1
|
Feng P, Yang F, Zang D, Bai D, Xu L, Fu Y, You R, Liu T, Yang X. Deciphering the roles of cellular and extracellular non-coding RNAs in chemotherapy-induced cardiotoxicity. Mol Cell Biochem 2024:10.1007/s11010-024-05143-5. [PMID: 39485641 DOI: 10.1007/s11010-024-05143-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Chemotherapy-induced cardiotoxicity is a major adverse effect, driven by multiple factors in its pathogenesis. Notably, RNAs have emerged as significant contributors in both cancer and heart failure (HF). RNAs carry genetic and metabolic information that mirrors the current state of cells, making them valuable as potential biomarkers and therapeutic tools for diagnosing, predicting, and treating a range of diseases, including cardiotoxicity. Over 97% of the genome is transcribed into non-coding RNAs (ncRNAs), including ribosomal RNA (rRNAs), transfer RNAs (tRNAs), and newly identified microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs). NcRNAs function not only within their originating cells but also in recipient cells by being transported through extracellular compartments, referred to as extracellular RNAs (exRNAs). Since ncRNAs were identified as key regulators of gene expression, numerous studies have highlighted their significance in both cancer and cardiovascular diseases. Nevertheless, the role of ncRNAs in cardiotoxicity remains not fully elucidated. The study aims to review the existing knowledge on ncRNAs in Cardio-Oncology and explore the potential of ncRNA-based biomarkers and therapies. These investigations could advance the clinical application of ncRNA research, improving early detection and mitigating of chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Pan Feng
- Baoji Hospital of Traditional Chinese Medicine, Baoji, 721000, China
| | - Fan Yang
- Guang'an Men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Dongmei Zang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Dapeng Bai
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Liyan Xu
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Yueyun Fu
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Ranran You
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Tao Liu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710068, China.
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China.
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
2
|
Qin T, Sun M, Huang Y, Guo J, Hong A, Zheng Q, Wei T, He Q, Ren Z. MiR-221 on protective oxidative induced by selenium modified Codonopsis pilosula polysaccharide. Int J Biol Macromol 2024; 279:134815. [PMID: 39154690 DOI: 10.1016/j.ijbiomac.2024.134815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Oxidative stress plays an important role in various diseases. miR-221 has been reported to regulate oxidative stress. However, the mechanism of miR-221 in regulating oxidative stress induced by sCPPS5 remains unclear. This study aimed to investigate the protective effects and mechanisms of miR-221 on oxidative stress induced by sCPPS5. The expression of SOD, CAT, MDA, LDH, MMP, caspase-3 activity and apoptosis were measured. In addition, the key signaling factors in the Keap1-Nrf2-ARE signaling pathway were determined by real-time PCR and Western blot. Mice were employed to evaluate the effects of sCPPS5 and the possible mechanism in vivo. sCPPS5 promoted the expression of SOD and CAT and activated Keap1-Nrf2-ARE signaling pathway inhibit the MDA content, MMP, caspase-3 activity, apoptosis and LDH release rate after transfection with miR-221 mimics and inhibitors. Consistently, sCPPS5 has the potential to enhance the expression of antioxidant enzymes as well as upregulate mRNA expression of crucial signal proteins in vivo. miR-221 on oxidative stress protection induced by sCPPS5 possibly through regulating the Keap1-Nrf2-ARE signaling pathway in macrophages.
Collapse
Affiliation(s)
- Tao Qin
- Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health in Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Mengke Sun
- Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health in Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Yongyuan Huang
- Fujian Key Laboratory of Chinese Traditional and Western Veterinary Medicine and Animal Health, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Jinhang Guo
- Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health in Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Ancan Hong
- Fujian Key Laboratory of Chinese Traditional and Western Veterinary Medicine and Animal Health, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Qiang Zheng
- Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health in Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Tiantian Wei
- Fujian Key Laboratory of Chinese Traditional and Western Veterinary Medicine and Animal Health, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Qiuyue He
- Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health in Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China
| | - Zhe Ren
- Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health in Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China.
| |
Collapse
|
3
|
Kura B, Pavelkova P, Kalocayova B, Pobijakova M, Slezak J. MicroRNAs as Regulators of Radiation-Induced Oxidative Stress. Curr Issues Mol Biol 2024; 46:7097-7113. [PMID: 39057064 PMCID: PMC11276491 DOI: 10.3390/cimb46070423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
microRNAs (miRNAs) represent small RNA molecules involved in the regulation of gene expression. They are implicated in the regulation of diverse cellular processes ranging from cellular homeostasis to stress responses. Unintended irradiation of the cells and tissues, e.g., during medical uses, induces various pathological conditions, including oxidative stress. miRNAs may regulate the expression of transcription factors (e.g., nuclear factor erythroid 2 related factor 2 (Nrf2), nuclear factor kappa B (NF-κB), tumor suppressor protein p53) and other redox-sensitive genes (e.g., mitogen-activated protein kinase (MAPKs), sirtuins (SIRTs)), which trigger and modulate cellular redox signaling. During irradiation, miRNAs mainly act with reactive oxygen species (ROS) to regulate the cell fate. Depending on the pathway involved and the extent of oxidative stress, this may lead to cell survival or cell death. In the context of radiation-induced oxidative stress, miRNA-21 and miRNA-34a are among the best-studied miRNAs. miRNA-21 has been shown to directly target superoxide dismutase (SOD), or NF-κB, whereas miRNA-34a is a direct regulator of NADPH oxidase (NOX), SIRT1, or p53. Understanding the mechanisms underlying radiation-induced injury including the involvement of redox-responsive miRNAs may help to develop novel approaches for modulating the cellular response to radiation exposure.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| | - Patricia Pavelkova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia
| | - Margita Pobijakova
- Department of Radiation Oncology, Bory Hospital–Penta Hospitals, 841 03 Bratislava, Slovakia;
- Radiological Science, Faculty of Nursing and Medical Professional Studies, Slovak Medical University, 831 01 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| |
Collapse
|
4
|
Zhao MM, Wang B, Huang WX, Zhang L, Peng R, Wang C. Verteporfin suppressed mitophagy via PINK1/parkin pathway in endometrial cancer. Am J Cancer Res 2024; 14:1935-1946. [PMID: 38726279 PMCID: PMC11076261 DOI: 10.62347/pmyv3832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 03/15/2024] [Indexed: 05/12/2024] Open
Abstract
Endometrial cancer (EC) is a malignancy that poses a threat to woman's health worldwide. Building upon prior work, we explored the inhibitory effect of verteporfin on EC. We showed that verteporfin can damage the mitochondria of EC cells, leading to a decrease of mitochondrial membrane potential and an increase in ROS (reactive oxygen species). In addition, verteporfin treatment was shown to inhibit the proliferation and migration of EC cells, promote apoptosis, and reduce the expression of mitophagy-related proteins PINK1/parkin and TOM20. The ROS inhibitor N-Acetyl Cysteine was able to rescue the expression of PINK1/parkin proteins. This suggests that verteporfin may inhibit mitophagy by elevating ROS levels, thereby inhibiting EC cell viability. The effect of verteporfin on mitophagy supports further investigation as a potential therapeutic option for EC.
Collapse
Affiliation(s)
- Ming-Ming Zhao
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Bo Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Wen-Xi Huang
- School of Basic Medical Sciences, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Li Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Rui Peng
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| | - Chao Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai 200011, China
| |
Collapse
|
5
|
Agwa SHA, Elzahwy SS, Hossam N, Yahia YA, Hamady S, Sherif N, Elshazly A, Darwish RM, Hashim JO, Adly MA, Abd Elsamee AM, Shamekh R, Roushdy MMS, Matboli M. Discriminatory power of a circulating multi-noncoding RNA panel in acute coronary syndrome subtypes: Towards precision detection. Int J Biochem Cell Biol 2024; 169:106531. [PMID: 38280541 DOI: 10.1016/j.biocel.2024.106531] [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: 06/22/2023] [Revised: 11/24/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Acute Coronary Syndrome (ACS) stands as a significant contributor to cardiovascular mortality, necessitating improved diagnostic tools for early detection and tailored therapeutic interventions. Current diagnostic modalities, exhibit limitations in sensitivity and specificity, urging the quest for novel biomarkers to enhance discrimination of the different stages of ACS including unstable angina, Non-ST-segment Elevation Myocardial Infarction (NSTEMI), and ST-segment Elevation Myocardial Infarction (STEMI). METHODS This study investigated the potential of a plasma-circulating multi-noncoding RNA (ncRNA) panel, comprising four miRNAs (miR-182-5p, miR-23a-3p, miR-146a-5p, and miR-183-5p) and three lncRNAs (SNHG15, SNHG5, and RMRP), selected based on their intricate involvement in ACS pathogenesis and signaling pathways regulating post-myocardial infarction (MI) processes. The differential expression of these ncRNAs was validated in sera of ACS patients and healthy controls via real-time polymerase chain reaction (RT-PCR). RESULTS Analysis revealed a marked upregulation of the multi-ncRNAs panel in ACS patients. Notably, miRNA-182-5p and lncRNA-RMRP exhibited exceptional discriminatory power, indicated by the high area under the curve (AUC) values (0.990 and 0.980, respectively). Importantly, this panel displayed superior efficacy in discriminating between STEMI and NSTEMI, outperforming conventional biomarkers like creatine kinase-MB and cardiac troponins. Additionally, the four miRNAs and lncRNA RMRP showcased remarkable proficiency in distinguishing between STEMI and unstable angina. CONCLUSION The findings underscore the promising potential of the multi-ncRNA panel as a robust tool for early ACS detection, and precise differentiation among ACS subtypes, and as a potential therapeutic target.
Collapse
Affiliation(s)
- Sara H A Agwa
- Clinical pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt.
| | - Sherif Samir Elzahwy
- Cardiovascular Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 1382, Egypt
| | - Nourhan Hossam
- Medicinal Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Yahia A Yahia
- Biochemistry Department, Faculty of Pharmacy, Misr University for Science and Technology, Giza 12566, Egypt
| | - Shaimaa Hamady
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Nadine Sherif
- Clinical pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Ahmed Elshazly
- Cardiovascular Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 1382, Egypt
| | - Reham M Darwish
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Jomana Osama Hashim
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Mahmoud Ashraf Adly
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Aya M Abd Elsamee
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Rania Shamekh
- Department of Pathology, University of South Florida, Tampa, FL 33620, USA
| | - Marian Maher Salib Roushdy
- Medicinal Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Marwa Matboli
- Medicinal Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt.
| |
Collapse
|
6
|
Zheng H, Xu Y, Liehn EA, Rusu M. Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction. Int J Mol Sci 2024; 25:3114. [PMID: 38542087 PMCID: PMC10970003 DOI: 10.3390/ijms25063114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/31/2024] [Accepted: 02/10/2024] [Indexed: 06/26/2024] Open
Abstract
Currently, coronary artery bypass and reperfusion therapies are considered the gold standard in long-term treatments to restore heart function after acute myocardial infarction. As a drawback of these restoring strategies, reperfusion after an ischemic insult and sudden oxygen exposure lead to the exacerbated synthesis of additional reactive oxidative species and the persistence of increased oxidation levels. Attempts based on antioxidant treatment have failed to achieve an effective therapy for cardiovascular disease patients. The controversial use of vitamin C as an antioxidant in clinical practice is comprehensively systematized and discussed in this review. The dose-dependent adsorption and release kinetics mechanism of vitamin C is complex; however, this review may provide a holistic perspective on its potential as a preventive supplement and/or for combined precise and targeted therapeutics in cardiovascular management therapy.
