and acute myocardial infarction. Lupus 2023; 32:1296-1309. [PMID: 37800460 DOI: 10.1177/09612033231202659] [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] [Indexed: 10/07/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) patients have a higher risk of acute myocardial infarction (AMI) compared to the general population. However, the underlying common mechanism of this association is not fully understood. This study aims to investigate the molecular mechanism of this complication. METHODS Gene expression profiles of SLE (GSE50772) and AMI (GSE66360) were obtained from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (DEGs) in SLE and AMI were identified, and functional annotation, protein-protein interaction (PPI) network analysis, module construction, and hub gene identification were performed. Additionally, transcription factor (TF)-gene regulatory network and TF-miRNA regulatory network were constructed for the hub genes. RESULTS 70 common DEGs (7 downregulated genes and 63 upregulated genes) were identified and were mostly enriched in signaling pathways such as the IL-17 signaling pathway, TNF signaling pathway, lipid metabolism, and atherosclerosis. Using cytoHubba, 12 significant hub genes were identified, including IL1B, TNF, FOS, CXCL8, JUN, PTGS2, FN1, EGR1, CXCL1, DUSP1, MMP9, and ZFP36. CONCLUSIONS This study reveals a common pathogenesis of SLE and AMI and provides new perspectives for further mechanism research. The identified common pathways and hub genes may have important clinical implications for the prevention and treatment of AMI in SLE patients.
Collapse
Affiliation(s)
- Yang Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Yunxia Wang
- Department of Radiology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Muwei Li
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Chang C, Cai RP, Su YM, Wu Q, Su Q. Mesenchymal Stem Cell-Derived Exosomal Noncoding RNAs as Alternative Treatments for Myocardial Ischemia-Reperfusion Injury: Current Status and Future Perspectives. J Cardiovasc Transl Res 2023; 16:1085-1098. [PMID: 37286924 PMCID: PMC10246878 DOI: 10.1007/s12265-023-10401-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023]
Abstract
Ischemic cardiomyopathy is treated mainly with thrombolytic drugs, percutaneous coronary intervention, and coronary artery bypass grafting to recanalize blocked vessels. Myocardial ischemia-reperfusion injury (MIRI) is an unavoidable complication of obstructive revascularization. Compared with those of myocardial ischemic injury, few effective therapeutic options are available for MIRI treatment. The pathophysiological mechanisms of MIRI involve the inflammatory response, the immune response, oxidative stress, apoptosis, intracellular Ca2+ overload, and cardiomyocyte energy metabolism. These mechanisms exacerbate MIRI. Mesenchymal stem cell-derived exosomes (MSC-EXOs) can alleviate MIRI through these mechanisms and, to some extent, prevent the limitations caused by direct MSC administration. Therefore, using MSC-EXOs instead of MSCs to treat MIRI is a potentially beneficial cell-free treatment strategy. In this review, we describe the mechanism of action of MSC-EXO-derived noncoding RNAs in the treatment of MIRI and discuss the advantages and limitations of this strategy, as well as possible future research directions.
Collapse
Affiliation(s)
- Chen Chang
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541000, China
| | - Ru-Ping Cai
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Ying-Man Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541000, China
| | - Qiang Wu
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100048, China.
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qiang Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541000, China.
| |
Collapse
|
11
|
Moatar AI, Chis AR, Romanescu M, Ciordas PD, Nitusca D, Marian C, Oancea C, Sirbu IO. Plasma miR-195-5p predicts the severity of Covid-19 in hospitalized patients. Sci Rep 2023; 13:13806. [PMID: 37612439 PMCID: PMC10447562 DOI: 10.1038/s41598-023-40754-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 08/16/2023] [Indexed: 08/25/2023] Open
Abstract
Predicting the clinical course of Covid-19 is a challenging task, given the multi-systemic character of the disease and the paucity of minimally invasive biomarkers of disease severity. Here, we evaluated the early (first two days post-admission) level of circulating hsa-miR-195-5p (miR-195, a known responder to viral infections and SARS-CoV-2 interactor) in Covid-19 patients and assessed its potential as a biomarker of disease severity. We show that plasma miR-195 correlates with several clinical and paraclinical parameters, and is an excellent discriminator between the severe and mild forms of the disease. Our Gene Ontology analysis of miR-195 targets differentially expressed in Covid-19 indicates a strong impact on cardiac mitochondria homeostasis, suggesting a possible role in long Covid and chronic fatigue syndrome (CFS) syndromes.
Collapse
Affiliation(s)
- Alexandra Ioana Moatar
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
- Doctoral School, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
| | - Aimee Rodica Chis
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
- Center for Complex Network Science, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
| | - Mirabela Romanescu
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
- Doctoral School, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
| | - Paula-Diana Ciordas
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
- Doctoral School, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
| | - Diana Nitusca
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
- Doctoral School, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
| | - Catalin Marian
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
- Center for Complex Network Science, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania
| | - Cristian Oancea
- Department of Infectious Diseases, Discipline of Pulmonology, University of Medicine and Pharmacy "Victor Babes", E. Murgu Square no.2, 300041, Timisoara, Romania
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, "Victor Babes" University of Medicine and Pharmacy Timisoara, E. Murgu Square 2, 300041, Timisoara, Romania
| | - Ioan-Ovidiu Sirbu
- Department of Biochemistry and Pharmacology, Discipline of Biochemistry, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania.
- Center for Complex Network Science, University of Medicine and Pharmacy "Victor Babes", E Murgu Square no.2, 300041, Timisoara, Romania.
- Timisoara Institute of Complex Systems, 18 Vasile Lucaciu Str, 300044, Timisoara, Romania.
| |
Collapse
|
12
|
Danckwardt S, Trégouët DA, Castoldi E. Post-transcriptional control of haemostatic genes: mechanisms and emerging therapeutic concepts in thrombo-inflammatory disorders. Cardiovasc Res 2023; 119:1624-1640. [PMID: 36943786 PMCID: PMC10325701 DOI: 10.1093/cvr/cvad046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 03/23/2023] Open
Abstract
The haemostatic system is pivotal to maintaining vascular integrity. Multiple components involved in blood coagulation have central functions in inflammation and immunity. A derailed haemostasis is common in prevalent pathologies such as sepsis, cardiovascular disorders, and lately, COVID-19. Physiological mechanisms limit the deleterious consequences of a hyperactivated haemostatic system through adaptive changes in gene expression. While this is mainly regulated at the level of transcription, co- and posttranscriptional mechanisms are increasingly perceived as central hubs governing multiple facets of the haemostatic system. This layer of regulation modulates the biogenesis of haemostatic components, for example in situations of increased turnover and demand. However, they can also be 'hijacked' in disease processes, thereby perpetuating and even causally entertaining associated pathologies. This review summarizes examples and emerging concepts that illustrate the importance of posttranscriptional mechanisms in haemostatic control and crosstalk with the immune system. It also discusses how such regulatory principles can be used to usher in new therapeutic concepts to combat global medical threats such as sepsis or cardiovascular disorders.
Collapse
Affiliation(s)
- Sven Danckwardt
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK),
Berlin, Germany
- Posttranscriptional Gene Regulation, University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University
Medical Centre Mainz, Langenbeckstr. 1, 55131
Mainz, Germany
- Center for Healthy Aging (CHA), Mainz,
Germany
| | - David-Alexandre Trégouët
- INSERM, Bordeaux Population Health Research Center, UMR 1219, Department of
Molecular Epidemiology of Vascular and Brain Disorders (ELEANOR), University of
Bordeaux, Bordeaux, France
| | - Elisabetta Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht
(CARIM), Maastricht University, Universiteitsingel 50, 6229
ER Maastricht, The Netherlands
| |
Collapse
|
13
|
Raffee LA, Alawneh KZ, Alshehabat MAM, Haddad H, Jaradat SA. MicroRNA profiling in dogs undergoing induced ischemic heart infarction: An experimental study. Vet World 2023; 16:1319-1324. [PMID: 37577186 PMCID: PMC10421551 DOI: 10.14202/vetworld.2023.1319-1324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/08/2023] [Indexed: 08/15/2023] Open
Abstract
Background and Aim MicroRNAs (miRNAs) play an important role in various biological functions. According to many studies, miRNA expression is tissue-specific, strongly controlled throughout embryogenesis, and over- or under-expressed in numerous disorders, including cardiovascular pathologies. This study aimed to screen, characterize, and profile many induced biomarkers (miRNAs) in dog serum before and after experimentally inducing a regional myocardial infarction (MI) by occluding the coronary arteries under general anesthesia. Materials and Methods A preclinical experimental animal study recruited 12 healthy canine dogs. The selected canine dogs were anesthetized with 1 mg/kg xylazine and 15 mg/kg ketamine before undergoing femoral arterial catheterization under fluoroscopic supervision. Commercial assay kits were used to purify total RNA and miRNA before the occlusion and 2 h after the occlusion according to the manufacturer's guidelines, and the samples were stored in RNase/DNase-free water at -80°C. Data were analyzed by GraphPad Prism 5.0 software (GraphPad Prism, San Diego, CA) SPSS, and GenEx software (www.multid.se) or (REST V3). Results Among 325 transcribed genes, 20 were identified in 2 h. After MI, 14 biomarkers were negative, indicating downregulation, and 6 (3-F08, 3-B10, 4-A11, 1-A06, 2-E01, 3-F10) were positive, indicating upregulation. Polymerase chain reaction assay results showed a normalized fold-change in gene expression in the test sample. Fold values >1 represented a biologically significant change. Conclusion Profiling of miRNAs before and after MI in a dog model revealed upregulation of six previously unidentified biomarkers (3-F08, 3-B10, 4-A11, 1-A06, 2-E01, and 3-F10), indicating various miRNA regulatory patterns.
