1
|
Park M, Cho S, Jeong D. Restoration of Sestrin 3 Expression Mitigates Cardiac Oxidative Damage in Ischemia-Reperfusion Injury Model. Antioxidants (Basel) 2025; 14:61. [PMID: 39857395 PMCID: PMC11763094 DOI: 10.3390/antiox14010061] [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: 12/12/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Cardiac ischemia-reperfusion injury (IRI) occurs when blood flow is restored to the myocardium after a period of ischemia, leading to oxidative stress and subsequent myocardial cell damage, primarily due to the accumulation of reactive oxygen species (ROS). In our previous research, we identified that miR-25 is significantly overexpressed in pressure overload-induced heart failure, and its inhibition improves cardiac function by restoring the expression of SERCA2a, a key protein involved in calcium regulation. In this study, we aimed to investigate the role of miR-25 in the context of ischemia-reperfusion injury. We found that miR-25 was markedly upregulated under hypoxic conditions in both in vitro and in vivo models. Through in silico analysis, we identified Sestrin3 (SESN3), an antioxidant protein known for its protective effects against oxidative stress, as a novel target of miR-25. Based on these findings, we hypothesized that inhibiting miR-25 would restore Sestrin3 expression, thereby reducing ROS-induced myocardial cell damage and improving cardiac function. To test this hypothesis, we employed two model systems: a hypoxia/reoxygenation (H/R) stress model using H9c2 myoblasts and a surgically induced ischemia-reperfusion injury mouse model. Our results demonstrated that the use of miR-25 inhibitors significantly improved cardiac function and reduced myocardial damage in both models through the restoration of SESN3 expression. In conclusion, our findings suggest that targeting miR-25 may serve as a novel therapeutic modality to alleviate oxidative damage in the heart.
Collapse
Affiliation(s)
| | | | - Dongtak Jeong
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University—ERICA, Ansan 15588, Republic of Korea; (M.P.); (S.C.)
| |
Collapse
|
2
|
Turkieh A, Beseme O, Saura O, Charrier H, Michel JB, Amouyel P, Thum T, Bauters C, Pinet F. LIPCAR levels in plasma-derived extracellular vesicles is associated with left ventricle remodeling post-myocardial infarction. J Transl Med 2024; 22:31. [PMID: 38184604 PMCID: PMC10771704 DOI: 10.1186/s12967-023-04820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/20/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Long Intergenic noncoding RNA predicting CARdiac remodeling (LIPCAR) is a long noncoding RNA identified in plasma of patients after myocardial infarction (MI) to be associated with left ventricle remodeling (LVR). LIPCAR was also shown to be a predictor of early death in heart failure (HF) patients. However, no information regarding the expression of LIPCAR and its function in heart as well as the mechanisms involved in its transport to the circulation is known. The aims of this study are (1) to characterize the transporter of LIPCAR from heart to circulation; (2) to determine whether LIPCAR levels in plasma isolated-extracellular vesicles (EVs) reflect the alteration of its expression in total plasma and could be used as biomarkers of LVR post-MI. METHODS Since expression of LIPCAR is restricted to human species and the limitation of availability of cardiac biopsy samples, serum-free conditioned culture media from HeLa cells were first used to characterize the extracellular transporter of LIPCAR before validation in EVs isolated from human cardiac biopsies (non-failing and ischemic HF patients) and plasma samples (patients who develop or not LVR post-MI). Differential centrifugation at 20,000g and 100,000g were performed to isolate the large (lEVs) and small EVs (sEVs), respectively. Western blot and nanoparticle tracking (NTA) analysis were used to characterize the isolated EVs. qRT-PCR analysis was used to quantify LIPCAR in all samples. RESULTS We showed that LIPCAR is present in both lEVs and sEVs isolated from all samples. The levels of LIPCAR are higher in lEVs compared to sEVs isolated from HeLa conditioned culture media and cardiac biopsies. No difference of LIPCAR expression was observed in tissue or EVs isolated from cardiac biopsies obtained from ischemic HF patients compared to non-failing patients. Interestingly, LIPCAR levels were increased in lEVs and sEVs isolated from MI patients who develop LVR compared to patients who did not develop LVR. CONCLUSION Our data showed that large EVs are the main extracellular vesicle transporter of LIPCAR from heart into the circulation independently of the status, non-failing or HF, in patients. The levels of LIPCAR in EVs isolated from plasma could be used as biomarkers of LVR in post-MI patients.