Collapse
Affiliation(s)
- Huabo Zheng
- Department of Cardiology, Angiology and Intensive Care, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany;
- Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark;
| | - Yichen Xu
- Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark;
- Department of Histology and Embryology, Medicine and Life Sciences, Hainan Medical University, Haikou 571199, China
| | - Elisa A. Liehn
- Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark;
- National Institute of Pathology “Victor Babes”, Splaiul Independentei Nr. 99-101, 050096 Bucharest, Romania
| | - Mihaela Rusu
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany
| |
Collapse
|
7
|
Wang S, Sun Y, Yao L, Xing Y, Yang H, Ma Q. The Role of microRNA-23a-3p in the Progression of Human Aging Process by Targeting FOXO3a. Mol Biotechnol 2024; 66:277-287. [PMID: 37087718 PMCID: PMC10803409 DOI: 10.1007/s12033-023-00746-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/02/2023] [Indexed: 04/24/2023]
Abstract
Aging results in deterioration of body functions and, ultimately, death. miRNAs contribute to the regulation of aging. The aim of this study was to explore the contribution of miRNAs to aging and senescence-related changes in gene expression. The expression changes of miRNAs in the blood of people and animal samples collected from different age subjects were examined using Affymetrix miRNA 4.0 microarray and qRT-PCR. MTT assay and flow cytometry were used to examine the effect of miR-23a on cell functions in WI-38 cells. The expression levels of 48 miRNAs, including miR-23a, miR-21, and miR-100, in the blood samples were higher in the middle-aged group than in the young or elderly group. Animal studies further suggested that the expression of miR-23a increased with age. In addition, upregulation of miR-23a dramatically suppressed the cell proliferation and arrested the WI-38 cell cycle in vitro. FOXO3a has been identified as a target gene of miR-23a. MiR-23a downregulated the expression of FOXO3a in WI-38 cells. MiRNAs have different expression levels in different age groups. miR-23a could suppress cell proliferation and arrest the cell cycle in WI-38 cells, which elucidated the mechanism through which miR-23a exerts pivotal role in WI-38 cells by targeting FOXO3a.
Collapse
Affiliation(s)
- Shan Wang
- Department of Geriatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Sun
- Department of Geriatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lan Yao
- Department of Geriatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yunli Xing
- Department of Geriatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Huayu Yang
- Department of Geriatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Qing Ma
- Department of Geriatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
8
|
Hyttinen JMT, Blasiak J, Kaarniranta K. Non-Coding RNAs Regulating Mitochondrial Functions and the Oxidative Stress Response as Putative Targets against Age-Related Macular Degeneration (AMD). Int J Mol Sci 2023; 24:ijms24032636. [PMID: 36768958 PMCID: PMC9917342 DOI: 10.3390/ijms24032636] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Age-related macular degeneration (AMD) is an ever-increasing, insidious disease which reduces the quality of life of millions of elderly people around the world. AMD is characterised by damage to the retinal pigment epithelium (RPE) in the macula region of the retina. The origins of this multi-factorial disease are complex and still not fully understood. Oxidative stress and mitochondrial imbalance in the RPE are believed to be important factors in the development of AMD. In this review, the regulation of the mitochondrial function and antioxidant stress response by non-coding RNAs (ncRNAs), newly emerged epigenetic factors, is discussed. These molecules include microRNAs, long non-coding RNAs, and circular non-coding RNAs. They act mainly as mRNA suppressors, controllers of other ncRNAs, or by interacting with proteins. We include here examples of these RNA molecules which affect various mitochondrial processes and antioxidant signaling of the cell. As a future prospect, the possibility to manipulate these ncRNAs to strengthen mitochondrial and antioxidant response functions is discussed. Non-coding RNAs could be used as potential diagnostic markers for AMD, and in the future, also as therapeutic targets, either by suppressing or increasing their expression. In addition to AMD, it is possible that non-coding RNAs could be regulators in other oxidative stress-related degenerative diseases.
Collapse
Affiliation(s)
- Juha M. T. Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- Correspondence:
| | - Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029 Kuopio, Finland
| |
Collapse
|
9
|
Sigutova R, Evin L, Stejskal D, Ploticova V, Svagera Z. Specific microRNAs and heart failure: time for the next step toward application? Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2022; 166:359-368. [PMID: 35726831 DOI: 10.5507/bp.2022.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
A number of microRNAs are involved in the pathophysiological events associated with heart disease. In this review, we discuss miR-21, miR-1, miR-23a, miR-142-5p, miR-126, miR-29, miR-195, and miR-499 because they are most often mentioned as important specific indicators of myocardial hypertrophy and fibrosis leading to heart failure. The clinical use of microRNAs as biomarkers and for therapeutic interventions in cardiovascular diseases appears highly promising. However, there remain many unresolved details regarding their specific actions in distinct pathological phenomena. The introduction of microRNAs into routine practice, as part of the cardiovascular examination panel, will require additional clinically relevant and reliable data. Thus, there remains a need for additional research in this area, as well as the optimization and standardization of laboratory procedures which could significantly shorten the determination time, and make microRNA analysis simpler and more affordable. In this review, we aim to summarize the current knowledge about selected microRNAs related to heart failure, including their potential use in diagnosis, prognosis, and treatment, and options for their laboratory determination.
Collapse
Affiliation(s)
- Radka Sigutova
- Institute of Laboratory Medicine, Department of Clinical Biochemistry, University Hospital Ostrava and Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.,Department of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Lukas Evin
- Department of Internal Medicine and Cardiology, Department of Cardiovascular, University Hospital Ostrava, Ostrava, Czech Republic
| | - David Stejskal
- Institute of Laboratory Medicine, Department of Clinical Biochemistry, University Hospital Ostrava and Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Vera Ploticova
- Institute of Laboratory Medicine, Department of Clinical Biochemistry, University Hospital Ostrava and Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Zdenek Svagera
- Institute of Laboratory Medicine, Department of Clinical Biochemistry, University Hospital Ostrava and Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
10
|
Cardiovascular Disease-Associated MicroRNAs as Novel Biomarkers of First-Trimester Screening for Gestational Diabetes Mellitus in the Absence of Other Pregnancy-Related Complications. Int J Mol Sci 2022; 23:ijms231810635. [PMID: 36142536 PMCID: PMC9501303 DOI: 10.3390/ijms231810635] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
We assessed the diagnostic potential of cardiovascular disease-associated microRNAs for the early prediction of gestational diabetes mellitus (GDM) in singleton pregnancies of Caucasian descent in the absence of other pregnancy-related complications. Whole peripheral venous blood samples were collected within 10 to 13 weeks of gestation. This retrospective study involved all pregnancies diagnosed with only GDM (n = 121) and 80 normal term pregnancies selected with regard to equality of sample storage time. Gene expression of 29 microRNAs was assessed using real-time RT-PCR. Upregulation of 11 microRNAs (miR-1-3p, miR-20a-5p, miR-20b-5p, miR-23a-3p, miR-100-5p, miR-125b-5p, miR-126-3p, miR-181a-5p, miR-195-5p, miR-499a-5p, and miR-574-3p) was observed in pregnancies destinated to develop GDM. Combined screening of all 11 dysregulated microRNAs showed the highest accuracy for the early identification of pregnancies destinated to develop GDM. This screening identified 47.93% of GDM pregnancies at a 10.0% false positive rate (FPR). The predictive model for GDM based on aberrant microRNA expression profile was further improved via the implementation of clinical characteristics (maternal age and BMI at early stages of gestation and an infertility treatment by assisted reproductive technology). Following this, 69.17% of GDM pregnancies were identified at a 10.0% FPR. The effective prediction model specifically for severe GDM requiring administration of therapy involved using a combination of these three clinical characteristics and three microRNA biomarkers (miR-20a-5p, miR-20b-5p, and miR-195-5p). This model identified 78.95% of cases at a 10.0% FPR. The effective prediction model for GDM managed by diet only required the involvement of these three clinical characteristics and eight microRNA biomarkers (miR-1-3p, miR-20a-5p, miR-20b-5p, miR-100-5p, miR-125b-5p, miR-195-5p, miR-499a-5p, and miR-574-3p). With this, the model identified 50.50% of GDM pregnancies managed by diet only at a 10.0% FPR. When other clinical variables such as history of miscarriage, the presence of trombophilic gene mutations, positive first-trimester screening for preeclampsia and/or fetal growth restriction by the Fetal Medicine Foundation algorithm, and family history of diabetes mellitus in first-degree relatives were included in the GDM prediction model, the predictive power was further increased at a 10.0% FPR (72.50% GDM in total, 89.47% GDM requiring therapy, and 56.44% GDM managed by diet only). Cardiovascular disease-associated microRNAs represent promising early biomarkers to be implemented into routine first-trimester screening programs with a very good predictive potential for GDM.
Collapse
|
11
|
Lei S, Feng Y, Huang P, Chen B, Bao K, Wu Q, Zhang H, Huang X. Ophiopogonin D'-induced mitophagy and mitochondrial damage are associated with dysregulation of the PINK1/Parkin signaling pathway in AC16 cells. Toxicology 2022; 477:153275. [PMID: 35905946 DOI: 10.1016/j.tox.2022.153275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Abstract
Shenmai injection (SMI) is a patented traditional Chinese medicine that is extracted from Panax ginseng and Ophiopogon japonicus and is commonly used to treat cardiovascular diseases and tumors. The O. japonicus extract Ophiopogonin D' (OPD') is highly cardiotoxic. Mitochondria are central to OPD'-induced cardiotoxicity, although the precise mechanisms remain unclear. Excessive mitophagy activation and mitochondrial dysfunction lead to apoptosis, and the PTEN-induced kinase 1(PINK1)/Parkin pathway is critical in regulating mitophagy and mitochondrial function. We investigated the role of the PINK1/Parkin pathway in OPD'-induced mitochondrial damage and cardiotoxicity in AC16 cells. Concentrations of 2μM OPD' and above inhibited cardiomyocyte viability and increased lactate dehydrogenase (LDH) release in a concentration- and time-dependent manner. OPD' was toxic to cells and mitochondria and increased the rate of apoptosis, triggering pyknosis, decreasing mitochondrial membrane potential (MMP), and decreasing the protein expression of the biogenesis regulator peroxisome proliferator-activated receptor γ coactivator-1 alpha (PGC-1α). The increased ratio of microtubule-associated proteins 1A/1B light chain 3B (LC3-II/LC3-I) in mitochondria indicated that OPD' induced mitophagy. OPD' significantly induced oxidative stress and apoptosis, including increased reactive oxygen species (ROS) generation and decreased nuclear factor erythroid-2 related factor 2 (Nrf2), heme oxygenase-1(HO-1), and B-cell lymphoma 2 (Bcl-2) protein expression. OPD' activated the PINK1/Parkin pathway and promoted PINK1/Parkin translocation to mitochondria. Inhibiting mitophagy attenuated OPD'-induced PINK1/Parkin pathway activation and preserved mitochondrial biogenesis, consequently mitigating OPD'-induced mitochondrial dysfunction and apoptosis. These findings suggest that OPD'-induced cardiomyocyte mitophagy and mitochondrial damage are at least partially mediated by dysregulation of the PINK1/Parkin pathway.
Collapse
Affiliation(s)
- Sisi Lei
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Yuchao Feng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Peiying Huang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China
| | - BoJun Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Kun Bao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Qihua Wu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Haobo Zhang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Xiaoyan Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
12
|
Reily-Bell M, Bahn A, Katare R. Reactive Oxygen Species-Mediated Diabetic Heart Disease: Mechanisms and Therapies. Antioxid Redox Signal 2022; 36:608-630. [PMID: 34011169 DOI: 10.1089/ars.2021.0098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Significance: Diabetic heart disease (DHD) is the primary cause of mortality in people with diabetes. A significant contributor to the development of DHD is the disruption of redox balance due to reactive oxygen species (ROS) overproduction resulting from sustained high glucose levels. Therapies specifically focusing on the suppression of ROS will hugely benefit patients with DHD. Recent Advances: In addition to the gold standard pharmacological therapies, the recent development of gene therapy provides an exciting avenue for developing new therapeutics to treat ROS-mediated DHD. In particular, microRNAs (miRNAs) are gaining interest due to their crucial role in several physiological and pathological processes, including DHD. Critical Issues: miRNAs have many targets and differential function depending on the environment. Therefore, a proper understanding of the function of miRNAs in specific cell types and cell states is required for the successful application of this technology. In the present review, we first provide an overview of the role of ROS in contributing to DHD and the currently available treatments. We then discuss the newer gene therapies with a specific focus on the role of miRNAs as the causative factors and therapeutic targets to combat ROS-mediated DHD. Future Directions: The future of miRNA therapeutics in tackling ROS-mediated DHD is dependent on a complete understanding of how miRNAs behave in different cells and environments. Future research should also aim to develop conditional miRNA therapeutic platforms capable of switching on and off in response to disruptions in the redox state. Antioxid. Redox Signal. 36, 608-630.