Collapse
Affiliation(s)
- Liqaa A. Raffee
- Department of Accident and Emergency Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Khaled Z. Alawneh
- Department of Diagnostic Radiology and Nuclear Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Musa Ahmed Mohammed Alshehabat
- Department of Clinical Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Hazem Haddad
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Saied A. Jaradat
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
14
|
Da Fonseca Ferreira A, Wei J, Zhang L, Macon CJ, Degnan B, Jayaweera D, Hare JM, Kolber MA, Bellio M, Khan A, Pan Y, Dykxhoorn DM, Wang L, Dong C. HIV Promotes Atherosclerosis via Circulating Extracellular Vesicle MicroRNAs. Int J Mol Sci 2023; 24:7567. [PMID: 37108729 PMCID: PMC10146407 DOI: 10.3390/ijms24087567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
People living with HIV (PLHIV) are at a higher risk of having cerebrocardiovascular diseases (CVD) compared to HIV negative (HIVneg) individuals. The mechanisms underlying this elevated risk remains elusive. We hypothesize that HIV infection results in modified microRNA (miR) content in plasma extracellular vesicles (EVs), which modulates the functionality of vascular repairing cells, i.e., endothelial colony-forming cells (ECFCs) in humans or lineage negative bone marrow cells (lin- BMCs) in mice, and vascular wall cells. PLHIV (N = 74) have increased atherosclerosis and fewer ECFCs than HIVneg individuals (N = 23). Plasma from PLHIV was fractionated into EVs (HIVposEVs) and plasma depleted of EVs (HIV PLdepEVs). HIVposEVs, but not HIV PLdepEVs or HIVnegEVs (EVs from HIVneg individuals), increased atherosclerosis in apoE-/- mice, which was accompanied by elevated senescence and impaired functionality of arterial cells and lin- BMCs. Small RNA-seq identified EV-miRs overrepresented in HIVposEVs, including let-7b-5p. MSC (mesenchymal stromal cell)-derived tailored EVs (TEVs) loaded with the antagomir for let-7b-5p (miRZip-let-7b) counteracted, while TEVs loaded with let-7b-5p recapitulated the effects of HIVposEVs in vivo. Lin- BMCs overexpressing Hmga2 (a let-7b-5p target gene) lacking the 3'UTR and as such is resistant to miR-mediated regulation showed protection against HIVposEVs-induced changes in lin- BMCs in vitro. Our data provide a mechanism to explain, at least in part, the increased CVD risk seen in PLHIV.
Collapse
Affiliation(s)
- Andrea Da Fonseca Ferreira
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jianqin Wei
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lukun Zhang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Conrad J. Macon
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Bernard Degnan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dushyantha Jayaweera
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michael A. Kolber
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michael Bellio
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yue Pan
- Biostatistics Division, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Derek M. Dykxhoorn
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Liyong Wang
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chunming Dong
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Section of Cardiology, Department of Medicine, Miami VA Health System, University of Miami, Miami, FL 33146, USA
| |
Collapse
|
15
|
Venugopal P, George M, Kandadai SD, Balakrishnan K, Uppugunduri CRS. Prioritization of microRNA biomarkers for a prospective evaluation in a cohort of myocardial infarction patients based on their mechanistic role using public datasets. Front Cardiovasc Med 2022; 9:981335. [PMID: 36407428 PMCID: PMC9668885 DOI: 10.3389/fcvm.2022.981335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Background MicroRNAs (miR) have proven to be promising biomarkers for several diseases due to their diverse functions, stability and tissue/organ-specific nature. Identification of new markers with high sensitivity and specificity will help in risk reduction in acute myocardial infarction (AMI) patients with chest pain and also prevent future adverse outcomes. Hence the aim of this study was to perform a detailed in silico analysis for identifying the mechanistic role of miRs involved in the pathogenesis/prognosis of AMI for prospective evaluation in AMI patients. Methods miR profiling data was extracted from GSE148153 and GSE24591 datasets using the GEO2R gene expression omnibus repository and analyzed using limma algorithm. Differentially expressed miRs were obtained by comparing MI patients with corresponding controls after multiple testing corrections. Data mining for identifying candidate miRs from published literature was also performed. Target prediction and gene enrichment was done using standard bioinformatics tools. Disease specific analysis was performed to identify target genes specific for AMI using open targets platform. Protein-protein interaction and pathway analysis was done using STRING database and Cytoscape platform. Results and conclusion The analysis revealed significant miRs like let-7b-5p, let-7c-5p, miR-4505, and miR-342-3p in important functions/pathways including phosphatidylinositol-3-kinase/AKT and the mammalian target of rapamycin, advanced glycation end products and its receptor and renin–angiotensin–aldosterone system by directly targeting angiotensin II receptor type 1, forkhead box protein O1, etc. With this approach we were able to prioritize the miR candidates for a prospective clinical association study in AMI patients of south Indian origin.
Collapse
Affiliation(s)
| | - Melvin George
- Clinical Research Department, Hindu Mission Hospital, Chennai, India
| | | | | | - Chakradhara Rao S. Uppugunduri
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- *Correspondence: Chakradhara Rao S. Uppugunduri,
| |
Collapse
|
16
|
Sacubitril/valsartan (LCZ696) ameliorates hyperthyroid-induced cardiac hypertrophy in male rats through modulation of miR-377, let-7 b, autophagy, and fibrotic signaling pathways. Sci Rep 2022; 12:14654. [PMID: 36030321 PMCID: PMC9420135 DOI: 10.1038/s41598-022-18860-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022] Open
Abstract
Hyperthyroidism is associated with cardiac hypertrophy, fibrosis, and increased risk of cardiovascular mortality. Sacubitril/valsartan (LCZ696) is a new combined drug that has shown promise for the treatment of hyperthyroidism-associated heart failure; however, the underlying molecular mechanisms, including the contributions of epigenetic regulation, remain unclear. The present study was designed to investigate the therapeutic efficacy of LCZ696 and the potential contributions of microRNA regulation in a rat model of hyperthyroidism-induced cardiac hypertrophy. Cardiac hypertrophy was induced by intraperitoneal administration of levothyroxine. Sixty adult male Wistar rats were randomly allocated to four equal groups (15 rats each): control, cardiac hypertrophy (CH), CH + valsartan, and CH + LCZ696. Treatment with LCZ696 or valsartan significantly improved hemodynamic abnormalities, normalized serum concentrations of natriuretic peptide, fibroblast growth factor-23, and cardiac inflammatory markers compared to CH group rats. Treatment with LCZ696 or valsartan also normalized myocardial expression levels of autophagy markers, fibrotic markers, PPAR-ϒ, mir-377, and let-7b. In addition, both valsartan and LCZ696 ameliorated collagen deposition, ventricular degeneration, and various ultrastructural abnormalities induced by levothyroxine. The beneficial effects of LCZ696 were superior to those of valsartan alone. The superior efficacy of LCZ696 may be explained by the stronger modulation of miR-377 and let-7b.
Collapse
|
17
|
Nikolajevic J, Ariaee N, Liew A, Abbasnia S, Fazeli B, Sabovic M. The Role of MicroRNAs in Endothelial Cell Senescence. Cells 2022; 11:cells11071185. [PMID: 35406749 PMCID: PMC8997793 DOI: 10.3390/cells11071185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/05/2022] Open
Abstract
Cellular senescence is a complex, dynamic process consisting of the irreversible arrest of growth and gradual deterioration of cellular function. Endothelial senescence affects the cell’s ability to repair itself, which is essential for maintaining vascular integrity and leads to the development of endothelial dysfunction, which has an important role in the pathogenesis of cardiovascular diseases. Senescent endothelial cells develop a particular, senescence-associated secretory phenotype (SASP) that detrimentally affects both surrounding and distant endothelial cells, thereby facilitating the ageing process and development of age-related disorders. Recent studies highlight the role of endothelial senescence and its dysfunction in the pathophysiology of several age-related diseases. MicroRNAs are small noncoding RNAs that have an important role in the regulation of gene expression at the posttranscriptional level. Recently, it has been discovered that miRNAs could importantly contribute to endothelial cell senescence. Overall, the research focus has been shifting to new potential mechanisms and targets to understand and prevent the structural and functional changes in ageing senescent endothelial cells in order to prevent the development and limit the progression of the wide spectrum of age-related diseases. The aim of this review is to provide some insight into the most important pathways involved in the modulation of endothelial senescence and to reveal the specific roles of several miRNAs involved in this complex process. Better understanding of miRNA’s role in endothelial senescence could lead to new approaches for prevention and possibly also for the treatment of endothelial cells ageing and associated age-related diseases.
Collapse
Affiliation(s)
- Jovana Nikolajevic
- Department of Vascular Diseases, University Medical Center, 1000 Ljubljana, Slovenia;
- Correspondence:
| | - Nazila Ariaee
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad 1696700, Iran;
| | - Aaron Liew
- Department of Medicine, National University of Galway, H91 CF50 Galway, Ireland;
| | - Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad 1696700, Iran;
| | - Bahare Fazeli
- Vascular Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 1696700, Iran;
| | - Miso Sabovic
- Department of Vascular Diseases, University Medical Center, 1000 Ljubljana, Slovenia;
- Medical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
18
|
Kucher AN, Koroleva IA, Zarubin AA, Nazarenko MS. MicroRNAs as the Potential Regulators of SARS-CoV-2 Infection and Modifiers of the COVID-19 Clinical Features. Mol Biol 2022; 56:29-45. [PMID: 35464324 PMCID: PMC9016216 DOI: 10.1134/s0026893322010034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/14/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) warrants the identification of factors that may determine both risk and severity of infection. The factors include microRNAs that have a wide regulatory potential and hence are particularly interesting. The review focuses on the potential roles of human microRNAs and the viral genome as well as microRNAs in SARS-CoV-2 infection and clinical features of COVID-19. The review summarizes the information about the human microRNAs that are thought to specifically bind to the SARS-CoV-2 genome and considers their expression levels in various organs (cells) in both healthy state and pathologies that are risk factors for severe COVID-19. Potential mechanisms whereby SARS-CoV-2 may affect the clinical features of COVID-19 are discussed in brief. The mechanisms include blocking of human microRNAs and RNA-binding proteins, changes in gene expression in infected cells, and possible epigenetic modifications of the human genome with the participation of coronavirus microRNAs.