Collapse
Affiliation(s)
- Annie Turkieh
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France.
| | - Olivia Beseme
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Ouriel Saura
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Henri Charrier
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | | | - Philippe Amouyel
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christophe Bauters
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France
| | - Florence Pinet
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167- RID-AGE, Université de Lille, Lille, France.
| |
Collapse
|
3
|
Soliman AG, Mahmoud B, Eldin ZE, El-Shahawy AAG, Abdel-Gabbar M. Optimized synthesis characterization and protective activity of quercetin and quercetin–chitosan nanoformula against cardiotoxicity that was induced in male Wister rats via anticancer agent: doxorubicin. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
AbstractThe study’s goal was to look into the protective properties of quercetin (QU) in natural form and QU nanoparticles-loaded chitosan nanoparticles (QU-CHSNPs) against cardiotoxicity. The ionotropic gelation approach was adopted to form QU-CHSNPs. The characterizations were performed using advanced techniques. In vitro, the release profile of QU was studied. Cardiotoxicity was induced by doxorubicin (DOX) and protected via concurrent administration of QU and QU-CHSNPs. The heart's preventive effects of QU and QU-CHSNPs were manifested by a decrease in elevated serum activities of cardiac enzymes, as well as an improvement in the heart's antioxidant defence system and histological changes. The findings substantiated QU-CHSNPs' structure with an entrapment efficiency of 92.56%. The mean of the zeta size distribution was 150 nm, the real average particle size was 50 nm, and the zeta potential value was − 27.9 mV, exhibiting low physical stability. The percent of the free QU-cumulative release was about 70% after 12 h, and QU-CHSNPs showed a 49% continued release with a pattern of sustained release, reaching 98% after 48 h. And as such, QU and QU-CHSNPs restrained the induced cardiotoxicity of DOX in male Wistar rats, with the QU-CHSNPs being more efficient.
Collapse
|
4
|
Maries L, Moatar AI, Sala-Cirtog M, Sima L, Anghel A, Marian C, Chis AR, Sirbu IO. Clinical Variables Influence the Ability of miR-101, miR-150, and miR-21 to Predict Ventricular Remodeling after ST-Elevation Myocardial Infarction. Biomedicines 2023; 11:2738. [PMID: 37893111 PMCID: PMC10604279 DOI: 10.3390/biomedicines11102738] [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: 09/13/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Left ventricle remodeling (LVR) after acute myocardial infarction (MI) leads to impairment of both systolic and diastolic function, a significant contributor to heart failure (HF). Despite extensive research in the field, predicting post-MI LVR and HF is still a challenge. Several circulant microRNAs have been proposed as LVR predictors; however, their clinical value is controversial. Here, we used real-time quantitative PCR to quantify the plasma levels of hsa-miR-101, hsa-miR-150, and hsa-miR-21 on the first day of hospital admission of MI patients with ST-elevation (STEMI). We analyzed their correlation to the patient's clinical and paraclinical variables and evaluated their ability to discriminate between post-MI LVR and non-LVR. We show that, despite being excellent MI discriminators, none of these microRNAs can distinguish between LVR and non-LVR patients. Furthermore, we found that diabetes mellitus (DM), Hb level, and the number of erythrocytes significantly influence all three plasma microRNA levels. This suggests that plasma microRNAs' diagnostic and prognostic value in STEMI patients should be reevaluated and interpreted in the context of associated pathologies.
Collapse
Affiliation(s)
- Liana Maries
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Alexandra Ioana Moatar
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Maria Sala-Cirtog
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Laurentiu Sima
- Surgical Semiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Andrei Anghel
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
| | - Catalin Marian
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Aimee Rodica Chis
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ioan-Ovidiu Sirbu
- Biochemistry Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (L.M.); (A.I.M.); (M.S.-C.); (A.A.); (C.M.); (I.-O.S.)