Collapse
Affiliation(s)
- Matthew Reily-Bell
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Andrew Bahn
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Gouhar SA, Abo-Elfadl MT, Gamal-Eldeen AM, El-Daly SM. Involvement of miRNAs in response to oxidative stress induced by the steroidal glycoalkaloid α-solanine in hepatocellular carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:212-223. [PMID: 34655286 DOI: 10.1002/tox.23390] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND α-Solanine is a natural toxic glycoalkaloid produced in some species of the Solanaceae family with antiproliferative activity in various cancers. OBJECTIVE This study aimed to investigate the effect of α-solanine on the oxidative stress status in human hepatocellular carcinoma HepG2 cells and to evaluate its influence on microRNAs (miRNAs) associated with oxidative stress and NF-κB regulation. METHODS The prooxidant effect of α-solanine was tested by the decay rate of the fluorescent probe, β-phycoerythrin, and by measuring malondialdehyde, reduced Glutathione, catalase, and superoxide dismutase following treatment of HepG2 cells with low doses of α-solanine. Immunocytochemical techniques were used to detect mitochondrial membrane potential (ΔΨm) and NF-κB protein. The gene expression of NF-κB and miRNAs was evaluated by real-time PCR. RESULTS α-Solanine is a prooxidant that causes a rapid decay in the fluorescence intensity of β-phycoerythrin. It induces oxidative stress-related alterations such as increased lipid peroxidation and reduced antioxidant markers. Oxidative stress induced by α-solanine was mediated by decreased ΔΨm, increased NF-κB expression, upregulation of miRNAs that control oxidative stress by regulating the NF-κB pathway, and downregulation of oncogenic miRNAs that inhibit the NF-κB pathway. CONCLUSION α-Solanine-induced oxidative stress is mediated by alterations in the NF-κB pathway with a detected crosstalk between α-solanine treatment and the expression of oxidative stress-responsive miRNAs.
Collapse
Affiliation(s)
- Shaimaa A Gouhar
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Mahmoud T Abo-Elfadl
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| | - Amira M Gamal-Eldeen
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| | - Sherien M El-Daly
- Medical Biochemistry Department, Medical Research Division, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| |
Collapse
|
14
|
Majka M, Kleibert M, Wojciechowska M. Impact of the Main Cardiovascular Risk Factors on Plasma Extracellular Vesicles and Their Influence on the Heart's Vulnerability to Ischemia-Reperfusion Injury. Cells 2021; 10:3331. [PMID: 34943838 PMCID: PMC8699798 DOI: 10.3390/cells10123331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
The majority of cardiovascular deaths are associated with acute coronary syndrome, especially ST-elevation myocardial infarction. Therapeutic reperfusion alone can contribute up to 40 percent of total infarct size following coronary artery occlusion, which is called ischemia-reperfusion injury (IRI). Its size depends on many factors, including the main risk factors of cardiovascular mortality, such as age, sex, systolic blood pressure, smoking, and total cholesterol level as well as obesity, diabetes, and physical effort. Extracellular vesicles (EVs) are membrane-coated particles released by every type of cell, which can carry content that affects the functioning of other tissues. Their role is essential in the communication between healthy and dysfunctional cells. In this article, data on the variability of the content of EVs in patients with the most prevalent cardiovascular risk factors is presented, and their influence on IRI is discussed.
Collapse
Affiliation(s)
- Miłosz Majka
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (M.M.); (M.K.)
| | - Marcin Kleibert
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (M.M.); (M.K.)
| | - Małgorzata Wojciechowska
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (M.M.); (M.K.)
- Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
| |
Collapse
|
15
|
Contribution of Oxidative Stress to HIF-1-Mediated Profibrotic Changes during the Kidney Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6114132. [PMID: 34712385 PMCID: PMC8548138 DOI: 10.1155/2021/6114132] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/09/2021] [Indexed: 12/01/2022]
Abstract
Hypoxia and oxidative stress are the common causes of various types of kidney injury. During recent years, the studies on hypoxia inducible factor- (HIF-) 1 attract more and more attention, which can not only mediate hypoxia adaptation but also contribute to profibrotic changes. Through analyzing related literatures, we found that oxidative stress can regulate the expression and activity of HIF-1α through some signaling molecules, such as prolyl hydroxylase domain-containing protein (PHD), PI-3K, and microRNA. And oxidative stress can take part in inflammation, epithelial-mesenchymal transition, and extracellular matrix deposition mediated by HIF-1 via interacting with classical NF-κB and TGF-β signaling pathways. Therefore, based on previous literatures, this review summarizes the contribution of oxidative stress to HIF-1-mediated profibrotic changes during the kidney damage, in order to further understand the role of oxidative stress in renal fibrosis.
Collapse
|
16
|
Tanase DM, Gosav EM, Ouatu A, Badescu MC, Dima N, Ganceanu-Rusu AR, Popescu D, Floria M, Rezus E, Rezus C. Current Knowledge of MicroRNAs (miRNAs) in Acute Coronary Syndrome (ACS): ST-Elevation Myocardial Infarction (STEMI). Life (Basel) 2021; 11:life11101057. [PMID: 34685428 PMCID: PMC8541211 DOI: 10.3390/life11101057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023] Open
Abstract
Regardless of the newly diagnostic and therapeutic advances, coronary artery disease (CAD) and more explicitly, ST-elevation myocardial infarction (STEMI), remains one of the leading causes of morbidity and mortality worldwide. Thus, early and prompt diagnosis of cardiac dysfunction is pivotal in STEMI patients for a better prognosis and outcome. In recent years, microRNAs (miRNAs) gained attention as potential biomarkers in myocardial infarction (MI) and acute coronary syndromes (ACS), as they have key roles in heart development, various cardiac processes, and act as indicators of cardiac damage. In this review, we describe the current available knowledge about cardiac miRNAs and their functions, and focus mainly on their potential use as novel circulating diagnostic and prognostic biomarkers in STEMI.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Correspondence: (E.M.G.); (M.F.); (E.R.)
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Ana Roxana Ganceanu-Rusu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, Emergency Military Clinical Hospital Iasi, 700483 Iasi, Romania
- Correspondence: (E.M.G.); (M.F.); (E.R.)
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence: (E.M.G.); (M.F.); (E.R.)
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (M.C.B.); (N.D.); (A.R.G.-R.); (D.P.); (C.R.)
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| |
Collapse
|
17
|
Ruiz GP, Camara H, Fazolini NPB, Mori MA. Extracellular miRNAs in redox signaling: Health, disease and potential therapies. Free Radic Biol Med 2021; 173:170-187. [PMID: 33965563 DOI: 10.1016/j.freeradbiomed.2021.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Extracellular microRNAs (miRNAs) have emerged as important mediators of cell-to-cell communication and intertissue crosstalk. MiRNAs are produced by virtually all types of eukaryotic cells and can be selectively packaged and released to the extracellular medium, where they may reach distal cells to regulate gene expression cell non-autonomously. By doing so, miRNAs participate in integrative physiology. Oxidative stress affects miRNA expression, while miRNAs control redox signaling. Disruption in miRNA expression, processing or release to the extracellular compartment are associated with aging and a number of chronic diseases, such as obesity, type 2 diabetes, neurodegenerative diseases and cancer, all of them being conditions related to oxidative stress. Here we discuss the interplay between redox balance and miRNA function and secretion as a determinant of health and disease states, reviewing the findings that support this notion and highlighting novel and yet understudied venues of research in the field.
Collapse
Affiliation(s)
- Gabriel Palermo Ruiz
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Henrique Camara
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Narayana P B Fazolini
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
18
|
Xin R, Qu D, Su S, Zhao B, Chen D. Downregulation of miR-23b by transcription factor c-Myc alleviates ischemic brain injury by upregulating Nrf2. Int J Biol Sci 2021; 17:3659-3671. [PMID: 34512173 PMCID: PMC8416714 DOI: 10.7150/ijbs.61399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic brain injury (IBI) is a common acute cerebral vessel disease that occurs secondary to blockage in arteries, mainly characterized by insufficient blood supply to the brain. The transcription factor c-Myc in IBI continues to be implicated in numerous studies. This study was conducted with emphasis placed on the underlying mechanism of c-Myc in IBI. Clinical samples were collected from IBI patients. Middle cerebral artery occlusion (MCAO) was induced in mice by inserting a suture from the external carotid artery to the anterior cerebral artery through the internal carotid artery to mechanically block the blood supply at the origin of the middle cerebral artery, and cortical neurons from mice were exposed to oxygen glucose deprivation (OGD) conditions for IBI model in vitro construction. RT-qPCR was performed to determine microRNA-23b (miR-23b) expression. TUNEL staining and Western blot analysis was conducted to detect apoptosis. The regulatory relationship was analyzed by dual-luciferase reporter gene assay. After loss- and gain-of-function assays, triphenyltetrazolium chloride staining was carried out to detect the area of cerebral infarction, after which the spatial memory in mice was evaluated with Morris water maze test. As per our findings, miR-23b was upregulated in the serum of IBI patients and OGD-treated murine primary neurons. Silencing of miR-23b resulted in reduced OGD-induced neuronal apoptosis. miR-23b inversely targeted nuclear factor erythroid 2-related factor 2 (Nrf2) and c-Myc negatively regulated miR-23b expression. Overexpression of c-Myc and inhibition of miR-23b led to reduced neurological scores of infarction area, neuronal apoptosis, shortened platform arrival time and significantly increased the time spent on the platform quadrant and the times of crossing the platform in vivo. Collectively, downregulated miR-23b by c-Myc might alleviate IBI by upregulating Nrf2.
Collapse
Affiliation(s)
- Rui Xin
- Jilin University, Changchun 130000, P. R. China
- Department of Radiology, the Second Hospital of Jilin University, Changchun 130000, P. R. China
| | - Danhua Qu
- Jilin University, Changchun 130000, P. R. China
- Department of Respiratory and Critical Diseases, the Second Hospital of Jilin University, Changchun 130000, P. R. China
| | - Shuang Su
- Sinopec Research Institute of Safety Engineering, Qingdao 266000, P. R. China
| | - Bin Zhao
- Jilin University, Changchun 130000, P. R. China
- Department of Neurosurgery, the Second Hospital of Jilin University, Changchun 130000, P. R. China
| | - Dawei Chen
- Jilin University, Changchun 130000, P. R. China
- Department of Radiation Protection, School of Public Health, Jilin University, Changchun 130000, P. R. China
| |
Collapse
|
19
|
Changes in miR 21 and 23b expression in postnatal hypertrophic heart derived from gestational diabetes precede dilated cardiomyopathy. J Biosci 2021. [DOI: 10.1007/s12038-021-00201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Ishrat R, Ahmed MM, Tazyeen S, Alam A, Farooqui A, Ali R, Imam N, Tamkeen N, Ali S, Zubbair Malik M, Sultan A. In Silico Integrative Approach Revealed Key MicroRNAs and Associated Target Genes in Cardiorenal Syndrome. Bioinform Biol Insights 2021; 15:11779322211027396. [PMID: 34276211 PMCID: PMC8256246 DOI: 10.1177/11779322211027396] [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: 08/23/2020] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiorenal syndromes constellate primary dysfunction of either heart or kidney whereby one organ dysfunction leads to the dysfunction of another. The role of several microRNAs (miRNAs) has been implicated in number of diseases, including hypertension, heart failure, and kidney diseases. Wide range of miRNAs has been identified as ideal candidate biomarkers due to their stable expression. Current study was aimed to identify crucial miRNAs and their target genes associated with cardiorenal syndrome and to explore their interaction analysis. Three differentially expressed microRNAs (DEMs), namely, hsa-miR-4476, hsa-miR-345-3p, and hsa-miR-371a-5p, were obtained from GSE89699 and GSE87885 microRNA data sets, using R/GEO2R tools. Furthermore, literature mining resulted in the retrieval of 15 miRNAs from scientific research and review articles. The miRNAs-gene networks were constructed using miRNet (a Web platform of miRNA-centric network visual analytics). CytoHubba (Cytoscape plugin) was adopted to identify the modules and the top-ranked nodes in the network based on Degree centrality, Closeness centrality, Betweenness centrality, and Stress centrality. The overlapped miRNAs were further used in pathway enrichment analysis. We found that hsa-miR-21-5p was common in 8 pathways out of the top 10. Based on the degree, 5 miRNAs, namely, hsa-mir-122-5p, hsa-mir-222-3p, hsa-mir-21-5p, hsa-mir-146a-5p, and hsa-mir-29b-3p, are considered as key influencing nodes in a network. We suggest that the identified miRNAs and their target genes may have pathological relevance in cardiorenal syndrome (CRS) and may emerge as potential diagnostic biomarkers.