Collapse
Affiliation(s)
- A. N. Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - Iu. A. Koroleva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - A. A. Zarubin
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - M. S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| |
Collapse
|
19
|
Song F, Li JZ, Wu Y, Wu WY, Wang Y, Li G. Ubiquitinated ligation protein NEDD4L participates in MiR-30a-5p attenuated atherosclerosis by regulating macrophage polarization and lipid metabolism. MOLECULAR THERAPY - NUCLEIC ACIDS 2021; 26:1303-1317. [PMID: 34853729 PMCID: PMC8609110 DOI: 10.1016/j.omtn.2021.10.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/22/2021] [Accepted: 10/28/2021] [Indexed: 11/04/2022]
Abstract
MiR-30a-5p plays an important role in various cardiovascular diseases, but its effect in atherosclerosis has not been reported. Apolipoprotein E-deficient (Apo E−/−) mice were used to investigate the role of miR-30a-5p in atherosclerosis, and the underlying mechanism was investigated in vivo and in vitro. The fluorescence in situ hybridization test revealed that miR-30a-5p was expressed in Apo E−/− mice lesions. Nevertheless, in RAW264.7 macrophages, the expression of miR-30a-5p was reduced by lipopolysaccharide (LPS) or oxidized low-density lipoprotein. MiR-30a-5p-ago-treated Apo E−/− mice significantly reduced lesion areas in the aorta and aortic root, reduced levels of lipoprotein and pro-inflammatory cytokines, and increased levels of anti-inflammatory cytokines. The ratio of M1/M2 macrophages was decreased in miR-30a-5p-ago-treated Apo E−/− mice and LPS-treated RAW264.7 macrophages by the regulation of Smad-1/2 phosphorylation. MiR-30a-5p reduced lipid uptake in oxidized low-density lipoprotein-treated macrophages by regulating the expression of PPAR-γ, ABCA1, ABCG1, LDLR, and PCSK9. Ubiquitinated ligase NEDD4L was identified as a target of miR-30a-5p. Interestingly, knockdown of NEDD4L decreased the M1/M2 ratio and oxidized low-density lipoprotein uptake in macrophages by inhibiting the ubiquitination of PPAR-γ and phosphorylation of Smad-1/2 and regulating ABCA1, ABCG1, LDLR, and PCSK9. We demonstrated a novel effect and mechanism of miR-30a-5p in atherosclerosis.
Collapse
|
20
|
Wang B, Li Y, Hao X, Yang J, Han X, Li H, Li T, Wang D, Teng Y, Ma L, Li Y, Zhao M, Wang X. Comparison of the Clinical Value of miRNAs and Conventional Biomarkers in AMI: A Systematic Review. Front Genet 2021; 12:668324. [PMID: 34220945 PMCID: PMC8248539 DOI: 10.3389/fgene.2021.668324] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/04/2021] [Indexed: 01/22/2023] Open
Abstract
Background/Aims: This study aimed to compare the clinical value of the peak time point and area under the curve (AUC) of miRNAs and conventional biomarkers in acute myocardial infarction (AMI). Methods: A literature search was carried out in PubMed, Web of Science, Embase, and Cochrane systematically. Screening studies, extracting data, and assessing article quality were performed independently by two researchers. Also, the names of miRNAs in the included studies were standardized by the miRBase database. Results: A total of 40 studies, encompassing 6,960 participants, were included in this systematic review. The samples of circulating miRNAs were mainly from the plasma. The results of this systematic review displayed that miR-1-3p, miR-19b-3p, miR-22-5p, miR-122-5p, miR-124-3p, miR-133a/b, miR-134-5p, miR-150-5p, miR-186-5p, miR-208a, miR-223-3p, miR-483-5p, and miR-499a-5p reached peak time earlier and showed a shorter time window than the conventional biomarkers despite the different collection times of initial blood samples. miR-1-3p, miR-19b-3p, miR-133a/b, miR-208a/b, miR-223-3p, miR-483-5p, and miR-499a-5p were shown to be more valuable than classical biomarkers for the early diagnosis of AMI, and these miRNAs appeared to have the most potential biomarkers within 4 h of the onset of symptoms except miR-133a/b and miR-208b. Moreover, combined miRNAs or miRNAs combined with classical biomarkers could compensate for the deficiency of single miRNA and conventional biomarker in sensitivity or specificity for an optimal clinical value. Conclusions: miR-1-3p, miR-19b-3p, miR-208a, miR-223-3p, miR-483-5p, and miR-499a-5p are promising biomarkers for AMI due to their satisfactory diagnostic accuracy and short time window (within 4 h of the onset of symptoms).
Collapse
Affiliation(s)
- Baofu Wang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xuezeng Hao
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Yang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Haiyan Li
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Tong Li
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Dayang Wang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Liang Ma
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yao Li
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xian Wang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China.,Institute of Cardiovascular Diseases, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Kheyfets VO, Dufva MJ, Boehm M, Tian X, Qin X, Tabakh JE, Truong U, Ivy D, Spiekerkoetter E. The left ventricle undergoes biomechanical and gene expression changes in response to increased right ventricular pressure overload. Physiol Rep 2021; 8:e14347. [PMID: 32367677 PMCID: PMC7198956 DOI: 10.14814/phy2.14347] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 01/02/2023] Open
Abstract
Pulmonary hypertension (PH) results in right ventricular (RV) pressure overload and eventual failure. Current research efforts have focused on the RV while overlooking the left ventricle (LV), which is responsible for mechanically assisting the RV during contraction. The objective of this study is to evaluate the biomechanical and gene expression changes occurring in the LV due to RV pressure overload in a mouse model. Nine male mice were divided into two groups: (a) pulmonary arterial banding (PAB, N = 4) and (b) sham surgery (Sham, N = 5). Tagged and steady‐state free precision cardiac MRI was performed on each mouse at 1, 4, and 7 weeks after surgery. At/week7, the mice were euthanized following right/left heart catheterization with RV/LV tissue harvested for histology and gene expression (using RT‐PCR) studies. Compared to Sham mice, the PAB group revealed a significantly decreased LV and RV ejection fraction, and LV maximum torsion and torsion rate, within the first week after banding. In the PAB group, there was also a slight but significant increase in LV perivascular fibrosis, which suggests elevated myocardial stress. LV fibrosis was also accompanied with changes in gene expression in the hypertensive group, which was correlated with LV contractile mechanics. In fact, principal component (PC) analysis of LV gene expression effectively separated Sham and PAB mice along PC2. Changes in LV contractile mechanics were also significantly correlated with unfavorable changes in RV contractile mechanics, but a direct causal relationship was not established. In conclusion, a purely biomechanical insult of RV pressure overload resulted in biomechanical and transcriptional changes in both the RV and LV. Given that the RV relies on the LV for contractile energy assistance, considering the LV could provide prognostic and therapeutic targets for treating RV failure in PH.
Collapse
Affiliation(s)
- Vitaly O Kheyfets
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Department of Pediatrics, Section of Cardiology, Children's Hospital Colorado, Aurora, CO, USA
| | - Melanie J Dufva
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Department of Pediatrics, Section of Cardiology, Children's Hospital Colorado, Aurora, CO, USA
| | - Mario Boehm
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, USA.,German Center for Lung Research (DZL), Giessen, Germany
| | - Xuefeit Tian
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, USA
| | - Xulei Qin
- Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Jennifer E Tabakh
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Uyen Truong
- Department of Pediatrics, Section of Cardiology, Children's Hospital Colorado, Aurora, CO, USA.,Department of Pediatrics - Division of Cardiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Dunbar Ivy
- Department of Pediatrics, Section of Cardiology, Children's Hospital Colorado, Aurora, CO, USA
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, USA.,Cardiovascular Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
22
|
Xu L, Zhang H, Wang Y, Guo W, Gu L, Yang A, Ma S, Yang Y, Wu K, Jiang Y. H3K14 hyperacetylation‑mediated c‑Myc binding to the miR‑30a‑5p gene promoter under hypoxia postconditioning protects senescent cardiomyocytes from hypoxia/reoxygenation injury. Mol Med Rep 2021; 23:468. [PMID: 33880587 PMCID: PMC8097758 DOI: 10.3892/mmr.2021.12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 02/22/2021] [Indexed: 11/25/2022] Open
Abstract
Our previous study reported that microRNA (miR)‑30a‑5p upregulation under hypoxia postconditioning (HPostC) exert a protective effect on aged H9C2 cells against hypoxia/reoxygenation injury via DNA methyltransferase 3B‑induced DNA hypomethylation at the miR‑30a‑5p gene promoter. This suggests that miR‑30a‑5p may be a potential preventative and therapeutic target for ischemic heart disease in aged myocardium. The present study aimed to investigate the underlying mechanisms of miR‑30a‑5p transcription in aged myocardium in ischemic heart disease. Cardiomyocytes were treated with 8 mg/ml D‑galactose for 9 days, and then exposed to hypoxic conditions. Cell viability was determined using a cell viability assay. Expression levels of histone deacetylase 2 (HDAC2), LC3B‑II/I, beclin‑1 and p62 were detected via reverse transcription‑quantitative PCR and western blotting. Chromatin immunoprecipitation‑PCR and luciferase reporter assays were performed to evaluate the effect of c‑Myc binding and activity on the miR‑30a‑5p promoter in senescent cardiomyocytes following HPostC. It was found that HPostC enhanced the acetylation levels of H3K14 at the miR‑30a‑5p gene promoter in senescent cardiomyocytes, which attributed to the decreased expression of HDAC2. In addition, c‑Myc could positively regulate miR‑30a‑5p transcription to inhibit senescent cardiomyocyte autophagy. Mechanically, it was observed that increased H3K14 acetylation level exposed to romidepsin facilitated c‑Myc binding to the miR‑30a‑5p gene promoter region, which led to the increased transcription of miR‑30a‑5p. Taken together, these results demonstrated that HDAC2‑mediated H3K14 hyperacetylation promoted c‑Myc binding to the miR‑30a‑5p gene promoter, which contributed to HPostC senescent cardioprotection.
Collapse
Affiliation(s)
- Lingbo Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Huiping Zhang
- Department of Prenatal Diagnosis Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yanhua Wang
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Wei Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Lingyu Gu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Anning Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Shengchao Ma
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yong Yang
- Department of Nuclear Medicine, The People's Hospital in Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Kai Wu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yideng Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
23
|
Han X, Li T, Fan Y, Wang X, Gu W, Lu W, Yin Y, Meng Q, Zhang W, Zhao J, Zhang F, Fu Y. Screening of 20 Mycobacterium tuberculosis sRNAs in plasma for detection of active pulmonary tuberculosis. Tuberculosis (Edinb) 2021; 129:102086. [PMID: 34051642 DOI: 10.1016/j.tube.2021.102086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Mycobacterium tuberculosis (MTB) sRNAs are abundant. However, the level of MTB sRNA in peripheral blood remains elusive. METHODS Twenty MTB sRNAs annotated in the reference genome of H37Rv were detected in the plasma of 170 active pulmonary tuberculosis patients and 124 healthy people by qRT-PCR detection system. The differential expression of sRNAs were analyzed in two groups. The value of sRNAs for diagnosis of active tuberculosis were evaluated by ROC curve analysis. RESULTS Eight of the 20 sRNAs (MTS2823, MTS0997, MTS1338, ASdes, G2, C8, mcr15 and MTS1082) were found in at least 50% of the samples detected. The relative expression levels of MTS2823, MTS0997, MTS1338 and ASdes in plasma of tuberculosis patients were statistically higher than those in healthy controls. ROC curve analysis showed that the AUC of MTS0997, MTS1338, MTS2823 and ASdes were 0.8935 (95% CI 0.8109-0.9760), 0.8722 (95% CI 0.7862-0.9581), 0.8208 (95% CI 0.7246-0.9170) and 0.5792 (95% CI 0.4240-0.7344), respectively. The AUC value of combination of MTS0997, MTS1338 and MTS2823 was 0.914 (95% CI 0.8281-0.9926). CONCLUSIONS MTB sRNAs MTS2823, MTS0997 and MTS1338 have the potential to be plasma biomarkers for active pulmonary tuberculosis.