- Center for Complex Network Science, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
5
|
Swiderski J, Sakkal S, Apostolopoulos V, Zulli A, Gadanec LK. Combination of Taurine and Black Pepper Extract as a Treatment for Cardiovascular and Coronary Artery Diseases. Nutrients 2023; 15:nu15112562. [PMID: 37299525 DOI: 10.3390/nu15112562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The shift in modern dietary regimens to "Western style" and sedentary lifestyles are believed to be partly responsible for the increase in the global burden of cardiovascular diseases. Natural products have been used throughout human history as treatments for a plethora of pathological conditions. Taurine and, more recently, black pepper have gained attention for their beneficial health effects while remaining non-toxic even when ingested in excess. Taurine, black pepper, and the major terpene constituents found in black pepper (i.e., β-caryophyllene; α-pinene; β-pinene; α-humulene; limonene; and sabinene) that are present in PhytoCann BP® have been shown to have cardioprotective effects based on anti-inflammatory, antioxidative, anti-hypertensive and anti-atherosclerotic mechanisms. This comprehensive review of the literature focuses on determining whether the combination of taurine and black pepper extract is an effective natural treatment for reducing cardiovascular diseases risk factors (i.e., hypertension and hyperhomocysteinemia) and for driving anti-inflammatory, antioxidative and anti-atherosclerotic mechanisms to combat coronary artery disease, heart failure, myocardial infarction, and atherosclerotic disease.
Collapse
Affiliation(s)
- Jordan Swiderski
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| |
Collapse
|
6
|
Roberts JA, Rainbow RD, Sharma P. Mitigation of Cardiovascular Disease and Toxicity through NRF2 Signalling. Int J Mol Sci 2023; 24:ijms24076723. [PMID: 37047696 PMCID: PMC10094784 DOI: 10.3390/ijms24076723] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Cardiovascular toxicity and diseases are phenomena that have a vastly detrimental impact on morbidity and mortality. The pathophysiology driving the development of these conditions is multifactorial but commonly includes the perturbance of reactive oxygen species (ROS) signalling, iron homeostasis and mitochondrial bioenergetics. The transcription factor nuclear factor erythroid 2 (NFE2)-related factor 2 (NRF2), a master regulator of cytoprotective responses, drives the expression of genes that provide resistance to oxidative, electrophilic and xenobiotic stresses. Recent research has suggested that stimulation of the NRF2 signalling pathway can alleviate cardiotoxicity and hallmarks of cardiovascular disease progression. However, dysregulation of NRF2 dynamic responses can be severely impacted by ageing processes and off-target toxicity from clinical medicines including anthracycline chemotherapeutics, rendering cells of the cardiovascular system susceptible to toxicity and subsequent tissue dysfunction. This review addresses the current understanding of NRF2 mechanisms under homeostatic and cardiovascular pathophysiological conditions within the context of wider implications for this diverse transcription factor.
Collapse
Affiliation(s)
- James A. Roberts
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| | - Parveen Sharma
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| |
Collapse
|
7
|
Luo Q, Tang S, Xiao X, Wei Y, Cheng B, Huang Y, Zhong K, Tian G, Lu H. Benomyl-induced development and cardiac toxicity in zebrafish embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:33090-33100. [PMID: 36471152 DOI: 10.1007/s11356-022-24213-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
Benomyl is a highly effective broad-spectrum fungicide widely used worldwide to control vegetable, fruit, and oil crop diseases. However, the mechanism of its toxicity to aquatic organisms and humans remains unknown. In this study, zebrafish were used to determine the toxicity of benomyl. It was found to be highly toxic, with a 72-h post-fertilization (hpf) lethal concentration 50 (LC50) of 1.454 mg/L. Benomyl induced severe developmental toxicity, including shorter body length, slower heart rate, and a reduced yolk absorption rate. Benomyl also increased oxidative stress in zebrafish, especially in the heart and head, as well as increasing malondialdehyde (MDA) content and decreasing catalase (CAT) and superoxide dismutase (SOD) activities. This indicates that benomyl induced reactive oxygen species (ROS) production and cell membrane peroxidation in vivo. Acridine orange (AO) staining and apoptosis factor detection further indicated that benomyl induced apoptosis in zebrafish. Overall, these findings demonstrate that benomyl disrupts cellular homeostasis by activating oxidative stress in zebrafish, resulting in an imbalance of cardiac development-related gene expression and apoptosis, which causes severe developmental toxicity and cardiac dysfunction. This study evaluated the in vivo toxicity of benomyl, which is a potential threat to aquatic organisms and humans. Possible toxicity mechanisms are explored, providing a valuable reference for the safe use of benomyl.