Collapse
Affiliation(s)
- Romana Ishrat
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Mohd Murshad Ahmed
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Safia Tazyeen
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Aftab Alam
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Anam Farooqui
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Rafat Ali
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Nikhat Imam
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Naaila Tamkeen
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Shahnawaz Ali
- Centre for Interdisciplinary Research
in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi,
India
| | - Md Zubbair Malik
- School of Computational and Integrative
Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Armiya Sultan
- Department of Biosciences, Jamia Millia
Islamia (A Central University), New Delhi, India
| |
Collapse
|
21
|
Ciesielska S, Slezak-Prochazka I, Bil P, Rzeszowska-Wolny J. Micro RNAs in Regulation of Cellular Redox Homeostasis. Int J Mol Sci 2021; 22:6022. [PMID: 34199590 PMCID: PMC8199685 DOI: 10.3390/ijms22116022] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 02/08/2023] Open
Abstract
In living cells Reactive Oxygen Species (ROS) participate in intra- and inter-cellular signaling and all cells contain specific systems that guard redox homeostasis. These systems contain both enzymes which may produce ROS such as NADPH-dependent and other oxidases or nitric oxide synthases, and ROS-neutralizing enzymes such as catalase, peroxiredoxins, thioredoxins, thioredoxin reductases, glutathione reductases, and many others. Most of the genes coding for these enzymes contain sequences targeted by micro RNAs (miRNAs), which are components of RNA-induced silencing complexes and play important roles in inhibiting translation of their targeted messenger RNAs (mRNAs). In this review we describe miRNAs that directly target and can influence enzymes responsible for scavenging of ROS and their possible role in cellular redox homeostasis. Regulation of antioxidant enzymes aims to adjust cells to survive in unstable oxidative environments; however, sometimes seemingly paradoxical phenomena appear where oxidative stress induces an increase in the levels of miRNAs which target genes which are supposed to neutralize ROS and therefore would be expected to decrease antioxidant levels. Here we show examples of such cellular behaviors and discuss the possible roles of miRNAs in redox regulatory circuits and further cell responses to stress.
Collapse
Affiliation(s)
- Sylwia Ciesielska
- Department of Systems Biology and Engineering, Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland; (P.B.); (J.R.-W.)
- Biotechnology Centre, Silesian University of Technology, 44-100 Gliwice, Poland;
| | | | - Patryk Bil
- Department of Systems Biology and Engineering, Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland; (P.B.); (J.R.-W.)
- Biotechnology Centre, Silesian University of Technology, 44-100 Gliwice, Poland;
| | - Joanna Rzeszowska-Wolny
- Department of Systems Biology and Engineering, Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland; (P.B.); (J.R.-W.)
- Biotechnology Centre, Silesian University of Technology, 44-100 Gliwice, Poland;
| |
Collapse
|
22
|
Huang J, Zhang W, Zhang CL, Wang L. Interleukin-17 aggravates right ventricular remodeling via activating STAT3 under both normoxia and hypoxia. BMC Cardiovasc Disord 2021; 21:249. [PMID: 34020615 PMCID: PMC8139008 DOI: 10.1186/s12872-021-02069-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/17/2021] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Proinflammatory cytokine interleukin 17 (IL-17) is involved in ventricular remodeling, mainly of the left ventricle. This study was designed to explore the role of IL-17 played in the pathogenesis of right ventricular hypertrophy (RVH), aiming to provide a novel treatment target or diagnostic biomarker options for improving the care of RVH patients. METHODS C57BL/6 mice were maintained in 10% O2 chamber or room air for four weeks. Right ventricular hypertrophy index (RVHI), RV/body weight ratio, pulmonary arteriolar remodeling determined by percent media thickness (%MT), and the cardiomyocyte diameter of RV were evaluated. Mice were treated with exogenous recombinant mouse IL-17 (rmIL-17, 1 μg per dose twice a week) for four weeks. H9c2 cardiomyocytes were cultured and treated with IL-17 (10 ng/mL) and STAT3 inhibitor (10 ng/mL) either under normoxia (21% O2, 5% CO2, 74% N2) or under hypoxia (3% O2, 5% CO2, 92% N2). Cardiomyocyte viability was assessed by Cell counting kit 8 (CCK-8) assay. The mRNA level was detected by RT-PCR, where as the protein expression was measured by Western blot, immunohistochemistry, and immunofluorescent analyses. RESULTS In vivo experiments showed that IL-17 did not affect the pulmonary artery under normoxia, after treatment with rmIL-17, %MT was not changed, while RVHI and the RV/body weight ratio were increased, indicating that IL-17 directly induced right ventricular hypertrophy. In a time-course study, the mice were exposed to hypoxia for 0, 1, 2, 3, 4 weeks, respectively. We found that the expression of IL-17 was gradually upregulated in RV tissue in a time-dependent manner after one week of hypoxia exposure, especially at the third and fourth week. Cardiomyocyte hypertrophy and apoptosis were observed after the exposure of the mice to hypoxia for four weeks, rmIL-17 further aggravated the hypoxia-induced cardiomyocyte hypertrophy and apoptosis. The expression of p-STAT3 in the IL-17-deficient mice was lower than in the wild-type mice. In vitro, IL-17 inhibited cardiomyocyte viability and induced cardiomyocyte apoptosis via STAT3 under both normoxic and hypoxic conditions. CONCLUSIONS These findings support a role for IL-17 as a mediator in the pathogenesis RVH, which might be considered as a potential novel anti-inflammation therapeutic strategy or diagnostic biomarker for RVH.
Collapse
MESH Headings
- Animals
- Cell Hypoxia
- Cell Line
- Disease Models, Animal
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia/physiopathology
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Interleukin-17/toxicity
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphorylation
- Rats
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Ventricular Function, Right/drug effects
- Ventricular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Jing Huang
- Department of Rheumatism and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Wei Zhang
- Department of Emergency Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Cai-Lian Zhang
- Department of Pulmonary and Critical Care Medicine, Yanan University Affiliated Hospital, Yanan, 716000, People's Republic of China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xin Cheng District, Xi'an, 710004, People's Republic of China.
| |
Collapse
|
23
|
Liu L, Wang C, Luo X, Wang Y, Li F. Leonurine Alleviates Hypoxia-Induced Myocardial Damage by Regulating miRNAs. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211007274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective miRNAs as pharmaco-targets have been investigated in multifarious diseases. Our study aimed to determine whether leonurine was a potential cardioprotective agent by targeting miRNAs in hypoxia-stimulated mice and H9c2 cardiomyocytes. Methods Cell proliferation and apoptosis were examined by CCK-8 and TUNEL assay in hypoxia-stimulated rat H9c2 cardiomyocytes. miRNAs expression levels in cardiomyocytes in response to hypoxia stimulation were detected by RT-qPCR. Mice with myocardial injury were induced by chronic intermittent hypoxia stimulation. Results Leonurine alleviated hypoxia-induced cardiac hypertrophy in mice. Moreover, up-regulation of miR-31 and down-regulation of miR-210 in hypoxia-stimulated mice were reversed by leonurine administration. Leonurine exhibited cardioprotective activity in an vitro cell model of hypoxia-stimulated rat H9c2 cardiomyocytes, reflecting that the compound improved hypoxia-induced growth inhibition and apoptosis of cardiomyocytes. TUNEL assay revealed that transfection of miR-31 inhibitors or miR-210 mimics abrogated hypoxia-induced cardiomyocyte apoptosis. In contrast to that, miR-31 mimics or miR-210 inhibitors counteracted the anti-apoptotic effect of leonurine on hypoxia-treated rat H9c2 cardiomyocytes. Conclusion Our findings suggest that miR-31 and miR-210 as the upstream regulators of leonurine are involved in hypoxia-induced cardiomyocyte apoptosis. Leonurine can target miRNAs to protect against hypoxia-induced myocardial damage. miRNAs as potential drug targets may provide prospective therapeutic strategies for the treatment of myocardial damage.
Collapse
Affiliation(s)
- Liping Liu
- Department of Pediatric Cardiovasology, Children’s Medical Center, the Second Xiangya Hospital, Central South University & Institute of Pediatrics, Central South University, Changsha, China
| | - Cheng Wang
- Department of Pediatric Cardiovasology, Children’s Medical Center, the Second Xiangya Hospital, Central South University & Institute of Pediatrics, Central South University, Changsha, China
| | - Xuemei Luo
- Department of Pediatric Cardiovasology, Children’s Medical Center, the Second Xiangya Hospital, Central South University & Institute of Pediatrics, Central South University, Changsha, China
| | - Yuwen Wang
- Department of Pediatric Cardiovasology, Children’s Medical Center, the Second Xiangya Hospital, Central South University & Institute of Pediatrics, Central South University, Changsha, China
| | - Fang Li
- Department of Pediatric Cardiovasology, Children’s Medical Center, the Second Xiangya Hospital, Central South University & Institute of Pediatrics, Central South University, Changsha, China
| |
Collapse
|
24
|
Ferrero G, Carpi S, Polini B, Pardini B, Nieri P, Impeduglia A, Grioni S, Tarallo S, Naccarati A. Intake of Natural Compounds and Circulating microRNA Expression Levels: Their Relationship Investigated in Healthy Subjects With Different Dietary Habits. Front Pharmacol 2021; 11:619200. [PMID: 33519486 PMCID: PMC7840481 DOI: 10.3389/fphar.2020.619200] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Diet has a strong influence on many physiological processes, which in turn have important implications on a variety of pathological conditions. In this respect, microRNAs (miRNAs), a class of small non-coding RNAs playing a relevant epigenetic role in controlling gene expression, may represent mediators between the dietary intake and the healthy status. Despite great advances in the field of nutri-epigenomics, it remains unclear how miRNA expression is modulated by the diet and, specifically, the intake of specific nutrients. We investigated the whole circulating miRNome by small RNA-sequencing performed on plasma samples of 120 healthy volunteers with different dietary habits (vegans, vegetarians, and omnivores). Dietary intakes of specific nutrients were estimated for each subject from the information reported in the food-frequency questionnaire previously validated in the EPIC study. We focused hereby on the intake of 23 natural compounds (NCs) of the classes of lipids, micro-elements, and vitamins. We identified 78 significant correlations (rho > 0.300, p-value < 0.05) among the estimated daily intake of 13 NCs and the expression levels of 58 plasma miRNAs. Overall, vitamin D, sodium, and vitamin E correlated with the largest number of miRNAs. All the identified correlations were consistent among the three dietary groups and 22 of them were confirmed as significant (p-value < 0.05) by age-, gender-, and body-mass index-adjusted Generalized Linear regression Model analysis. miR-23a-3p expression levels were related with different NCs including a significant positive correlation with sodium (rho = 0.377) and significant negative correlations with lipid-related NCs and vitamin E. Conversely, the estimated intake of vitamin D was negatively correlated with the expression of the highest number of circulating miRNAs, particularly miR-1277-5p (rho = −0.393) and miR-144-3p (rho = −0.393). Functional analysis of the targets of sodium intake-correlated miRNAs highlighted terms related to cardiac development. A similar approach on targets of those miRNAs correlated with vitamin D intake showed an enrichment in genes involved in hormone metabolisms, while the response to chronic inflammation was among the top enriched processes involving targets of miRNAs negatively related with vitamin E intake. Our findings show that nutrients through the habitual diet influence circulating miRNA profiles and highlight that this aspect must be considered in the nutri-epigenomic research.