Collapse
Affiliation(s)
- Xue Han
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Ting Li
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Yunfan Fan
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Xinyang Wang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Wei Gu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Weinan Lu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Yian Yin
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Qingtai Meng
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Wenli Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Jizi Zhao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China.
| | - Fengmin Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, China.
| | - Yingmei Fu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 194, Xuefu Road, Nangang District, Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, China.
| |
Collapse
|
24
|
Alimoradi N, Firouzabadi N, Fatehi R. Metformin and insulin-resistant related diseases: Emphasis on the role of microRNAs. Biomed Pharmacother 2021; 139:111662. [PMID: 34243629 DOI: 10.1016/j.biopha.2021.111662] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Metformin is one of the most prescribed drugs in type II diabetes (T2DM) which has recently found new applications in the prevention and treatment of various illnesses, from metabolic disorders to cardiovascular and age-related diseases. Metformin improves insulin resistance (IR) by modulating metabolic mechanisms and mitochondrial biogenesis. Alternation of microRNAs (miRs) in the treatment of IR-related illnesses has been observed by metformin therapy. MiRs are small non-coding RNAs that play important roles in RNA silencing, targeting the 3'untranslated region (3'UTR) of most mRNAs and inhibiting the translation of related proteins. As a result, their dysregulation is associated with many diseases. Metformin may alter miRs levels in the treatment of various diseases by AMPK-dependent or AMPK-independent mechanisms. Here, we summarized the therapeutic role of metformin by modifying the aberrant expression of miRs as potential biomarkers or therapeutic targets in diseases in which IR plays a key role.
Collapse
Affiliation(s)
- Nahid Alimoradi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
25
|
Kaur A, Mackin ST, Schlosser K, Wong FL, Elharram M, Delles C, Stewart DJ, Dayan N, Landry T, Pilote L. Systematic review of microRNA biomarkers in acute coronary syndrome and stable coronary artery disease. Cardiovasc Res 2021; 116:1113-1124. [PMID: 31782762 DOI: 10.1093/cvr/cvz302] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/24/2019] [Accepted: 11/26/2019] [Indexed: 01/03/2023] Open
Abstract
The aim of this systematic review was to assess dysregulated miRNA biomarkers in coronary artery disease (CAD). Dysregulated microRNA (miRNAs) have been shown to be linked to cardiovascular pathologies including CAD and may have utility as diagnostic and prognostic biomarkers. We compared miRNAs identified in acute coronary syndrome (ACS) compared with stable CAD and control populations. We conducted a systematic search of controlled vocabulary and free text terms related to ACS, stable CAD and miRNA in Biosis Previews (OvidSP), The Cochrane Library (Wiley), Embase (OvidSP), Global Health (OvidSP), Medline (PubMed and OvidSP), Web of Science (Clarivate Analytics), and ClinicalTrials.gov which yielded 7370 articles. Of these, 140 original articles were appropriate for data extraction. The most frequently reported miRNAs in any CAD (miR-1, miR-133a, miR-208a/b, and miR-499) are expressed abundantly in the heart and play crucial roles in cardiac physiology. In studies comparing ACS cases with stable CAD patients, miR-21, miR-208a/b, miR-133a/b, miR-30 family, miR-19, and miR-20 were most frequently reported to be dysregulated in ACS. While a number of miRNAs feature consistently across studies in their expression in both ACS and stable CAD, when compared with controls, certain miRNAs were reported as biomarkers specifically in ACS (miR-499, miR-1, miR-133a/b, and miR-208a/b) and stable CAD (miR-215, miR-487a, and miR-502). Thus, miR-21, miR-133, and miR-499 appear to have the most potential as biomarkers to differentiate the diagnosis of ACS from stable CAD, especially miR-499 which showed a correlation between the level of their concentration gradient and myocardial damage. Although these miRNAs are potential diagnostic biomarkers, these findings should be interpreted with caution as the majority of studies conducted predefined candidate-driven assessments of a limited number of miRNAs (PROSPERO registration: CRD42017079744).
Collapse
Affiliation(s)
- Amanpreet Kaur
- Centre for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, 5252 de Maisonneuve West, 2B.39, Montreal QC H4A 3S5, Canada
| | - Sharon T Mackin
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Kenny Schlosser
- Ottawa Hospital Research Institute and Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Fui Lin Wong
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Malik Elharram
- Department of Medicine, McGill University Health Centre, Montreal, Canada
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Duncan J Stewart
- Ottawa Hospital Research Institute and Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Natalie Dayan
- Centre for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, 5252 de Maisonneuve West, 2B.39, Montreal QC H4A 3S5, Canada.,Department of Medicine, McGill University Health Centre, Montreal, Canada
| | - Tara Landry
- Medical Library, Montreal General Hospital, McGill University Health Centre, Montreal, Canada
| | - Louise Pilote
- Centre for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, 5252 de Maisonneuve West, 2B.39, Montreal QC H4A 3S5, Canada.,Department of Medicine, McGill University Health Centre, Montreal, Canada
| |
Collapse
|
26
|
A novel rationale for targeting FXI: Insights from the hemostatic microRNA targetome for emerging anticoagulant strategies. Pharmacol Ther 2021; 218:107676. [DOI: 10.1016/j.pharmthera.2020.107676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
|
27
|
Zheng M, Wang M. A narrative review of the roles of the miR-15/107 family in heart disease: lessons and prospects for heart disease. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:66. [PMID: 33553359 PMCID: PMC7859774 DOI: 10.21037/atm-20-6073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heart disease is one of the leading causes of morbidity and mortality globally. To reduce morbidity and mortality among patients with heart disease, it is important to identify drug targets and biomarkers for more effective diagnosis, prognosis, and treatment. MicroRNAs (miRNAs) are characterized as a group of endogenous, small non-coding RNAs, which function by directly inhibiting target genes. The miR-15/107 family is a group of evolutionarily conserved miRNAs comprising 10 members that share an identical motif of AGCAGC, which determines overlapping target genes and cooperation in the biological process. Accumulating evidence has demonstrated the predominant dysregulation of the miR-15/107 family in cardiovascular disease, neurodegenerative disease, and cancer. In this review, we summarize the current understanding of the miR-15/107 family, focusing on its role in the regulation in the development of the heart and the progression of heart disease. We also discuss the potential of different members of the miR-15/107 family as biomarkers for diverse heart disease, as well as the current applications and challenges in the use of the miR-15/107 family in clinical trials for various disease. This paper hopes to explore the potential of the miR-15/107 family as therapeutic targets or biomarkers and to provide directions for future research.
Collapse
Affiliation(s)
- Manni Zheng
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Videira RF, da Costa Martins PA, Falcão-Pires I. Non-Coding RNAs as Blood-Based Biomarkers in Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21239285. [PMID: 33291434 PMCID: PMC7730567 DOI: 10.3390/ijms21239285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
In 2020, cardiovascular diseases (CVDs) remain a leading cause of mortality and morbidity, contributing to the burden of the already overloaded health system. Late or incorrect diagnosis of patients with CVDs compromises treatment efficiency and patient's outcome. Diagnosis of CVDs could be facilitated by detection of blood-based biomarkers that reliably reflect the current condition of the heart. In the last decade, non-coding RNAs (ncRNAs) present on human biofluids including serum, plasma, and blood have been reported as potential biomarkers for CVDs. This paper reviews recent studies that focus on the use of ncRNAs as biomarkers of CVDs.
Collapse
Affiliation(s)
- Raquel Figuinha Videira
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.F.V.); (P.A.d.C.M.)
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Paula A. da Costa Martins
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.F.V.); (P.A.d.C.M.)
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, 6229 ER Maastricht, The Netherlands
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Inês Falcão-Pires
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
29
|
De Martinis M, Ginaldi L, Allegra A, Sirufo MM, Pioggia G, Tonacci A, Gangemi S. The Osteoporosis/Microbiota Linkage: The Role of miRNA. Int J Mol Sci 2020; 21:E8887. [PMID: 33255179 PMCID: PMC7727697 DOI: 10.3390/ijms21238887] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Hundreds of trillions of bacteria are present in the human body in a mutually beneficial symbiotic relationship with the host. A stable dynamic equilibrium exists in healthy individuals between the microbiota, host organism, and environment. Imbalances of the intestinal microbiota contribute to the determinism of various diseases. Recent research suggests that the microbiota is also involved in the regulation of the bone metabolism, and its alteration may induce osteoporosis. Due to modern molecular biotechnology, various mechanisms regulating the relationship between bone and microbiota are emerging. Understanding the role of microbiota imbalances in the development of osteoporosis is essential for the development of potential osteoporosis prevention and treatment strategies through microbiota targeting. A relevant complementary mechanism could be also constituted by the permanent relationships occurring between microbiota and microRNAs (miRNAs). miRNAs are a set of small non-coding RNAs able to regulate gene expression. In this review, we recapitulate the physiological and pathological meanings of the microbiota on osteoporosis onset by governing miRNA production. An improved comprehension of the relations between microbiota and miRNAs could furnish novel markers for the identification and monitoring of osteoporosis, and this appears to be an encouraging method for antagomir-guided tactics as therapeutic agents.