Collapse
Affiliation(s)
- Qiang Luo
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Shuqiong Tang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Xiaoping Xiao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
- Provincal Key Laboratory of Low-Carbon Solid Waste Recycling, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - You Wei
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
- College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Bo Cheng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yong Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
- College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Guiyou Tian
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Huiqiang Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
8
|
Zabielska-Kaczorowska MA, Bogucka AE, Macur K, Czaplewska P, Watson SA, Perbellini F, Terracciano CM, Smolenski RT. Label-free quantitative SWATH-MS proteomic analysis of adult myocardial slices in vitro after biomimetic electromechanical stimulation. Sci Rep 2022; 12:16533. [PMID: 36192624 PMCID: PMC9529937 DOI: 10.1038/s41598-022-20494-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
A special in vitro model maintained with ultrathin cardiac slices with a preserved architecture, multi-cellularity, and physiology of the heart tissue was used. In our experiments, we performed label-free quantitative SWATH-MS proteomic analysis of the adult myocardial slices in vitro after biomimetic electromechanical stimulation. Rat myocardial slices were stretched to sarcomere lengths (SL) within the physiological range of 1.8–2.2 μm. Electromechanically stimulated slices were compared with slices cultured without electromechanical stimulation (unloaded and nonstimulated-TW) on a liquid–air interface and with fresh myocardial slices (0 h-C). Quantitative (relative) proteomic analyses were performed using a label-free SWATH-MS technique on a high-resolution microLC-MS/MS TripleTOF 5600+ system (SCIEX). The acquired MS/MS spectra from the DDA LC–MS/MS analyses of the rat heart samples were searched against the UniProt Rattus norvegicus database (version of 15.05.2018) using the Paragon algorithm incorporated into ProteinPilot 4.5 (SCIEX) software. The highest number of differential proteins was observed in the TW group—121 when compared to the C group. In the 1.8 and 2.2 groups, 79 and 52 proteins present at a significantly different concentration from the control samples were found, respectively. A substantial fraction of these proteins were common for two or more comparisons, resulting in a list of 169 significant proteins for at least one of the comparisons. This study found the most prominent changes in the proteomic pattern related to mitochondrial respiration, energy metabolism, and muscle contraction in the slices that were stretched and fresh myocardial slices cultured without electromechanical stimulation.
Collapse
Affiliation(s)
- M A Zabielska-Kaczorowska
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland. .,Department of Physiology, Medical University of Gdansk, Gdansk, Poland.
| | - A E Bogucka
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland.,Institute of Biochemistry, Medical Faculty, Justus Liebig University of Giessen, Giessen, Germany
| | - K Macur
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - P Czaplewska
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - S A Watson
- National Heart & Lung Institute, Imperial College London, London, UK
| | - F Perbellini
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - C M Terracciano
- National Heart & Lung Institute, Imperial College London, London, UK
| | - R T Smolenski
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
9
|
Hormesis and Oxidative Distress: Pathophysiology of Reactive Oxygen Species and the Open Question of Antioxidant Modulation and Supplementation. Antioxidants (Basel) 2022; 11:antiox11081613. [PMID: 36009331 PMCID: PMC9405171 DOI: 10.3390/antiox11081613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Alterations of redox homeostasis leads to a condition of resilience known as hormesis that is due to the activation of redox-sensitive pathways stimulating cell proliferation, growth, differentiation, and angiogenesis. Instead, supraphysiological production of reactive oxygen species (ROS) exceeds antioxidant defence and leads to oxidative distress. This condition induces damage to biomolecules and is responsible or co-responsible for the onset of several chronic pathologies. Thus, a dietary antioxidant supplementation has been proposed in order to prevent aging, cardiovascular and degenerative diseases as well as carcinogenesis. However, this approach has failed to demonstrate efficacy, often leading to harmful side effects, in particular in patients affected by cancer. In this latter case, an approach based on endogenous antioxidant depletion, leading to ROS overproduction, has shown an interesting potential for enhancing susceptibility of patients to anticancer therapies. Therefore, a deep investigation of molecular pathways involved in redox balance is crucial in order to identify new molecular targets useful for the development of more effective therapeutic approaches. The review herein provides an overview of the pathophysiological role of ROS and focuses the attention on positive and negative aspects of antioxidant modulation with the intent to find new insights for a successful clinical application.