Collapse
Affiliation(s)
- Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy.,Department of Computer Science, University of Turin, Torino, Italy
| | - Sara Carpi
- Department of Pharmacy, University of Pisa, Pisa, Italy.,NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Pisa, Italy
| | | | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Torino, Italy
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Sara Grioni
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Sonia Tarallo
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Torino, Italy
| | - Alessio Naccarati
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Torino, Italy
| |
Collapse
|
25
|
Akbari G. Emerging roles of microRNAs in intestinal ischemia/reperfusion-induced injury: a review. J Physiol Biochem 2020; 76:525-537. [PMID: 33140255 DOI: 10.1007/s13105-020-00772-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
Intestinal ischemia/reperfusion (II/R) injury is a serious pathological phenomenon in underlying hemorrhagic shock, trauma, strangulated intestinal obstruction, and acute mesenteric ischemia which associated with high morbidity and mortality. MicroRNAs (miRNAs, miRs) are endogenous non-coding RNAs that regulate post-transcriptionally target mRNA translation via degrading it and/or suppressing protein synthesis. This review discusses on the role of some miRNAs in underlying II/R injury. Some of these miRNAs can have protective action through agomiR or specific antagomiR, and others can have destructive effects in the basal level of II/R insult. Based on these literature reviews, II/R injury affects several miRNAs and their specific target genes. Some miRNAs upregulate under condition of II/R injury, and multiple miRNAs downregulate following II/R damage. Data of this review have been collected from the scientific articles published in databases such as Science Direct, Scopus, PubMed, Web of Science, and Scientific Information Database from 2000 to 2020. It is shown a correlation between changes in the expression of miRNAs and autophagy, inflammation, oxidative stress, apoptosis, and epithelial barrier function. Taken together, agomiR or antagomiR of some miRNAs can be considered as one new target for the research and development of innovative drugs to the prevention or treatment of II/R damage.
Collapse
Affiliation(s)
- Ghaidafeh Akbari
- Medicinal Plants Research Center, Department of Physiology, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
26
|
Yu FR, Xia YW, Wang SB, Xiao LH. Long noncoding RNA PVT1 facilitates high glucose-induced cardiomyocyte death through the miR-23a-3p/CASP10 axis. Cell Biol Int 2020; 45:154-163. [PMID: 33049089 DOI: 10.1002/cbin.11479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022]
Abstract
Dilated cardiomyopathy (DCM) is the leading cause of morbidity and mortality in diabetic patients. Long noncoding RNA plasmacytoma variant translocation 1 (PVT1) has been shown to be related to the pathogenesis of DCM. However, the mechanism by which PVT1 regulates DCM pathogenesis is unclear. High glucose level was employed to construct a DCM cell model in vitro. Cell viability was determined via cell counting kit-8 assay. The level of lactate dehydrogenase (LDH) was measured with the corresponding kit. Expression levels of PVT1, miR-23a-3p, and caspase-10 (CASP10) messenger RNA were evaluated with a quantitative real-time polymerase chain reaction. Cell apoptosis was assessed by flow cytometry assay. Protein levels of B-cell lymphoma 2-associated X (Bax), cleaved-caspase-3 (cleaved-casp-3), and CASP10 were examined via western blot analysis. The relationship between PVT1 or CASP10 and miR-23a-3p was verified with dual-luciferase reporter assay. We observed that PVT1 and CASP10 were upregulated while miR-23a-3p was downregulated in high glucose-induced cardiomyocytes. High glucose levels repressed cardiomyocyte activity and induced cardiomyocyte apoptosis, but this influence was antagonized by PVT1 knockdown or miR-23a-3p overexpression. Furthermore, PVT1 acted as a sponge for miR-23a-3p, and miR-23a-3p inhibition counterbalanced the influence of PVT1 silencing on viability and apoptosis of cardiomyocytes under high glucose level treatment. PVT1 could increase CASP10 expression via sponging miR-23a-3p. In conclusion, PVT1 acted as a deleterious lncRNA in DCM. PVT1 facilitated cardiomyocyte death by regulating the miR-23a-3p/CASP10, which offered a new mechanism to comprehend the pathogenesis of DCM.
Collapse
Affiliation(s)
- Feng-Rong Yu
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| | - Yin-Wen Xia
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| | - Shao-Bo Wang
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| | - Li-Hua Xiao
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| |
Collapse
|
27
|
Bhaskar S, Sheshadri P, Joseph JP, Potdar C, Prasanna J, Kumar A. Mitochondrial Superoxide Dismutase Specifies Early Neural Commitment by Modulating Mitochondrial Dynamics. iScience 2020; 23:101564. [PMID: 33083732 PMCID: PMC7522125 DOI: 10.1016/j.isci.2020.101564] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 07/28/2020] [Accepted: 09/11/2020] [Indexed: 12/02/2022] Open
Abstract
Studies revealing molecular mechanisms underlying neural specification have majorly focused on the role played by different transcription factors, but less on non-nuclear components. Earlier, we reported mitochondrial superoxide dismutase (SOD2) to be essential for self-renewal and pluripotency of mouse embryonic stem cells (mESCs). In the present study, we found SOD2 to be specifically required for neural lineage, but not the meso- or endoderm specification. Temporally, SOD2 regulated early neural genes, but not the matured genes, by modulating mitochondrial dynamics—specifically by enhancing the mitochondrial fusion protein Mitofusin 2 (MFN2). Bio-complementation strategy further confirmed SOD2 to enhance mitochondrial fusion process independent of its antioxidant activity. Over-expression of SOD2 along with OCT4, but neither alone, transdifferentiated mouse fibroblasts to neural progenitor-like colonies, conclusively proving the neurogenic potential of SOD2. In conclusion, our findings accredit a novel role for SOD2 in early neural lineage specification. SOD2 is essential for early neural differentiation of mESCs Up-regulation of MFN2, but not MFN1, underlies SOD2-mediated neurogenesis Antioxidant enzymatic activity of SOD2 is dispensable for mitochondrial fusion Overexpression of SOD2 with OCT4 transdifferentiate MEFs to iNPLCs
Collapse
Affiliation(s)
- Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalasandra, Yelahanka, Bengaluru, 560065 Karnataka, India
| | - Preethi Sheshadri
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalasandra, Yelahanka, Bengaluru, 560065 Karnataka, India
| | - Joel P Joseph
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalasandra, Yelahanka, Bengaluru, 560065 Karnataka, India
| | - Chandrakanta Potdar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalasandra, Yelahanka, Bengaluru, 560065 Karnataka, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalasandra, Yelahanka, Bengaluru, 560065 Karnataka, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalasandra, Yelahanka, Bengaluru, 560065 Karnataka, India
| |
Collapse
|
28
|
Desjarlais M, Dussault S, Rivera JC, Chemtob S, Rivard A. MicroRNA Expression Profiling of Bone Marrow-Derived Proangiogenic Cells (PACs) in a Mouse Model of Hindlimb Ischemia: Modulation by Classical Cardiovascular Risk Factors. Front Genet 2020; 11:947. [PMID: 32973881 PMCID: PMC7472865 DOI: 10.3389/fgene.2020.00947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Classical cardiovascular risk factors (CRFs) are associated with impaired angiogenic activities of bone marrow–derived proangiogenic cells (PACs) related to peripheral artery diseases (PADs) and ischemia-induced neovascularization. MicroRNAs (miRs) are key regulators of gene expression, and they are involved in the modulation of PAC function and PAC paracrine activity. However, the effects of CRFs on the modulation of miR expression in PACs are unknown. Aims and Methods We used a model of hindlimb ischemia and next-generation sequencing to perform a complete profiling of miRs in PACs isolated from the bone marrow of mice subjected to three models of CRFs: aging, smoking (SMK) and hypercholesterolemia (HC). Results Approximately 570 miRs were detected in PACs in the different CRF models. When excluding miRs with a very low expression level (<100 RPM), 40 to 61 miRs were found to be significantly modulated by aging, SMK, or HC. In each CRF condition, we identified downregulated proangiogenic miRs and upregulated antiangiogenic miRs that could contribute to explain PAC dysfunction. Interestingly, several miRs were similarly downregulated (e.g., miR-542-3p, miR-29) or upregulated (e.g., miR-501, miR-92a) in all CRF conditions. In silico approaches including Kyoto Encyclopedia of Genes and Genomes and cluster dendogram analyses identified predictive effects of these miRs on pathways having key roles in the modulation of angiogenesis and PAC function, including vascular endothelial growth factor signaling, extracellular matrix remodeling, PI3K/AKT/MAPK signaling, transforming growth factor beta (TGFb) pathway, p53, and cell cycle progression. Conclusion This study describes for the first time the effects of CRFs on the modulation of miR profile in PACs related to PAD and ischemia-induced neovascularization. We found that several angiogenesis-modulating miRs are similarly altered in different CRF conditions. Our findings constitute a solid framework for the identification of miRs that could be targeted in PACs in order to improve their angiogenic function and for the future development of novel therapies to improve neovascularization and reduce tissue damage in patients with severe PAD.
Collapse
Affiliation(s)
- Michel Desjarlais
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada.,Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Sylvie Dussault
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada
| | - José Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alain Rivard
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada
| |
Collapse
|
29
|
Abstract
Heart failure is a growing health problem resulting in the decreased life expectancy of patients and severely increased the healthcare burden. Penetrating research on the pathogenesis and regulation mechanism of heart failure is important for treatment of heart failure. Epicardial adipose tissue (EAT) has been demonstrated as not only a dynamic organ with biological functions but also an inert lipid store with regulating systemic metabolism. EAT mediates physiological and pathophysiological processes of heart failure by regulating adipogenesis, cardiac remodeling, insulin resistance, cardiac output, and renin angiotensin aldosterone system (RAAS). Moreover, EAT secretes a wide range of adipokines, adrenomedullin, adiponectin, and miRNAs through paracrine, endocrine, and vasocrine pathways, which involve in various extracellular and intracellular mechanism of cardiac-related cells in the progress of cardiovascular disease especially in heart failure. Nevertheless, mechanisms and roles of EAT on heart failure are barely summarized. Understanding the regulating mechanisms of EAT on heart failure may give rise to novel therapeutic targets and will open up innovative strategies to myocardial injury as well as in heart failure.
Collapse
Affiliation(s)
- Ying Song
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China.,Xiamen Branch of Zhongshan Hospital, Fudan University, Xiamen, 361015, Fujian, China
| | - Fei Song
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, No. 2999 Jinshan Road, Xiamen, 361015, Fujian, China.
| |
Collapse
|
30
|
Substantially Altered Expression Profile of Diabetes/Cardiovascular/Cerebrovascular Disease Associated microRNAs in Children Descending from Pregnancy Complicated by Gestational Diabetes Mellitus-One of Several Possible Reasons for an Increased Cardiovascular Risk. Cells 2020; 9:cells9061557. [PMID: 32604801 PMCID: PMC7349356 DOI: 10.3390/cells9061557] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Gestational diabetes mellitus (GDM), one of the major pregnancy-related complications, characterized as a transitory form of diabetes induced by insulin resistance accompanied by a low/absent pancreatic beta-cell compensatory adaptation to the increased insulin demand, causes the acute, long-term, and transgenerational health complications. The aim of the study was to assess if alterations in gene expression of microRNAs associated with diabetes/cardiovascular/cerebrovascular diseases are present in whole peripheral blood of children aged 3-11 years descending from GDM complicated pregnancies. A substantially altered microRNA expression profile was found in children descending from GDM complicated pregnancies. Almost all microRNAs with the exception of miR-92a-3p, miR-155-5p, and miR-210-3p were upregulated. The microRNA expression profile also differed between children after normal and GDM complicated pregnancies in relation to the presence of overweight/obesity, prehypertension/hypertension, and/or valve problems and heart defects. Always, screening based on the combination of microRNAs was superior over using individual microRNAs, since at 10.0% false positive rate it was able to identify a large proportion of children with an aberrant microRNA expression profile (88.14% regardless of clinical findings, 75.41% with normal clinical findings, and 96.49% with abnormal clinical findings). In addition, the higher incidence of valve problems and heart defects was found in children with a prior exposure to GDM. The extensive file of predicted targets of all microRNAs aberrantly expressed in children descending from GDM complicated pregnancies indicates that a large group of these genes is involved in ontologies of diabetes/cardiovascular/cerebrovascular diseases. In general, children with a prior exposure to GDM are at higher risk of later development of diabetes mellitus and cardiovascular/cerebrovascular diseases, and would benefit from dispensarisation as well as implementation of primary prevention strategies.