Collapse
Affiliation(s)
- Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.G.); (M.M.S.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.G.); (M.M.S.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Maria Maddalena Sirufo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (L.G.); (M.M.S.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy;
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
30
|
Lv XB, Niu QH, Zhang M, Feng L, Feng J. Critical functions of microRNA-30a-5p-E2F3 in cardiomyocyte apoptosis induced by hypoxia/reoxygenation. Kaohsiung J Med Sci 2020; 37:92-100. [PMID: 33058540 DOI: 10.1002/kjm2.12309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 01/01/2023] Open
Abstract
The high-mortality rate of cardiovascular diseases (CVDs) is associated with the myocardial ischemia and reperfusion (I/R). Recent investigations have revealed that microRNAs (miRNAs) exert vital functions in the apoptosis of cardiomyocyte cell. Nevertheless, the potential role of miR-30a-5p in the regulation of cardiomyocyte cell apoptosis needs to be illuminated. In the current study, we observed that hypoxia/reoxygenation (H/R) remarkably raised the level of miR-30a-5p but reduced the expression of E2F transcription factor 3 (E2F3) in H9c2 cardiomyocytes. In vivo, miR-30a-5p was found to be significantly upregulated in the hearts of rats following I/R. Downregulation of miR-30a-5p using anti-miR-30a-5p decreased H9c2 cardiomyocytes apoptosis caused by H/R and promoted the proliferation of H9c2 inhibited by H/R. Moreover, E2F3 was a possible target gene of miR-30a-5p and upregulation of miR-30a-5p reduced the expression level of E2F3 in H9c2 cardiomyocytes. We further identified that E2F3 silencing reversed the effect of anti-miR-30a-5p on the proliferation and apoptosis in H/R treated H9c2 cells. These studies suggested that downregulation of miR-30a-5p attenuated the impact of H/R on H9c2 cardiomyocytes through targeting E2F3.
Collapse
Affiliation(s)
- Xiao-Bing Lv
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing-Hui Niu
- Liver Disease Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Min Zhang
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Feng
- Department of Cardiology, the Central Hospital of Lijin County, Dongying, China
| | - Jia Feng
- Department of Pediatrics, the Central Hospital of Shengli Oil Field, Dongying, China
| |
Collapse
|
31
|
miR-30e-3p Promotes Cardiomyocyte Autophagy and Inhibits Apoptosis via Regulating Egr-1 during Ischemia/Hypoxia. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7231243. [PMID: 32879888 PMCID: PMC7448244 DOI: 10.1155/2020/7231243] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 04/09/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
Background Microvascular obstruction (MVO) can result in coronary microcirculation embolism and myocardial microinfarction. Myocardial injury induced by MVO is characterized by continuous ischemia and hypoxia of cardiomyocytes. Autophagy and apoptosis are closely associated with various cardiovascular diseases. Based on our previous study, we observed a decrease in miR-30e-3p expression and an increase in Egr-1 expression in a rat coronary microembolization model. However, the specific function of miR-30e-3p in regulating autophagy and apoptosis in an ischemia/hypoxia (IH) environment remains to be deciphered. We exposed cardiomyocytes to an IH environment and then determined whether miR-30e-3p was involved in promoting cardiomyocyte autophagy and inhibiting apoptosis by regulating Egr-1. Methods Cardiomyocytes were isolated from rats for our in vitro study. miR-30e-3p was either overexpressed or inhibited by transfection with lentiviral vectors into cardiomyocytes. 3-Methyladenine (3-MA) was used to inhibit autophagy. RT-qPCR and western blotting were used to determine the expression levels of miR-30e-3p, Egr-1, and proteins related to the autophagy and apoptosis process. Autophagic vacuoles and autophagic flux were evaluated using transmission electron microscopy (TEM) and confocal microscopy, respectively. Cardiomyocyte viability was evaluated using the MTS assay. Cell injury was assessed by lactate dehydrogenase (LDH) leakage, and apoptosis was determined by flow cytometry. Results Both miR-30e-3p expression and autophagy were significantly inhibited, and apoptosis was increased in cardiomyocytes after 9 hours of IH exposure. Overexpression of miR-30e-3p increased autophagy and inhibited apoptosis, as well as suppressed Egr-1 expression and decreased cell injury. In addition, inhibition of miR-30e-3p reduced autophagy and increased apoptosis and cell injury. Conclusions miR-30e-3p may be involved in promoting cardiomyocyte autophagy and inhibiting apoptosis by indirectly regulating Egr-1 expression in an IH environment.
Collapse
|
32
|
Zhai C, Li R, Hou K, Chen J, Alzogool M, Hu Y, Zhang J, Zhang Y, Wang L, Zhang R, Cong H. Value of Blood-Based microRNAs in the Diagnosis of Acute Myocardial Infarction: A Systematic Review and Meta-Analysis. Front Physiol 2020; 11:691. [PMID: 32922300 PMCID: PMC7456928 DOI: 10.3389/fphys.2020.00691] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Recent studies have shown that blood-based miRNAs are dysregulated in patients with acute myocardial infarction (AMI) and are therefore a potential tool for the diagnosis of AMI. Therefore, this study summarized and evaluated studies focused on microRNAs as novel biomarkers for the diagnosis of AMI from the last ten years. Methods: MEDLINE, the Cochrane Central database, and EMBASE were searched between January 2010 and December 2019. Studies that assessed the diagnostic accuracy of circulating microRNAs in AMI were chosen. The pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, diagnostic odds ratio, and area under the curve (AUC) were used to assess the test performance of miRNAs. Results: A total of 58 studies that included 8,206 participants assessed the diagnostic accuracy of circulating miRNAs in AMI. The main results of the meta-analyses are as follows: (1) Total miRNAs: the overall pooled sensitivity and specificity were 0.82 (95% CI: 0.79-0.85) and 0.87 (95% CI: 0.84-0.90), respectively. The AUC value was 0.91 (95% CI: 0.88-0.93) in the overall summary receiver operator characteristic (SROC) curve. (2) The panel of two miRNAs: sensitivity: 0.88 (95% CI: 0.77-0.94), specificity: 0.84 (95% CI: 0.72-0.91), AUC: 0.92 (95% CI: 0.90-0.94). (3) The panel of three miRNAs: sensitivity: 0.91 (95% CI: 0.85-0.94), specificity: 0.87 (95% CI: 0.77-0.92), AUC: 0.92 (95% CI: 0.89-0.94). (4) Results by types of miRNAs: miRNA-1: sensitivity: 0.78 (95% CI: 0.71-0.84), specificity: 0.86 (95% CI: 0.77-0.91), AUC: 0.88 (95% CI: 0.85-0.90); miRNA-133a: sensitivity: 0.85 (95% CI: 0.69-0.94), specificity: 0.92 (95% CI: 0.61-0.99), AUC: 0.93 (95% CI: 0.91-0.95); miRNA-208b: sensitivity: 0.80 (95% CI: 0.69-0.88), specificity: 0.96 (95% CI: 0.77-0.99), AUC: 0.91 (95% CI: 0.88-0.93); miRNA-499: sensitivity: 0.85 (95% CI: 0.77-0.91), specificity: 0.95 (95% CI: 0.89-0.98), AUC: 0.96 (95% CI: 0.94-0.97). Conclusion: miRNAs may be used as potential biomarkers for the detection of AMI. For single, stand-alone miRNAs, miRNA-499 may have better diagnostic accuracy compared to other miRNAs. We propose that a panel of multiple miRNAs with high sensitivity and specificity should be tested.
Collapse
Affiliation(s)
- ChuanNan Zhai
- School of Medicine, NanKai University, Tianjin, China.,Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Rui Li
- Tianjin GongAn Hospital, Tianjin, China
| | - Kai Hou
- School of Medicine, NanKai University, Tianjin, China.,Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - JingYi Chen
- School of Medicine, NanKai University, Tianjin, China
| | | | - YueCheng Hu
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - JingXia Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - YingYi Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Le Wang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Rui Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - HongLiang Cong
- School of Medicine, NanKai University, Tianjin, China.,Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
33
|
Liu X, Li S, Yang Y, Sun Y, Yang Q, Gu N, Li J, Huang T, Liu Y, Dong H, Sun S, Fu G, Wu J, Yu B. The lncRNA ANRIL regulates endothelial dysfunction by targeting the let-7b/TGF-βR1 signalling pathway. J Cell Physiol 2020; 236:2058-2069. [PMID: 32783191 DOI: 10.1002/jcp.29993] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 02/04/2023]
Abstract
The long noncoding RNA antisense noncoding RNA in the INK4 locus (ANRIL) plays a critical role in the development of atherosclerosis. However, the precise effect of ANRIL on endothelial dysfunction remains unclear. In this study, we investigated ANRIL expression in patients with coronary artery disease and elucidated the molecular mechanism underlying its effect. ANRIL expression was detected in the blood plasma of 111 patients. We analysed the correlation between ANRIL and endothelial dysfunction markers. We also examined the effect of ANRIL on the regulation of endothelial dysfunction. ANRIL levels were increased in patients with acute coronary syndrome. The expression of ANRIL is associated with the inflammatory cytokines monocyte chemoattractant protein-1 and interleukin-10, which are secreted in response to endothelial dysfunction. Knockdown of ANRIL significantly promoted cell proliferation and tubule formation and inhibited inflammatory activation and apoptosis of human umbilical vein endothelial cells (HUVEC). ANRIL-mediated inhibition of let-7b regulates HUVEC dysfunction by targeting the TGF-βR1/Smad signalling pathway. This study highlights a new therapeutic strategy for preventing endothelial dysfunction associated with cardiovascular disease.
Collapse
Affiliation(s)
- Xianglan Liu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shufeng Li
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Yang
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Sun
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingyuan Yang
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Nan Gu
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Li
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tuo Huang
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Liu
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hui Dong
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Song Sun
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Wu
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bo Yu
- The Key Laboratory of Myocardial Ischaemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
34
|
Xie X, Ji Q, Han X, Zhang L, Li J. Knockdown of long non-coding RNA TTTY15 protects cardiomyocytes from hypoxia-induced injury by regulating let-7b/MAPK6 axis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1951-1961. [PMID: 32922590 PMCID: PMC7476952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/19/2020] [Indexed: 06/11/2023]
Abstract
Acute myocardial infarction (AMI) is a serious threat to human health. Long non-coding RNAs (lncRNAs) are known to be involved in the progression of AMI. The objective of this paper was to explore the functional effect of lncRNA testis-specific transcript Y-linked 15 (TTTY15) on hypoxia-induced cardiomyocyte injury. Human cardiomyocytes AC16 were cultured under hypoxic conditions to induce cardiomyocyte injury. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to check the expression of TTTY15, microRNA let-7b, and Mitogen-activated protein kinase 6 (MAPK6). Western blot was implemented for protein detection. Cell viability and apoptosis were examined by Cell counting kit-8 (CCK-8) assay and flow cytometry, respectively. The target association among TTTY15, let-7b, and MAPK6 was validated by dual-luciferase reporter assay, pull-down assay and RNA immunoprecipitation (RIP) assay. We found that the abundances of TTTY15 and MAPK6 were elevated, while let-7b level declined in hypoxia-induced AC16 cells. Knockdown of TTTY15 increased cell viability, and inhibited apoptosis of hypoxia-induced AC16 cells. TTTY15 bound to and inversely regulated let-7b. Likewise, MAPK6 was a target of let-7b and was negatively regulated by let-7b. Silencing of TTTY15 ameliorated the impact of let-7b downregulation or MAPK6 upregulation on hypoxia-induced cardiomyocyte injury. TTTY15 modulated MAPK6 enrichment by sponging let-7b. In conclusion, knockdown of TTTY15 suppressed hypoxia-induced cardiomyocyte injury through the let-7b/MAPK6 axis.