Collapse
|
10
|
Panda P, Verma HK, Lakkakula S, Merchant N, Kadir F, Rahman S, Jeffree MS, Lakkakula BVKS, Rao PV. Biomarkers of Oxidative Stress Tethered to Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9154295. [PMID: 35783193 PMCID: PMC9249518 DOI: 10.1155/2022/9154295] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) is a broad term that incorporated a group of conditions that affect the blood vessels and the heart. CVD is a foremost cause of fatalities around the world. Multiple pathophysiological mechanisms are involved in CVD; however, oxidative stress plays a vital role in generating reactive oxygen species (ROS). Oxidative stress occurs when the concentration of oxidants exceeds the potency of antioxidants within the body while producing reactive nitrogen species (RNS). ROS generated by oxidative stress disrupts cell signaling, DNA damage, lipids, and proteins, thereby resulting in inflammation and apoptosis. Mitochondria is the primary source of ROS production within cells. Increased ROS production reduces nitric oxide (NO) bioavailability, which elevates vasoconstriction within the arteries and contributes to the development of hypertension. ROS production has also been linked to the development of atherosclerotic plaque. Antioxidants can decrease oxidative stress in the body; however, various therapeutic drugs have been designed to treat oxidative stress damage due to CVD. The present review provides a detailed narrative of the oxidative stress and ROS generation with a primary focus on the oxidative stress biomarker and its association with CVD. We have also discussed the complex relationship between inflammation and endothelial dysfunction in CVD as well as oxidative stress-induced obesity in CVD. Finally, we discussed the role of antioxidants in reducing oxidative stress in CVD.
Collapse
Affiliation(s)
- Poojarani Panda
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lung Biology and Disease, Helmholtz Zentrum, 85764 Neuherberg, Munich, Germany
| | | | - Neha Merchant
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali, 304022 Rajasthan, India
| | - Fairrul Kadir
- Department of Emergency Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Shamsur Rahman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Mohammad Saffree Jeffree
- Department of Community and Family Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, 88400 Sabah, Malaysia
| | | | - Pasupuleti Visweswara Rao
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Abdurrab University, Jalan Riau Ujung No. 73, Pekanbaru, 28292 Riau, Indonesia
- Centre for International Relations and Research Collaborations, Reva University, Rukmini Knowledge Park, Kattigenahalli, Yelahanka, Bangalore, 560064 Karnataka, India
| |
Collapse
|
11
|
Jusic A, Thomas PB, Wettinger SB, Dogan S, Farrugia R, Gaetano C, Tuna BG, Pinet F, Robinson EL, Tual-Chalot S, Stellos K, Devaux Y. Noncoding RNAs in age-related cardiovascular diseases. Ageing Res Rev 2022; 77:101610. [PMID: 35338919 DOI: 10.1016/j.arr.2022.101610] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/28/2022] [Accepted: 03/15/2022] [Indexed: 11/01/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality in the adult population worldwide and represent a severe economic burden and public health concern. The majority of human genes do not code for proteins. However, noncoding transcripts play important roles in ageing that significantly increases the risk for CVDs. Noncoding RNAs (ncRNAs) are critical regulators of multiple biological processes related to ageing such as oxidative stress, mitochondrial dysfunction and chronic inflammation. NcRNAs are also involved in pathophysiological developments within the cardiovascular system including arrhythmias, cardiac hypertrophy, fibrosis, myocardial infarction and heart failure. In this review article, we cover the roles of ncRNAs in cardiovascular ageing and disease as well as their potential therapeutic applications in CVDs.
Collapse
|
12
|
Peugnet V, Chwastyniak M, Mulder P, Lancel S, Bultot L, Fourny N, Renguet E, Bugger H, Beseme O, Loyens A, Heyse W, Richard V, Amouyel P, Bertrand L, Pinet F, Dubois-Deruy E. Mitochondrial-Targeted Therapies Require Mitophagy to Prevent Oxidative Stress Induced by SOD2 Inactivation in Hypertrophied Cardiomyocytes. Antioxidants (Basel) 2022; 11:antiox11040723. [PMID: 35453408 PMCID: PMC9029275 DOI: 10.3390/antiox11040723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 12/24/2022] Open
Abstract
Heart failure, mostly associated with cardiac hypertrophy, is a major cause of illness and death. Oxidative stress causes accumulation of reactive oxygen species (ROS), leading to mitochondrial dysfunction, suggesting that mitochondria-targeted therapies could be effective in this context. The purpose of this work was to determine whether mitochondria-targeted therapies could improve cardiac hypertrophy induced by mitochondrial ROS. We used neonatal (NCMs) and adult (ACMs) rat cardiomyocytes hypertrophied by isoproterenol (Iso) to induce mitochondrial ROS. A decreased interaction between sirtuin 3 and superoxide dismutase 2 (SOD2) induced SOD2 acetylation on lysine 68 and inactivation, leading to mitochondrial oxidative stress and dysfunction and hypertrophy after 24 h of Iso treatment. To counteract these mechanisms, we evaluated the impact of the mitochondria-targeted antioxidant mitoquinone (MitoQ). MitoQ decreased mitochondrial ROS and hypertrophy in Iso-treated NCMs and ACMs but altered mitochondrial structure and function by decreasing mitochondrial respiration and mitophagy. The same decrease in mitophagy was found in human cardiomyocytes but not in fibroblasts, suggesting a cardiomyocyte-specific deleterious effect of MitoQ. Our data showed the importance of mitochondrial oxidative stress in the development of cardiomyocyte hypertrophy. We observed that targeting mitochondria by MitoQ in cardiomyocytes impaired the metabolism through defective mitophagy, leading to accumulation of deficient mitochondria.