Collapse
|
31
|
Climent M, Viggiani G, Chen YW, Coulis G, Castaldi A. MicroRNA and ROS Crosstalk in Cardiac and Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21124370. [PMID: 32575472 PMCID: PMC7352701 DOI: 10.3390/ijms21124370] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) affect many cellular functions and the proper redox balance between ROS and antioxidants contributes substantially to the physiological welfare of the cell. During pathological conditions, an altered redox equilibrium leads to increased production of ROS that in turn may cause oxidative damage. MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level contributing to all major cellular processes, including oxidative stress and cell death. Several miRNAs are expressed in response to ROS to mediate oxidative stress. Conversely, oxidative stress may lead to the upregulation of miRNAs that control mechanisms to buffer the damage induced by ROS. This review focuses on the complex crosstalk between miRNAs and ROS in diseases of the cardiac (i.e., cardiac hypertrophy, heart failure, myocardial infarction, ischemia/reperfusion injury, diabetic cardiomyopathy) and pulmonary (i.e., idiopathic pulmonary fibrosis, acute lung injury/acute respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, lung cancer) compartments. Of note, miR-34a, miR-144, miR-421, miR-129, miR-181c, miR-16, miR-31, miR-155, miR-21, and miR-1/206 were found to play a role during oxidative stress in both heart and lung pathologies. This review comprehensively summarizes current knowledge in the field.
Collapse
Affiliation(s)
- Montserrat Climent
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano, MI, Italy;
| | - Giacomo Viggiani
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, MI, Italy;
| | - Ya-Wen Chen
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Gerald Coulis
- Department of Physiology and Biophysics, and Institute for Immunology, University of California Irvine, Irvine, CA 92697, USA;
| | - Alessandra Castaldi
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Correspondence:
| |
Collapse
|
32
|
Abstract
Kawasaki disease is an acute febrile illness and systemic vasculitis of unknown aetiology that predominantly afflicts young children, causes coronary artery aneurysms and can result in long-term cardiovascular sequelae. Kawasaki disease is the leading cause of acquired heart disease among children in the USA. Coronary artery aneurysms develop in some untreated children with Kawasaki disease, leading to ischaemic heart disease and myocardial infarction. Although intravenous immunoglobulin (IVIG) treatment reduces the risk of development of coronary artery aneurysms, some children have IVIG-resistant Kawasaki disease and are at increased risk of developing coronary artery damage. In addition, the lack of specific diagnostic tests and biomarkers for Kawasaki disease make early diagnosis and treatment challenging. The use of experimental mouse models of Kawasaki disease vasculitis has considerably improved our understanding of the pathology of the disease and helped characterize the cellular and molecular immune mechanisms contributing to cardiovascular complications, in turn leading to the development of innovative therapeutic approaches. Here, we outline the pathophysiology of Kawasaki disease and summarize and discuss the progress gained from experimental mouse models and their potential therapeutic translation to human disease. This Review outlines the pathophysiology of Kawasaki disease and discusses the progress gained from experimental mouse models and their potential therapeutic translation to human disease. Kawasaki disease is a childhood systemic vasculitis leading to the development of coronary artery aneurysms; it is the leading cause of acquired heart disease in children in developed countries. The cause of Kawasaki disease is unknown, although it is suspected to be triggered by an unidentified infectious pathogen in genetically predisposed children. Kawasaki disease might not be a normal immune response to an unusual environmental stimulus, but rather a genetically determined unusual and uncontrolled immune response to a common stimulus. Although the aetiological agent in humans is unknown, mouse models of Kawasaki disease vasculitis demonstrate similar pathological features and have substantially accelerated discoveries in the field. Genetic and transcriptomic analysis of blood samples from patients with Kawasaki disease and experimental evidence generated using mouse models have demonstrated the critical role of IL-1β in the pathogenesis of this disease and the therapeutic potential of targeting this pathway (currently under investigation in clinical trials).
Collapse
|
33
|
Diabetes Mellitus and Cardiovascular Risk Assessment in Mothers with a History of Gestational Diabetes Mellitus Based on Postpartal Expression Profile of MicroRNAs Associated with Diabetes Mellitus and Cardiovascular and Cerebrovascular Diseases. Int J Mol Sci 2020; 21:ijms21072437. [PMID: 32244558 PMCID: PMC7177375 DOI: 10.3390/ijms21072437] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Mothers with a history of gestational diabetes mellitus (GDM) have an increased risk of developing diabetes in the future and a lifelong cardiovascular risk. Postpartal expression profile of cardiovascular/cerebrovascular disease associated microRNAs was assessed 3–11 years after the delivery in whole peripheral blood of young and middle-aged mothers with a prior exposure to GDM with the aim to identify a high-risk group of mothers at risk of later development of diabetes mellitus and cardiovascular/cerebrovascular diseases who would benefit from implementation of early primary prevention strategies and long-term follow-up. The hypothesis of the assessment of cardiovascular risk in women was based on the knowledge that a series of microRNAs play a role in the pathogenesis of diabetes mellitus and cardiovascular/cerebrovascular diseases. Abnormal expression profile of multiple microRNAs was found in women with a prior exposure to GDM (miR-1-3p, miR-16-5p, miR-17-5p, miR-20a-5p, miR-20b-5p, miR-21-5p, miR-23a-3p, miR-24-3p, miR-26a-5p, miR-29a-3p, miR-100-5p, miR-103a-3p, miR-125b-5p, miR-126-3p, miR-130b-3p, miR-133a-3p, miR-143-3p, miR-145-5p, miR-146a-5p, miR-181a-5p, miR-195-5p, miR-199a-5p, miR-221-3p, miR-342-3p, miR-499a-5p, and-miR-574-3p). Postpartal combined screening of miR-1-3p, miR-16-5p, miR-17-5p, miR-20b-5p, miR-21-5p, miR-23a-3p, miR-26a-5p, miR-29a-3p, miR-103a-3p, miR-133a-3p, miR-146a-5p, miR-181a-5p, miR-195-5p, miR-199a-5p, miR-221-3p, and miR-499a-5p showed the highest accuracy for the identification of mothers with a prior exposure to GDM at a higher risk of later development of cardiovascular/cerebrovascular diseases (AUC 0.900, p < 0.001, sensitivity 77.48%, specificity 93.26%, cut off >0.611270413). It was able to identify 77.48% mothers with an increased cardiovascular risk at 10.0% FPR. Any of changes in epigenome (upregulation of miR-16-5p, miR-17-5p, miR-29a-3p, and miR-195-5p) that were induced by GDM-complicated pregnancy are long-acting and may predispose mothers affected with GDM to later development of diabetes mellitus and cardiovascular/cerebrovascular diseases. In addition, novel epigenetic changes (upregulation of serious of microRNAs) appeared in a proportion of women that were exposed to GDM throughout the postpartal life. Likewise, a previous occurrence of either GH, PE, and/or FGR, as well as a previous occurrence of GDM, is associated with the upregulation of miR-1-3p, miR-17-5p, miR-20a-5p, miR-20b-5p, miR-29a-3p, miR-100-5p, miR-125b-5p, miR-126-3p, miR-130b-3p, miR-133a-3p, miR-143-3p, miR-145-5p, miR-146a-5p, miR-181a-5p, miR-199a-5p, miR-221-3p, and miR-499a-5p. On the other hand, upregulation of miR-16-5p, miR-21-5p, miR-23a-3p, miR-24-3p, miR-26a-5p, miR-103a-3p, miR-195-5p, miR-342-3p, and miR-574-3p represents a unique feature of aberrant expression profile of women with a prior exposure to GDM. Screening of particular microRNAs may stratify a high-risk group of mothers with a history of GDM who might benefit from implementation of early primary prevention strategies.
Collapse
|
34
|
Evaluation of Vascular Endothelial Function in Young and Middle-Aged Women with Respect to a History of Pregnancy, Pregnancy-Related Complications, Classical Cardiovascular Risk Factors, and Epigenetics. Int J Mol Sci 2020; 21:ijms21020430. [PMID: 31936594 PMCID: PMC7013677 DOI: 10.3390/ijms21020430] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the study was to examine the effect of previous pregnancies and classical cardiovascular risk factors on vascular endothelial function in a group of 264 young and middle-aged women 3 to 11 years postpartum. We examined microvascular functions by peripheral arterial tonometry and EndoPAT 2000 device with respect to a history of gestational hypertension, preeclampsia, fetal growth restriction, the severity of the disease with regard to the degree of clinical signs and delivery date. Besides, we compared Reactive Hyperemia Index (RHI) values and the prevalence of vascular endothelial dysfunction among the groups of women with normal and abnormal values of BMI, waist circumference, systolic and diastolic blood pressures, heart rate, total serum cholesterol levels, serum high-density lipoprotein cholesterol levels, serum low-density lipoprotein cholesterol levels, serum triglycerides levels, serum lipoprotein A levels, serum C-reactive protein levels, serum uric acid levels, and plasma homocysteine levels. Furthermore, we determined the effect of total number of pregnancies and total parity per woman, infertility and blood pressure treatment, presence of trombophilic gene mutations, current smoking of cigarettes, and current hormonal contraceptive use on the vascular endothelial function. We also examined the association between the vascular endothelial function and postpartum whole peripheral blood expression of microRNAs involved in pathogenesis of cardiovascular/cerebrovascular diseases (miR-1-3p, miR-16-5p, miR-17-5p, miR-20a-5p, miR-20b-5p, miR-21-5p, miR-23a-3p, miR-24-3p, miR-26a-5p, miR-29a-3p, miR-92a-3p, miR-100-5p, miR-103a-3p, miR-125b-5p, miR-126-3p, miR-130b-3p, miR-133a-3p, miR-143-3p, miR-145-5p, miR-146a-5p, miR-155-5p, miR-181a-5p, miR-195-5p, miR-199a-5p, miR-210-3p, miR-221-3p, miR-342-3p, miR-499a-5p, and miR-574-3p). A proportion of overweight women (17.94% and 20.59%) and women with central obesity (18.64% and 21.19%) had significantly lower RHI values at 10.0% false positive rate (FPR) both before and after adjustment of the data for the age of patients. At 10.0% FPR, a proportion of women with vascular endothelial dysfunction (RHI ≤ 1.67) was identified to have up-regulated expression profile of miR-1-3p (11.76%), miR-23a-3p (17.65%), and miR-499a-5p (18.82%) in whole peripheral blood. RHI values also negatively correlated with expression of miR-1-3p, miR-23a-3p, and miR-499a-5p in whole peripheral blood. Otherwise, no significant impact of other studied factors on vascular endothelial function was found. We suppose that screening of these particular microRNAs associated with vascular endothelial dysfunction may help to stratify a highly risky group of young and middle-aged women that would benefit from early implementation of primary prevention strategies. Nevertheless, it is obvious, that vascular endothelial dysfunction is just one out of multiple cardiovascular risk factors which has only a partial impact on abnormal expression of cardiovascular and cerebrovascular disease associated microRNAs in whole peripheral blood of young and middle-aged women.