Collapse
Affiliation(s)
- Xiaofei Xie
- Department of Cardiology, Anhui Chest HospitalHefei, China
| | - Qinjiong Ji
- Department of Cardiology, Anhui Chest HospitalHefei, China
| | - Xiaoliang Han
- Department of Cardiology, Anhui Chest HospitalHefei, China
| | - Ling Zhang
- Department of Cardiology, Anhui Chest HospitalHefei, China
| | - Jingjing Li
- Department of Endocrinology, The 901th Hospital of Joint Logistics Support Force of PLAHefei, Anhui, China
| |
Collapse
|
35
|
Associations of Serum MicroRNA with Bone Mineral Density in Community-Dwelling Subjects: The Yakumo Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5047243. [PMID: 32802851 PMCID: PMC7414326 DOI: 10.1155/2020/5047243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Osteoporosis is a disease characterized by deterioration of bone tissue and mass, with an increasing global prevalence. Therefore, the discovery of biomarkers for osteoporosis would help to guide appropriate treatment. Circulating microRNAs (miRNAs) have become increasingly recognized as biomarkers for detecting diseases. However, few studies have investigated the association of circulating miRNA with osteoporosis in the general population. The aim of this study was to identify miRNA associated with osteoporosis in a general resident health check-up for potential use as an osteoporosis biomarker. We conducted a cross-sectional study as part of a health check-up program and recruited 352 volunteers (139 men, 213 women, mean age 64.1 ± 9.6 years). Osteoporosis was diagnosed according to the WHO classification. Twenty-two candidate microRNAs were screened through real-time quantitative PCR, and miRNAs associated with osteoporosis were analyzed using logistic regression analysis including other risk factors. In total, 95 females and 30 males were diagnosed with osteoporosis with bone mineral density tests (BMD: T‐score < −2.5). We found that miR195 was significantly lower in females, while miR150 and miR222 were significantly higher in males. The results of the logistic regression analysis indicated that in females, higher age and lower miR195 (odds ratio: 0.45, 95% confidential interval: 0.03–0.98) were significant risk factors for lower BMD, while the presence of a smoking habit and lower miR150 (odds ratio: 1.35, 95% confidential interval: 1.02–1.79) were significant risk factors for osteoporosis. Serum levels of miR195 and miR150 are independently associated with low bone mineral density in females and males, respectively.
Collapse
|
36
|
Weighed Gene Coexpression Network Analysis Screens the Potential Long Noncoding RNAs and Genes Associated with Progression of Coronary Artery Disease. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:8183420. [PMID: 32695216 PMCID: PMC7361886 DOI: 10.1155/2020/8183420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/17/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
Background Coronary artery disease (CAD) is a type of heart disease with a high morbidity rate. This study is aimed at identifying potential biomarkers closely related to the progression of CAD. Materials and Methods A microarray dataset of GSE59867 was downloaded from a public database, Gene Expression Omnibus, which included 46 cases of stable CAD without a history of myocardial infarction (MI), 30 cases of MI without heart failure (HF), and 34 cases of MI with HF. Differentially expressed long noncoding RNAs (DElncRNAs) and mRNAs (DEmRNAs) were identified by the limma package, and functions of DEmRNAs were annotated by Gene Ontology and KEGG pathways. In addition, weighed gene coexpression network analysis (WGCNA) was used to construct a coexpression network of DEmRNAs, and a disease-related lncRNAs-mRNAs-pathway network was constructed. Finally, the datasets of GSE61145 and GSE57338 were used to verify the expression levels of the above highly correlated candidates. Results A total of 2362 upregulated mRNAs and 2816 downregulated mRNAs, as well as 235 upregulated lncRNAs and 113 downregulated lncRNAs were screened. These genes were significantly enriched in “cytokine-cytokine receptor interaction,” “RIG-I-like receptor signaling pathway,” and “natural killer cell-mediated cytotoxicity.” Five modules including 1201 DEmRNAs were enriched in WGCNA. A coexpression network including 19 DElncRNAs and 413 DEmRNAs was constructed. These genes were significantly enriched in “phosphatidylinositol signaling system,” “insulin signaling pathway,” and “MAPK signaling pathway”. Disease-related gene-pathway network suggested FASN in “insulin signaling pathway,” DGKZ in “phosphatidylinositol signaling system,” and TNFRSF1A in “MAPK signaling pathway” were involved in MI. Conclusion FASN, DGKZ, and TNFRSF1A were revealed to be CAD progression-associated genes by WGCNA coexpression network analysis.
Collapse
|
37
|
Shi C, Xie H, Ma Y, Yang Z, Zhang J. Nanoscale Technologies in Highly Sensitive Diagnosis of Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 8:531. [PMID: 32582663 PMCID: PMC7289988 DOI: 10.3389/fbioe.2020.00531] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death and morbidity in the world and are a major contributor to healthcare costs. Although enormous progress has been made in diagnosing CVD, there is an urgent need for more efficient early detection and the development of novel diagnostic tools. Currently, CVD diagnosis relies primarily on clinical symptoms based on molecular imaging (MOI) or biomarkers associated with CVDs. However, sensitivity, specificity, and accuracy of the assay are still challenging for early-stage CVDs. Nanomaterial platform has been identified as a promising candidate for improving the practical usage of diagnostic tools because of their unique physicochemical properties. In this review article, we introduced cardiac biomarkers and imaging techniques that are currently used for CVD diagnosis. We presented the applications of various nanotechnologies on diagnosis within cardiac immunoassays (CIAs) and molecular imaging. We also summarized and compared different cardiac immunoassays based on their sensitivities and working ranges of biomarkers.
Collapse
Affiliation(s)
- Chaohong Shi
- Department of Rehabilitation Medicine, The First People’s Hospital of Wenling, Wenzhou Medical University, Wenling, China
| | - Haotian Xie
- Department of Mathematics, The Ohio State University, Columbus, OH, United States
| | - Yifan Ma
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Zhaogang Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jingjing Zhang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
38
|
Identification of a miRNA Based-Signature Associated with Acute Coronary Syndrome: Evidence from the FLORINF Study. J Clin Med 2020; 9:jcm9061674. [PMID: 32492915 PMCID: PMC7356017 DOI: 10.3390/jcm9061674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The discovery of novel biomarkers that improve risk prediction models of acute coronary syndrome (ACS) is needed to better identify and stratify very high-risk patients. MicroRNAs (miRNAs) are essential non-coding modulators of gene expression. Circulating miRNAs recently emerged as important regulators and fine-tuners of physiological and pathological cardiovascular processes; therefore, specific miRNAs expression profiles may represent new risk biomarkers. The aims of the present study were: i) to assess the changes in circulating miRNAs levels associated with ACS and ii) to evaluate the incremental value of adding circulating miRNAs to a clinical predictive risk model. METHODS AND RESULTS The study population included ACS patients (n = 99) and control subjects (n = 103) at high to very high cardiovascular risk but without known coronary event. Based on a miRNA profiling in a matched derivation case (n = -6) control (n = 6) cohort, 21 miRNAs were selected for validation. Comparing ACS cases versus controls, seven miRNAs were significantly differentially expressed. Multivariate logistic regression analyses demonstrated that among the seven miRNAs tested, five were independently associated with the occurrence of ACS. A receiver operating characteristic curve analysis revealed that the addition of miR-122 + miR-150 + miR-195 + miR-16 to the clinical model provided the best performance with an increased area under the curve (AUC) from 0.882 to 0.924 (95% CI 0.885-0.933, p = 0.003). CONCLUSIONS Our study identified a powerful signature of circulating miRNAs providing additive value to traditional risk markers for ACS.
Collapse
|
39
|
Chang L, Zhang W, Shi S, Peng Y, Wang D, Zhang L, Zhang J. microRNA-195 attenuates neuronal apoptosis in rats with ischemic stroke through inhibiting KLF5-mediated activation of the JNK signaling pathway. Mol Med 2020; 26:31. [PMID: 32272873 PMCID: PMC7146986 DOI: 10.1186/s10020-020-00150-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Accumulating evidence has implicated the regulation of microRNAs (miRs) in ischemia stroke. The current study aimed to elucidate the role of microRNA-195 (miR-195) in neuronal apoptosis and brain plasticity in rats with ischemic stroke via the JNK signaling pathway/KLF5 axis. METHODS Ischemic stroke rat models were established by middle cerebral artery occlusion (MCAO), and oxygen deprivation (OGD) models were constructed in rat neuronal cells, followed by gain- or loss-of-function of miR-195 and/or KLF5 in rats and cells. Infarct volume, neuronal loss and ultrastructure, the expression of GAP-43, SYP and KLF5 protein as well as cell apoptosis were determined in the rats. Caspase-3 activity as well as the expression of miR-195, KLF5, GAP-43, SYP, JNK, phosphorylated JNK, Bax and Bcl-2 was measured in the cells. RESULTS The infarct size, expression of GAP-43 and SYP protein and apoptotic cells were increased in the miR-195-/- MCAO rats, while reductions were detected in the miR-195 mimic MCAO and KLF5-/- MCAO rats. Bcl-2 expression was increased, Bax and Caspase-3 expression as well as the ratio of phosphorylated JNK/JNK was decreased in response to miR-195 overexpression or KLF5 knockdown. Interestingly, the silencing of KLF5 reversed the effects exerted by the miR-195 inhibitor on the expression of Bcl-2, phosphorylated JNK/JNK, Bax and Caspase-3. CONCLUSIONS Collectively, our study unraveled that miR-195 could down-regulate KLF5 and block the JNK signaling pathway, ultimately inhibiting neuronal apoptosis in rats with ischemic stroke.
Collapse
Affiliation(s)
- Lisha Chang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Wan Zhang
- Quality Control Office, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, People's Republic of China
| | - Songxin Shi
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Yanbo Peng
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Dali Wang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Li Zhang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Jiang Zhang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China.
| |
Collapse
|
40
|
Noncoding RNAs as Biomarkers for Acute Coronary Syndrome. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3298696. [PMID: 32337239 PMCID: PMC7154975 DOI: 10.1155/2020/3298696] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
Acute coronary syndrome (ACS), consisting of acute myocardial infarction and unstable angina, is the most dangerous and fatal form of coronary heart disease. Acute coronary syndrome has sudden onset and rapid development, which may lead to malignant life-threatening conditions at any time. Therefore, early detection and diagnosis are critical for patients with ACS. Recent studies have found that noncoding RNA is of great significance in the diagnosis and treatment of cardiovascular diseases. In this review, we summarized recent data on circulating noncoding RNAs (including microRNA, long noncoding RNA, and circular RNA) as diagnostic and prognostic markers in ACS including acute myocardial infarction and unstable angina. Specifically, microRNAs (miRNAs) as diagnostic markers are divided into three types: miRNAs of increased expression in ACS, miRNAs of decreased expression in ACS, and miRNAs of contradictory expression in ACS. Moreover, we described these miRNAs of increased expression in ACS based on miRNAs family. This review may result in a great guidance of noncoding RNAs as biomarkers for ACS in clinical practice.
Collapse
|
41
|
Zhao H, Meng L, Xu C, Lin B, Zheng X, Wang J, Feng D. Retracted Article: Long noncoding RNA ANRIL protects cardiomyocytes against hypoxia/reoxygenation injury by sponging miR-195-5p and upregulating Bcl-2. RSC Adv 2019; 9:35624-35635. [PMID: 35702641 PMCID: PMC9097494 DOI: 10.1039/c9ra04898g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/26/2019] [Indexed: 01/04/2023] Open
Abstract
Long noncoding RNAs have been widely accepted to play important roles in acute myocardial infarction (AMI). The dysregulation of cyclin-dependent kinase inhibitor 2B antisense RNA 1 (ANRIL) was discovered in AMI patients. Nevertheless, the detailed role and molecular mechanisms of ANRIL in AMI remain indistinct. The levels of ANRIL, miR-195-5p and Bcl-2 mRNA were determined by qRT-PCR. western blot was performed to assess the expression of Bcl-2, Bax, Cyclin D1 and p21. Cell proliferation was detected by CCK-8 assay, and cell apoptosis was measured by flow cytometry. The targeted correlation between ANRIL and miR-195-5p was confirmed by the dual-luciferase reporter and RNA pull-down assays. Our data revealed that ANRIL was downregulated and miR-195-5p was upregulated in the serum of AMI patients and hypoxia/reoxygenation (H/R)-induced myocardial cells. ANRIL upregulation or miR-195-5p knockdown alleviated H/R-induced myocardial cell injury. Moreover, ANRIL sequestered miR-195-5p by acting as a sponge of miR-195-5p. ANRIL upregulated Bcl-2 expression by sponging miR-195-5p. Additionally, ANRIL overexpression alleviated H/R-induced myocardial cell injury by upregulating Bcl-2. In conclusion, our study indicated that ANRIL upregulation alleviated H/R-induced myocardial cell injury partially through sponging miR-195-5p and upregulating Bcl-2, highlighting its role as a promising mediator for new therapies for AMI treatment.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University No. 1 Longhu Central Road, Jinshui District450052 Zhengzhou China +86-0371-66271808
| | - Li Meng
- Department of Intensive Care Unit, The Henan Provincial Chest Hospital Zhengzhou China
| | - Chengyang Xu
- Department of International Medical Center, The Henan Provincial PeopleS Hospital Zhengzhou China
| | - Bin Lin
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University No. 1 Longhu Central Road, Jinshui District450052 Zhengzhou China +86-0371-66271808
| | - Xiangming Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Jiaxiang Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University No. 1 Longhu Central Road, Jinshui District450052 Zhengzhou China +86-0371-66271808
| | - Deguang Feng
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University No. 1 Longhu Central Road, Jinshui District450052 Zhengzhou China +86-0371-66271808
| |
Collapse
|
42
|
Jaquenod De Giusti C, Santalla M, Das S. Exosomal non-coding RNAs (Exo-ncRNAs) in cardiovascular health. J Mol Cell Cardiol 2019; 137:143-151. [PMID: 31669445 DOI: 10.1016/j.yjmcc.2019.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) play a role in the pathophysiological processes and in different diseases, including cardiovascular disease. Out of several categories of EVs, exosomes (smallest - 30 to 150 nm) are gaining most of the focus as the next generation of biomarkers and in therapeutic strategies. This is because exosomes can be differentiated from other types of EVs based on the expression of tetraspanin molecules on the surface. More importantly, exosomes can be traced back to the cell of origin by identifying the unique cellular marker(s) on the exosomal surface. Recently, several researchs have demonstrated an important and underappreciated mechanism of paracrine cell-cell communication involving exosomal transfer, and its subsequent functional impact on recipient cells. Exosomes are enriched in proteins, mRNAs, miRNAs, and other non-coding RNAs, which can potentially alter myocardial function. Additionally, different stages of tissue damage can also be identified by measuring these bioactive molecules in the circulation. There are several aspects of this new concept still unknown. Therefore, in this review, we have summarized the knowledge we have so far and highlighted the potential of this novel concept of next generation biomarkers and therapeutic intervention.
Collapse
Affiliation(s)
- Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares UNLP-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Manuela Santalla
- Centro de Investigaciones Cardiovasculares UNLP-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina; Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Pergamino, Argentina
| | - Samarjit Das
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
43
|
Soler-Botija C, Gálvez-Montón C, Bayés-Genís A. Epigenetic Biomarkers in Cardiovascular Diseases. Front Genet 2019; 10:950. [PMID: 31649728 PMCID: PMC6795132 DOI: 10.3389/fgene.2019.00950] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are the number one cause of death worldwide and greatly impact quality of life and medical costs. Enormous effort has been made in research to obtain new tools for efficient and quick diagnosis and predicting the prognosis of these diseases. Discoveries of epigenetic mechanisms have related several pathologies, including cardiovascular diseases, to epigenetic dysregulation. This has implications on disease progression and is the basis for new preventive strategies. Advances in methodology and big data analysis have identified novel mechanisms and targets involved in numerous diseases, allowing more individualized epigenetic maps for personalized diagnosis and treatment. This paves the way for what is called pharmacoepigenetics, which predicts the drug response and develops a tailored therapy based on differences in the epigenetic basis of each patient. Similarly, epigenetic biomarkers have emerged as a promising instrument for the consistent diagnosis and prognosis of cardiovascular diseases. Their good accessibility and feasible methods of detection make them suitable for use in clinical practice. However, multicenter studies with a large sample population are required to determine with certainty which epigenetic biomarkers are reliable for clinical routine. Therefore, this review focuses on current discoveries regarding epigenetic biomarkers and its controversy aiming to improve the diagnosis, prognosis, and therapy in cardiovascular patients.
Collapse
Affiliation(s)
- Carolina Soler-Botija
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayés-Genís
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiology Service, HUGTiP, Badalona, Spain
- Department of Medicine, Barcelona Autonomous University (UAB), Badalona, Spain
| |
Collapse
|
44
|
Nonaka CKV, Macêdo CT, Cavalcante BRR, Alcântara ACD, Silva DN, Bezerra MDR, Caria ACI, Tavora FRF, Neto JDDS, Noya-Rabelo MM, Rogatto SR, Ribeiro Dos Santos R, Souza BSDF, Soares MBP. Circulating miRNAs as Potential Biomarkers Associated with Cardiac Remodeling and Fibrosis in Chagas Disease Cardiomyopathy. Int J Mol Sci 2019; 20:E4064. [PMID: 31434314 PMCID: PMC6721092 DOI: 10.3390/ijms20164064] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 12/21/2022] Open
Abstract
Chagas disease (CD) affects approximately 6-7 million people worldwide, from which 30% develop chronic Chagas cardiomyopathy (CCC), usually after being asymptomatic for years. Currently available diagnostic methods are capable of adequately identifying infected patients, but do not provide information regarding the individual risk of developing the most severe form of the disease. The identification of biomarkers that predict the progression from asymptomatic or indeterminate form to CCC, may guide early implementation of pharmacological therapy. Here, six circulating microRNAs (miR-19a-3p, miR-21-5p, miR-29b-3p, miR-30a-5p, miR-199b-5p and miR-208a-3p) were evaluated and compared among patients with CCC (n = 28), CD indeterminate form (n = 10) and healthy controls (n = 10). MiR-19a-3p, miR-21-5p, and miR-29b-3p were differentially expressed in CCC patients when compared to indeterminate form, showing a positive correlation with cardiac dysfunction, functional class, and fibrosis, and a negative correlation with ejection fraction and left ventricular strain. Cardiac tissue analysis confirmed increased expression of microRNAs in CCC patients. In vitro studies using human cells indicated the involvement of these microRNAs in the processes of cardiac hypertrophy and fibrosis. Our study suggests that miRNAs are involved in the process of cardiac fibrosis and remodeling presented in CD and indicate a group of miRNAs as potential biomarkers of disease progression in CCC.
Collapse
Affiliation(s)
- Carolina Kymie Vasques Nonaka
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, 41253-190 Salvador, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), 22281-100 Rio de Janeiro, Brazil
| | - Carolina Thé Macêdo
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil
- Department of Cardiology, São Rafael Hospital, 41253-190 Salvador, Brazil
| | - Bruno Raphael Ribeiro Cavalcante
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, 41253-190 Salvador, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), 22281-100 Rio de Janeiro, Brazil
| | | | - Daniela Nascimento Silva
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, 41253-190 Salvador, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil
| | | | - Alex Cleber Improta Caria
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, 41253-190 Salvador, Brazil
- Federal University of Bahia, UFBA, 40231-300 Salvador, Brazil
| | | | | | | | - Silvia Regina Rogatto
- Department of Clinical Genetics, Vejle Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Ricardo Ribeiro Dos Santos
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil
- National Institute of Science and Technology for Regenerative Medicine, 21941-902 Rio de Janeiro, Brazil
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell Therapy, Hospital São Rafael, 41253-190 Salvador, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), 22281-100 Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, 21941-902 Rio de Janeiro, Brazil
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, FIOCRUZ, 40296-710 Salvador, Brazil.