Collapse
|
35
|
Kura B, Szeiffova Bacova B, Kalocayova B, Sykora M, Slezak J. Oxidative Stress-Responsive MicroRNAs in Heart Injury. Int J Mol Sci 2020; 21:E358. [PMID: 31948131 PMCID: PMC6981696 DOI: 10.3390/ijms21010358] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are important molecules in the living organisms as a part of many signaling pathways. However, if overproduced, they also play a significant role in the development of cardiovascular diseases, such as arrhythmia, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction and heart transplantation), and heart failure. As a result of oxidative stress action, apoptosis, hypertrophy, and fibrosis may occur. MicroRNAs (miRNAs) represent important endogenous nucleotides that regulate many biological processes, including those involved in heart damage caused by oxidative stress. Oxidative stress can alter the expression level of many miRNAs. These changes in miRNA expression occur mainly via modulation of nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, calcineurin/nuclear factor of activated T cell (NFAT), or nuclear factor kappa B (NF-κB) pathways. Up until now, several circulating miRNAs have been reported to be potential biomarkers of ROS-related cardiac diseases, including myocardial infarction, hypertrophy, ischemia/reperfusion, and heart failure, such as miRNA-499, miRNA-199, miRNA-21, miRNA-144, miRNA-208a, miRNA-34a, etc. On the other hand, a lot of studies are aimed at using miRNAs for therapeutic purposes. This review points to the need for studying the role of redox-sensitive miRNAs, to identify more effective biomarkers and develop better therapeutic targets for oxidative-stress-related heart diseases.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| |
Collapse
|
36
|
Qiao Y, Wang C, Kou J, Wang L, Han D, Huo D, Li F, Zhou X, Meng D, Xu J, Murtaza G, Artyom B, Ma N, Luo S. MicroRNA-23a suppresses the apoptosis of inflammatory macrophages and foam cells in atherogenesis by targeting HSP90. Gene 2019; 729:144319. [PMID: 31884108 DOI: 10.1016/j.gene.2019.144319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 11/25/2022]
Abstract
In previous study, we have found that microRNA-23a is down regulated in atherosclerotic tissues. Here we demonstrate that miR-23a directly binds to 3'UTR of HSP90 mRNA to suppress the expression of HSP90. To investigate the potential roles of miR-23a in macrophage, THP-1 macrophages were transfected with miR-23a mimics or inhibitors. Our results showed inflammatory factors IL-6 and MCP-1 concentrations in cell culture medium of macrophage and foam cell transfected with miR-23a mimics were decreased. Furthermore, we find that apoptosis of macrophage and foam cells transfected with miR-23a mimics were inhibited. Over expression of miR-23a in foam cells could reduced lipid intake and accumulation in foam cells. Meanwhile, we found that in inflammatory macrophages and foam cells transfected with miR-23a mimcs, HSP90 and NF-κB proteins are significantly decreased. Our results have suggested a promising and potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Yu Qiao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Chuxuan Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Translational Medicine Center of Northern China, Harbin Medical University, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Lujing Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Dong Han
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Da Huo
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Fuyan Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Xiaoxi Zhou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Dehao Meng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Jiaran Xu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Ghulam Murtaza
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Bobkov Artyom
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China
| | - Ning Ma
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China; Medical Science Institute of Hei Longjiang Province, Harbin, China.
| | - Shanshun Luo
- Department of Gerontology, The First Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
37
|
Stoeva M. RETRACTED ARTICLE: Apoptotic suppression of inflammatory macrophages and foam cells in vascular tissue by miR-23a. HEALTH AND TECHNOLOGY 2019. [DOI: 10.1007/s12553-019-00301-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
38
|
Bukauskas T, Mickus R, Cereskevicius D, Macas A. Value of Serum miR-23a, miR-30d, and miR-146a Biomarkers in ST-Elevation Myocardial Infarction. Med Sci Monit 2019; 25:3925-3932. [PMID: 31130720 PMCID: PMC6556071 DOI: 10.12659/msm.913743] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The aim of this study was to analyze the relative expression level of miR-30d-5p, miR-23a-3p, and miR-146a-5p, and to comprehensively assess the diagnostic and predictive possibilities of these miRNAs. Their expression changes have not yet been sufficiently investigated during acute myocardial infarction. Therefore, it is important to comprehensively assess the diagnostic and predictive possibilities of these micro-ribonucleic acids (miRNAs). MATERIAL AND METHODS Random patients with ST‑elevated myocardial infarction (STEMI) were enrolled into the study group. The control group was comprised of patients with no inflammation or ischemic heart disease who were hospitalized for minor elective surgery. The relative expression level for each miRNA was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR)-analysis. RESULTS There were 88 participants enrolled into the study: 62 patients were diagnosed with STEMI and there were 26 healthy controls. Expressions of miR-30d-5p, miR-146a-5p, and miR-23a-3p were respectively 1.581-fold, 4.048-fold, and 4.857-fold lower in patients with STEMI compared to the control group patients (all P values were <0.001). Downregulation of miR-23a-3p was significantly negatively correlated with risk scores of GRACE (Global Registry of Acute Coronary Events) and APACHE II (Acute Physiology and Chronic Health Evaluation II). MiR-23a-3p was a fair predictor for STEMI: area under the curve (AUC)=0.806. Cox regression analysis revealed that expression levels of analyzed miRNAs were not significantly associated with negative endpoints at 1 month after the onset of STEMI. CONCLUSIONS All investigated miRNAs were differentially expressed when comparing patients with STEMI and control group individuals. The evaluation of miR-23a-3p expression levels in serum could be useful to assess the severity of STEMI and as a potential diagnostic biomarker of this condition. In addition, miR-23a-3p may provide limited short-term prognostic value for STEMI patients.
Collapse
Affiliation(s)
- Tomas Bukauskas
- Department of Anesthesiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rytis Mickus
- Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Darius Cereskevicius
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Andrius Macas
- Department of Anesthesiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
39
|
Fan PC, Chen CC, Peng CC, Chang CH, Yang CH, Yang C, Chu LJ, Chen YC, Yang CW, Chang YS, Chu PH. A circulating miRNA signature for early diagnosis of acute kidney injury following acute myocardial infarction. J Transl Med 2019; 17:139. [PMID: 31039814 PMCID: PMC6492315 DOI: 10.1186/s12967-019-1890-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/23/2019] [Indexed: 12/31/2022] Open
Abstract
Background Acute kidney injury (AKI) is a common complication of acute myocardial infarction (AMI), and is associated with adverse outcomes. The study aimed to identify a miRNA signature for the early diagnosis of post-AMI AKI. Methods A total of 108 patients admitted to a coronary care unit (CCU) were divided into four subgroups: AMI−AKI−, AMI+AKI−, AMI+AKI+, and AMI−AKI+. Thirty-six miRNA candidates were selected based on an extensive literature review. Real-time quantitative RT-PCR analysis was used to determine the expression levels of these miRNAs in the serum collected on the day of CCU admittance. TargetScan 7.1 and miRDB databases were used for target prediction and Metacore 6.13 was used for pathway analysis. Results Through a stepwise selection based on abundance, hemolytic effect and differential expression between four groups, 9 miRNAs were found to have significantly differential expression levels as potential biomarkers for post-AMI AKI specifically. Noticeably, the expression levels of miR-24, miR-23a and miR-145 were significantly down-regulated in AMI+AKI+ patients compared to those in AMI+AKI− patients. Combination of the three miRNAs as a panel showed the best performance in the early detection of AKI following AMI (AUC = 0.853, sensitivity 95.65%), compared to the analysis of serum neutrophil gelatinase-associated lipocalin (AUC = 0.735, sensitivity 63.16%). Furthermore, bioinformatic analysis indicated that these three miRNAs regulate the transforming growth factor beta signaling pathway and involve in apoptosis and fibrosis in AKI. Conclusions For the first time, this study identify a unique circulating miRNA signature (miR-24-3p, miR-23a-3p, miR-145-5p) that can potentially early detect AKI following AMI and may be involved in renal injury and fibrosis in post-AMI AKI pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12967-019-1890-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pei-Chun Fan
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University College of Medicine, No. 5 Fusing Street, Gueishan Dist., Taoyuan City, 333, Taiwan, ROC.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, No. 5 Fusing Street, Gueishan Dist., Taoyuan City, 333, Taiwan, ROC
| | - Chia-Chun Chen
- Molecular Medicine Research Center, Chang Gung University, No 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan City, 33302, Taiwan, ROC.,Department of Colorectal Surgery, Chang Gung Memorial Hospital, No 259 Wen-Hwa 1st Road, Kwei-Shan, Linkou, Taoyuan City, 33302, Taiwan, ROC
| | - Chen-Ching Peng
- Molecular Medicine Research Center, Chang Gung University, No 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan City, 33302, Taiwan, ROC
| | - Chih-Hsiang Chang
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University College of Medicine, No. 5 Fusing Street, Gueishan Dist., Taoyuan City, 333, Taiwan, ROC.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, No. 5 Fusing Street, Gueishan Dist., Taoyuan City, 333, Taiwan, ROC
| | - Chia-Hung Yang
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5 Fusing Street, Gueishan Dist., Taoyuan City, 333, Taiwan, ROC
| | - Chi Yang
- Molecular Medicine Research Center, Chang Gung University, No 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan City, 33302, Taiwan, ROC
| | - Lichieh Julie Chu
- Molecular Medicine Research Center, Chang Gung University, No 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan City, 33302, Taiwan, ROC
| | - Yung-Chang Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung Branch, Chang Gung University College of Medicine, No. 222, Maijin Rd., Anle Dist., Keelung City, 20401, Taiwan, ROC
| | - Chih-Wei Yang
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University College of Medicine, No. 5 Fusing Street, Gueishan Dist., Taoyuan City, 333, Taiwan, ROC
| | - Yu-Sun Chang
- Molecular Medicine Research Center, Chang Gung University, No 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan City, 33302, Taiwan, ROC
| | - Pao-Hsien Chu
- Department of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 199 Tung Hwa North Road, Taipei, 105, Taiwan.
| |
Collapse
|
40
|
Du J, Hang P, Pan Y, Feng B, Zheng Y, Chen T, Zhao L, Du Z. Inhibition of miR-23a attenuates doxorubicin-induced mitochondria-dependent cardiomyocyte apoptosis by targeting the PGC-1α/Drp1 pathway. Toxicol Appl Pharmacol 2019; 369:73-81. [DOI: 10.1016/j.taap.2019.02.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023]
|
41
|
Integrative System Biology Analyses Identify Seven MicroRNAs to Predict Heart Failure. Noncoding RNA 2019; 5:ncrna5010022. [PMID: 30866581 PMCID: PMC6468490 DOI: 10.3390/ncrna5010022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) has several etiologies including myocardial infarction (MI) and left ventricular remodeling (LVR), but its progression remains difficult to predict in clinical practice. Systems biology analyses of LVR after MI provide molecular insights into this event such as modulation of microRNA (miRNA) that could be used as a signature of HF progression. To define a miRNA signature of LVR after MI, we use 2 systems biology approaches, integrating either proteomic data generated from LV of post-MI rat induced by left coronary artery ligation or multi-omics data (proteins and non-coding RNAs) generated from plasma of post-MI patients from the REVE-2 study. The first approach predicted that 13 miRNAs and 3 of these miRNAs would be validated to be associated with LVR in vivo: miR-21-5p, miR-23a-3p and miR-222-3p. The second approach predicted that 24 miRNAs among 1310 molecules and 6 of these miRNAs would be selected to be associated with LVR in silico: miR-17-5p, miR-21-5p, miR-26b-5p, miR-222-3p, miR-335-5p and miR-375. We identified a signature of 7 microRNAs associated with LVR after MI that support the interest of integrative systems biology analyses to define a miRNA signature of HF progression.
Collapse
|
42
|
Hromadnikova I, Kotlabova K, Dvorakova L, Krofta L, Sirc J. Postnatal Expression Profile of microRNAs Associated with Cardiovascular and Cerebrovascular Diseases in Children at the Age of 3 to 11 Years in Relation to Previous Occurrence of Pregnancy-Related Complications. Int J Mol Sci 2019; 20:ijms20030654. [PMID: 30717412 PMCID: PMC6387366 DOI: 10.3390/ijms20030654] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
Children descending from pregnancies complicated by gestational hypertension (GH), preeclampsia (PE) or fetal growth restriction (FGR) have a lifelong cardiovascular risk. The aim of the study was to verify if pregnancy complications induce postnatal alterations in gene expression of microRNAs associated with cardiovascular/cerebrovascular diseases. Twenty-nine microRNAs were assessed in peripheral blood, compared between groups, and analyzed in relation to both aspects, the current presence of cardiovascular risk factors and cardiovascular complications and the previous occurrence of pregnancy complications with regard to the clinical signs, dates of delivery, and Doppler ultrasound examination. The expression profile of miR-21-5p differed between controls and children with a history of uncomplicated pregnancies with abnormal clinical findings. Abnormal expression profile of multiple microRNAs was found in children affected with GH (miR-1-3p, miR-17-5p, miR-20a-5p, miR-21-5p, miR-23a-3p, miR-26a-5p, miR-29a-3p, miR-103a-3p, miR-125b-5p, miR-126-3p, miR-133a-3p, miR-146a-5p, miR-181a-5p, miR-195-5p, and miR-342-3p), PE (miR-1-3p, miR-20a-5p, miR-20b-5p, miR-103a-3p, miR-133a-3p, miR-342-3p), and FGR (miR-17-5p, miR-126-3p, miR-133a-3p). The index of pulsatility in the ductus venosus showed a strong positive correlation with miR-210-3p gene expression in children exposed to PE and/or FGR. Any of changes in epigenome (up-regulation of miR-1-3p and miR-133a-3p) that were induced by pregnancy complications are long-acting and may predispose children affected with GH, PE, or FGR to later development of cardiovascular/cerebrovascular diseases. Novel epigenetic changes (aberrant expression profile of microRNAs) appeared in a proportion of children that were exposed to GH, PE, or FGR. Screening of particular microRNAs may stratify a highly risky group of children that might benefit from implementation of early primary prevention strategies.