- National Institute of Science and Technology for Regenerative Medicine, 21941-902 Rio de Janeiro, Brazil.
| |
Collapse
|
45
|
Hawkins PG, Sun Y, Dess RT, Jackson WC, Sun G, Bi N, Tewari M, Hayman JA, Kalemkerian GP, Gadgeel SM, Lawrence TS, Haken RKT, Matuszak MM, Kong FMS, Schipper MJ, Jolly S. Circulating microRNAs as biomarkers of radiation-induced cardiac toxicity in non-small-cell lung cancer. J Cancer Res Clin Oncol 2019; 145:1635-1643. [PMID: 30923943 DOI: 10.1007/s00432-019-02903-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/22/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Radiation-induced cardiac toxicity (RICT) is an increasingly well-appreciated source of morbidity and mortality in patients receiving thoracic radiotherapy (RT). Currently available methods to predict RICT are suboptimal. We investigated circulating microRNAs (c-miRNAs) as potential biomarkers of RICT in patients undergoing definitive RT for non-small-cell lung cancer (NSCLC). METHODS Data from 63 patients treated on institutional trials were analyzed. Prognostic models of grade 3 or greater (G3 +) RICT based on pre-treatment c-miRNA levels ('c-miRNA'), mean heart dose (MHD) and pre-existing cardiac disease (PCD) ('clinical'), and a combination of these ('c-miRNA + clinical') were developed. Elastic net Cox regression and full cross validation were used for variable selection, model building, and model evaluation. Concordance statistic (c-index) and integrated Brier score (IBS) were used to evaluate model performance. RESULTS MHD, PCD, and serum levels of 14 c-miRNA species were identified as jointly prognostic for G3 + RICT. The 'c-miRNA and 'clinical' models yielded similar cross-validated c-indices (0.70 and 0.72, respectively) and IBSs (0.26 and 0.28, respectively). However, prognostication was not improved by combining c-miRNA and clinical factors (c-index 0.70, IBS 0.28). The 'c-miRNA' and 'clinical' models were able to significantly stratify patients into high- and low-risk groups of developing G3 + RICT. Chi-square testing demonstrated a marginally significantly higher prevalence of PCD in patients with high- compared to low-risk c-miRNA profile (p = 0.09), suggesting an association between some c-miRNAs and PCD. CONCLUSIONS We identified a pre-treatment c-miRNA signature prognostic for G3 + RICT. With further development, pre- and mid-treatment c-miRNA profiling could contribute to patient-specific dose selection and treatment adaptation.
Collapse
Affiliation(s)
- Peter G Hawkins
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Yilun Sun
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Robert T Dess
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - William C Jackson
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Grace Sun
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Nan Bi
- Department of Radiation Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Muneesh Tewari
- Department of Biomedical Engineering, Biointerfaces Institute, and Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - James A Hayman
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Gregory P Kalemkerian
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shirish M Gadgeel
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Randall K Ten Haken
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Martha M Matuszak
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
| | - Feng-Ming Spring Kong
- Department of Radiation Oncology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matthew J Schipper
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Shruti Jolly
- Department of Radiation Oncology, University of Michigan, 1500 E Medical Center Drive, UH B2 C490 SPC 5010, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
46
|
Wang J, Bie Z, Sun C. Long noncoding RNA AK088388 regulates autophagy through miR‐30a to affect cardiomyocyte injury. J Cell Biochem 2019; 120:10155-10163. [PMID: 30635942 DOI: 10.1002/jcb.28300] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/28/2018] [Indexed: 01/30/2023]
Affiliation(s)
- Jing‐jing Wang
- Department of Medicine Xi’an Jiaotong University Xi’an Shanxi China
- Department of Cardiology General Hospital of Ningxia Medical University Yinchuan Ningxia China
| | - Zi‐dong Bie
- Department of Cardiology Yantai Yuhuangding Hospital Yantai Shandong China
| | - Chao‐feng Sun
- Department of Cardiology First Affiliated Hospital of Xi’an Jiaotong University Xi’an Shanxi China
| |
Collapse
|
47
|
Cheng HY, Wang YS, Hsu PY, Chen CY, Liao YC, Juo SHH. miR-195 Has a Potential to Treat Ischemic and Hemorrhagic Stroke through Neurovascular Protection and Neurogenesis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 13:121-132. [PMID: 30775405 PMCID: PMC6365409 DOI: 10.1016/j.omtm.2018.11.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023]
Abstract
Tissue plasminogen activator is the only U.S. FDA-approved therapy for ischemic stroke, while there is no specific medication for hemorrhagic stroke. Therefore, the treatment of acute stroke continues to be a major unmet clinical need. We explored the effects of miR-195 on neurovascular protection and its potential in treating acute stroke. Using both cellular and animal studies, we showed that miR-195’s beneficial effects are mediated by four mechanisms: (1) anti-apoptosis for injured neural cells by directly suppressing Sema3A/Cdc42/JNK signaling, (2) neural regeneration by promoting neural stem cell proliferation and migration, (3) anti-inflammation by directly blocking the NF-kB pathway, and (4) improvement of endothelial functions. We intravenously injected miR-195 carried by nanoparticles into rats with either ischemic or hemorrhagic stroke in the acute stage. The results showed that miR-195 reduced the size of brain damage and improved functional recovery in both types of stroke rats. The reduction of injured brain volume could be up to 45% in ischemic stroke and approximately 30% in hemorrhagic stroke. The therapeutic window between stroke onset and miR-195 treatment could be up to 6 h. Our data demonstrated that miR-195 possesses the potential to become a new drug to treat acute ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Hsin-Yun Cheng
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan
| | - Yung-Song Wang
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan.,Institute of Fisheries Science, National Taiwan University, 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan.,Department of Life Science, National Taiwan University, 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Po-Yuan Hsu
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan.,Department of Medical Research, China Medical University Hospital, 2 Yude Road, Taichung, 40447, Taiwan
| | - Chien-Yuan Chen
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan.,Department of Medical Research, China Medical University Hospital, 2 Yude Road, Taichung, 40447, Taiwan
| | - Yi-Chu Liao
- Department of Neurology, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Department of Neurology, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Road, Taipei 112, Taiwan
| | - Suh-Hang H Juo
- Department of Medical Research, China Medical University Hospital, 2 Yude Road, Taichung, 40447, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.,Institute of New Drug Development, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.,Drug Development Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
48
|
Xing J, Xie T, Tan W, Li R, Yu C, Han X. microRNA‐183 improve myocardial damager via NF‐kb pathway: In vitro and in vivo study. J Cell Biochem 2018; 120:10145-10154. [PMID: 30548682 DOI: 10.1002/jcb.28298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 11/20/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Jie Xing
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Ting Xie
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Wei Tan
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Ruzheng Li
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Cheng Yu
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| | - Xiaohu Han
- Department of Cardiac surgery Hainan General Hospital Haikou Hainan China
| |
Collapse
|
49
|
Ahmed FW, Bakhashab S, Bastaman IT, Crossland RE, Glanville M, Weaver JU. Anti-Angiogenic miR-222, miR-195, and miR-21a Plasma Levels in T1DM Are Improved by Metformin Therapy, Thus Elucidating Its Cardioprotective Effect: The MERIT Study. Int J Mol Sci 2018; 19:ijms19103242. [PMID: 30347712 PMCID: PMC6214022 DOI: 10.3390/ijms19103242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1DM) is associated with increased cardiovascular disease (CVD) and reduced life expectancy. We thus hypothesized that anti-angiogenic miRs are increased in T1DM, and the cardioprotective effect of metformin is mediated via reducing those miRs. In an open label, case-controlled study, 23 T1DM patients without CVD were treated with metformin for eight weeks (TG), matched with nine T1DM patients on standard treatment (SG) and 23 controls (CG). Plasma miR-222, miR-195, miR-21a and miR-126 were assayed by real-time RT-qPCR. The results were correlated with: endothelial function (RHI), circulating endothelial progenitor cells (cEPCs) (vascular repair marker, CD45dimCD34+VEGFR2+ cells) and circulating endothelial cells (cECs) (vascular injury marker, CD45dimCD34+CD133-CD144+ cells). miR-222, miR-195 and miR-21a were higher in T1DM than CG; p = 0.009, p < 0.0001, p = 0.0001, respectively. There was an inverse correlation between logmiR-222 and logRHI (p < 0.05) and a direct correlation between logmiR-222 and logCD34+ (p < 0.05) in TG. Metformin reduced miR-222, miR-195 and miR-21a levels in TG; p = 0.007, p = 0.002 p = 0.0012, respectively. miRs remained unchanged in SG. miR-126 was similar in all groups. There was a positive association between changes in logmiR-222 and logcECs after metformin in TG (p < 0.05). Anti-angiogenic miRs are increased in T1DM. Metformin has cardioprotective effects through downregulating miR-222, miR-195 and miR-21a, beyond improving glycemic control.
Collapse
Affiliation(s)
- Fahad W Ahmed
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle Upon Tyne NE9 6SH, UK.
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Department of Diabetes and Endocrinology, Royal Sussex County Hospital, Brighton BN2 5BE, UK.
| | - Sherin Bakhashab
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah P.O. Box 80218, Saudi Arabia.
- Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah P.O. Box 80216, Saudi Arabia.
| | - Inda T Bastaman
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle Upon Tyne NE9 6SH, UK.
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia.
| | - Rachel E Crossland
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
| | - Michael Glanville
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
| | - Jolanta U Weaver
- Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle Upon Tyne NE9 6SH, UK.
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
- Cardiovascular Research Centre, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK.
| |
Collapse
|
50
|
Gao J, Yang S, Wang K, Zhong Q, Ma A, Pan X. Plasma miR-126 and miR-143 as Potential Novel Biomarkers for Cerebral Atherosclerosis. J Stroke Cerebrovasc Dis 2018; 28:38-43. [PMID: 30309729 DOI: 10.1016/j.jstrokecerebrovasdis.2018.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 08/24/2018] [Accepted: 09/02/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cerebral atherosclerosis is the most important mechanism for ischemic stroke. However, specific plasma biomarkers to assess atherosclerosis susceptibility are still lacking. Circulating miRNAs have been shown to be promising biomarkers for various pathologic conditions. We investigated whether plasma miR-126 and miR-143 could be used as biomarkers for identifying and evaluating cerebral atherosclerosis. Results showed that miR-143 and miR-126 might participate in the process of atherosclerosis and were minimally affected by cerebral infarction. Using Pearson correlation analysis, we showed that miR-126 and miR-143 were correlated with the presence and severity of cerebral atherosclerosis. The ability of miR-126 and miR-143 to differentiate atherosclerosis patients from healthy controls was demonstrated via a receiving operating characteristic curve with high specificity and sensitivity. Our data thus indicate that miR-126 and miR-143 may be potential specific biomarkers for atherosclerosis.
Collapse
Affiliation(s)
- Jing Gao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Shuna Yang
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Kun Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianwei Zhong
- Department of Neurology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Aijun Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|