Collapse
Affiliation(s)
- Ilona Hromadnikova
- Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University, 10000 Prague, Czech Republic.
| | - Katerina Kotlabova
- Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University, 10000 Prague, Czech Republic.
| | - Lenka Dvorakova
- Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University, 10000 Prague, Czech Republic.
| | - Ladislav Krofta
- Institute for the Care of the Mother and Child, Third Faculty of Medicine, Charles University, 14700 Prague, Czech Republic.
| | - Jan Sirc
- Institute for the Care of the Mother and Child, Third Faculty of Medicine, Charles University, 14700 Prague, Czech Republic.
| |
Collapse
|
43
|
Dong Y, Xu W, Liu C, Liu P, Li P, Wang K. Reactive Oxygen Species Related Noncoding RNAs as Regulators of Cardiovascular Diseases. Int J Biol Sci 2019; 15:680-687. [PMID: 30745854 PMCID: PMC6367576 DOI: 10.7150/ijbs.30464] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) are a class of reactive molecules that have been implicated in a variety of cardiovascular diseases, accompanied by disorder of multiple signaling events. As cardiomyocytes maintain abundant of mitochondria, which supply the major source of endogenous ROS, oxidative damage to mitochondria often drives apoptotic cell death and initiates cardiac pathology. In recent years, non-coding RNAs (ncRNAs) have received much attention to uncover their roles in regulating gene expression during those pathological events in the heart, such as myocardial infarction, cardiac hypertrophy, and heart failure. Emerging evidences have highlighted that different ROS levels in response to diverse cardiac stresses result in differential expression of ncRNAs, subsequently altering the expression of pathogenetic genes. However, the knowledge about the ncRNA-linked ROS regulatory mechanisms in cardiac pathologies is still largely unexplored. In this review, we summarize the connections that exist among ROS, ncRNAs, and cardiac diseases to understand the interactions among the molecular entities underlying cardiac pathological events in the hopes of guiding novel therapies for heart diseases in the future.
Collapse
Affiliation(s)
- Yanhan Dong
- Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Wenhua Xu
- Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Cuiyun Liu
- Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Peijun Liu
- Biochemistry Department No.2 Middle School Qingdao Shandong P.R. China 266000
| | - Peifeng Li
- Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Kun Wang
- Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| |
Collapse
|
44
|
Rynning I, Arlt VM, Vrbova K, Neča J, Rossner Jr P, Klema J, Ulvestad B, Petersen E, Skare Ø, Haugen A, Phillips DH, Machala M, Topinka J, Mollerup S. Bulky DNA adducts, microRNA profiles, and lipid biomarkers in Norwegian tunnel finishing workers occupationally exposed to diesel exhaust. Occup Environ Med 2019; 76:10-16. [PMID: 30425118 PMCID: PMC6327869 DOI: 10.1136/oemed-2018-105445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/04/2018] [Accepted: 10/21/2018] [Indexed: 01/27/2023]
Abstract
OBJECTIVES This study aimed to assess the biological impact of occupational exposure to diesel exhaust (DE) including DE particles (DEP) from heavy-duty diesel-powered equipment in Norwegian tunnel finishing workers (TFW). METHODS TFW (n=69) and referents (n=69) were investigated for bulky DNA adducts (by 32P-postlabelling) and expression of microRNAs (miRNAs) (by small RNA sequencing) in peripheral blood mononuclear cells (PBMC), as well as circulating free arachidonic acid (AA) and eicosanoid profiles in plasma (by liquid chromatography-tandem mass spectrometry). RESULTS PBMC from TFW showed significantly higher levels of DNA adducts compared with referents. Levels of DNA adducts were also related to smoking habits. Seventeen miRNAs were significantly deregulated in TFW. Several of these miRNAs are related to carcinogenesis, apoptosis and antioxidant effects. Analysis of putative miRNA-gene targets revealed deregulation of pathways associated with cancer, alterations in lipid molecules, steroid biosynthesis and cell cycle. Plasma profiles showed higher levels of free AA and 15-hydroxyeicosatetraenoic acid, and lower levels of prostaglandin D2 and 9-hydroxyoctadecadienoic acid in TFW compared with referents. CONCLUSION Occupational exposure to DE/DEP is associated with biological alterations in TFW potentially affecting lung homoeostasis, carcinogenesis, inflammation status and the cardiovascular system. Of particular importance is the finding that tunnel finishing work is associated with an increased level of DNA adducts formation in PBMC.
Collapse
Affiliation(s)
- Iselin Rynning
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London, UK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King’s College London in Partnership with Public Health England and Imperial College London, London, UK
| | - Kristyna Vrbova
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Neča
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Pavel Rossner Jr
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Klema
- Department of Computer Science, Czech Technical University in Prague, Prague, Czech Republic
| | - Bente Ulvestad
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Elisabeth Petersen
- Department of Work Psychology and Physiology, National Institute of Occupational Health, Oslo, Norway
| | - Øivind Skare
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Aage Haugen
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London, UK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King’s College London in Partnership with Public Health England and Imperial College London, London, UK
| | - Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Jan Topinka
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Steen Mollerup
- Section for Toxicology and Biological Work Environment, Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| |
Collapse
|
45
|
Zhou T, Qin G, Yang L, Xiang D, Li S. LncRNA XIST regulates myocardial infarction by targeting miR-130a-3p. J Cell Physiol 2018; 234:8659-8667. [PMID: 29226319 DOI: 10.1002/jcp.26327] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/29/2017] [Indexed: 12/25/2022]
Abstract
The study was used to probe long noncoding RNA X-inactive specific transcript (lncRNA XIST) RNA expression profile and its influence on cell cycle, proliferation, and apoptosis in myocardial cells. We also aimed to explore the possible meditating relationship between XIST, PDE4D, and miR-130a-3p. Gene differential analysis was carried out using human lncRNA Microarray V3.0. quantitative real-time PCR was used to test mRNA expressions of XIST, miR-130a-3p, and PDE4D in normal cells and postmyocardial infarction (MI) cells. Western blot was applied to determine the protein expression profile of PED4D. Changes in viability and cell cycle/apoptosis of post-MI myocardial cells after silencing of XIST or PDE4D were investigated by MTT assay and flow cytometry, respectively. The targeting relationship between miR-130a-3p and XIST, PDE4D in myocardial cells were verified by dual luciferase reporter assay. Simulated MI environment was constructed by performing anoxic preconditioning in normal cells to probe the influence of XIST on myocardial cell apoptosis. XIST and PDE4D were overexpressed in post-MI myocardial cells, whereas miR-130a-3p was underexpressed in post-MI myocardial cells. High-expressed XIST and PDE4D both promoted myocardial cell apoptosis. High-expressed XIST also inhibited myocardial cell proliferation. XIST-downregulated miR-130a-3p and PDE4D was a direct target of miR-130a-3p. LncRNA XIST promotes MI by targeting miR-130a-3p. MI induced by PDE4D can be reversed by miR-130a-3p.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Cardiac Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Guowei Qin
- Department of Electrocardiogram, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Liehong Yang
- Department of Cardiac Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Daokang Xiang
- Department of Cardiac Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Suining Li
- Department of Cardiac Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
46
|
Witvrouwen I, Gevaert AB, Van Craenenbroeck EM, Van Craenenbroeck AH. MicroRNA Isolation from Plasma for Real-Time qPCR Array. ACTA ACUST UNITED AC 2018; 99:e69. [DOI: 10.1002/cphg.69] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Isabel Witvrouwen
- Laboratory of Cellular and Molecular Cardiology, Research Group Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp; Antwerp Belgium
- Department of Cardiology, Antwerp University Hospital (UZA); Edegem Belgium
| | - Andreas B. Gevaert
- Laboratory of Cellular and Molecular Cardiology, Research Group Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp; Antwerp Belgium
- Department of Cardiology, Antwerp University Hospital (UZA); Edegem Belgium
| | - Emeline M. Van Craenenbroeck
- Laboratory of Cellular and Molecular Cardiology, Research Group Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp; Antwerp Belgium
- Department of Cardiology, Antwerp University Hospital (UZA); Edegem Belgium
| | - Amaryllis H. Van Craenenbroeck
- Laboratory of Cellular and Molecular Cardiology, Research Group Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp; Antwerp Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp; Antwerp Belgium
- Department of Nephrology, Antwerp University Hospital (UZA); Edegem Belgium
| |
Collapse
|
47
|
Su Y, Tian H, Wei L, Fu G, Sun T. Integrin β3 inhibits hypoxia-induced apoptosis in cardiomyocytes. Acta Biochim Biophys Sin (Shanghai) 2018; 50:658-665. [PMID: 29800236 DOI: 10.1093/abbs/gmy056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Indexed: 01/01/2023] Open
Abstract
Hypoxia-induced apoptosis plays an important role in cardiovascular diseases. Integrin β3 is one of the main integrin heterodimer receptors on the surface of cardiac myocytes. However, despite the important role that integrin β3 plays in the cardiovascular disease, its exact role in the hypoxia response remains unclear. Hence, in the present investigation we aimed to study the role of integrin β3 in hypoxia-induced apoptosis in H9C2 cells and primary rat myocardial cells. MTT assay, flow cytometry and TUNEL assay results showed that hypoxia inhibited cardiomyocyte proliferation and induced cardiomyocyte apoptosis. The expression levels of integrin β3 and HIF1α were upregulated in hypoxia-induced cardiomyocytes as revealed by real-time PCR and western blot analysis. Furthermore, knockdown of integrin β3 expression by siRNA increased hypoxia-induced cardiomyocyte apoptosis. In addition, integrin β3 overexpression weakened hypoxia-induced cardiomyocyte apoptosis. The protein expressions of integrin β3 and HIF1α were upregulated in acute myocardial infarction rat cardiac tissues compared with the control rat cardiac tissues. Our data suggest that integrin β3 plays a protective role in cardiomyocytes during hypoxia-induced apoptosis.
Collapse
Affiliation(s)
- Yifan Su
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lijiang Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guohui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Kinoshita C, Aoyama K, Nakaki T. Neuroprotection afforded by circadian regulation of intracellular glutathione levels: A key role for miRNAs. Free Radic Biol Med 2018; 119:17-33. [PMID: 29198727 DOI: 10.1016/j.freeradbiomed.2017.11.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 01/17/2023]
Abstract
Circadian rhythms are approximately 24-h oscillations of physiological and behavioral processes that allow us to adapt to daily environmental cycles. Like many other biological functions, cellular redox status and antioxidative defense systems display circadian rhythmicity. In the central nervous system (CNS), glutathione (GSH) is a critical antioxidant because the CNS is extremely vulnerable to oxidative stress; oxidative stress, in turn, causes several fatal diseases, including neurodegenerative diseases. It has long been known that GSH level shows circadian rhythm, although the mechanism underlying GSH rhythm production has not been well-studied. Several lines of recent evidence indicate that the expression of antioxidant genes involved in GSH homeostasis as well as circadian clock genes are regulated by post-transcriptional regulator microRNA (miRNA), indicating that miRNA plays a key role in generating GSH rhythm. Interestingly, several reports have shown that alterations of miRNA expression as well as circadian rhythm have been known to link with various diseases related to oxidative stress. A growing body of evidence implicates a strong correlation between antioxidative defense, circadian rhythm and miRNA function, therefore, their dysfunctions could cause numerous diseases. It is hoped that continued elucidation of the antioxidative defense systems controlled by novel miRNA regulation under circadian control will advance the development of therapeutics for the diseases caused by oxidative stress.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Toshio Nakaki
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| |
Collapse
|