Collapse
Affiliation(s)
- Victoriane Peugnet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
| | - Maggy Chwastyniak
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
| | - Paul Mulder
- Normandie Univ, UNIROUEN, Inserm U1096, FHU-REMOD-HF, 76000 Rouen, France; (P.M.); (V.R.)
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
| | - Laurent Bultot
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, UCLouvain, 1200 Bruxelles, Belgium; (L.B.); (N.F.); (E.R.); (L.B.)
| | - Natacha Fourny
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, UCLouvain, 1200 Bruxelles, Belgium; (L.B.); (N.F.); (E.R.); (L.B.)
| | - Edith Renguet
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, UCLouvain, 1200 Bruxelles, Belgium; (L.B.); (N.F.); (E.R.); (L.B.)
| | - Heiko Bugger
- Department of Cardiology and Angiology I, Heart Center Freiburg, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany;
| | - Olivia Beseme
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
| | - Anne Loyens
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut de Recherche Contre le Cancer de Lille, UMR9020-UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France;
| | - Wilfried Heyse
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
| | - Vincent Richard
- Normandie Univ, UNIROUEN, Inserm U1096, FHU-REMOD-HF, 76000 Rouen, France; (P.M.); (V.R.)
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, UCLouvain, 1200 Bruxelles, Belgium; (L.B.); (N.F.); (E.R.); (L.B.)
| | - Florence Pinet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
- Correspondence: (F.P.); (E.D.-D.); Tel.: +33-(0)3-20-87-72-15 (F.P.); +33-(0)3-20-87-73-62 (E.D.-D.)
| | - Emilie Dubois-Deruy
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, 59000 Lille, France; (V.P.); (M.C.); (S.L.); (O.B.); (W.H.); (P.A.)
- Correspondence: (F.P.); (E.D.-D.); Tel.: +33-(0)3-20-87-72-15 (F.P.); +33-(0)3-20-87-73-62 (E.D.-D.)
| |
Collapse
|
13
|
Turkieh A, El Masri Y, Pinet F, Dubois-Deruy E. Mitophagy Regulation Following Myocardial Infarction. Cells 2022; 11:cells11020199. [PMID: 35053316 PMCID: PMC8774240 DOI: 10.3390/cells11020199] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023] Open
Abstract
Mitophagy, which mediates the selective elimination of dysfunctional mitochondria, is essential for cardiac homeostasis. Mitophagy is regulated mainly by PTEN-induced putative kinase protein-1 (PINK1)/parkin pathway but also by FUN14 domain-containing 1 (FUNDC1) or Bcl2 interacting protein 3 (BNIP3) and BNIP3-like (BNIP3L/NIX) pathways. Several studies have shown that dysregulated mitophagy is involved in cardiac dysfunction induced by aging, aortic stenosis, myocardial infarction or diabetes. The cardioprotective role of mitophagy is well described, whereas excessive mitophagy could contribute to cell death and cardiac dysfunction. In this review, we summarize the mechanisms involved in the regulation of cardiac mitophagy and its role in physiological condition. We focused on cardiac mitophagy during and following myocardial infarction by highlighting the role and the regulation of PI NK1/parkin-; FUNDC1-; BNIP3- and BNIP3L/NIX-induced mitophagy during ischemia and reperfusion.
Collapse
|
14
|
Thai SF, Jones CP, Robinette BL, Ren H, Vallanat B, Fisher AA, Kitchin KT. Effects of Silver Nanoparticles and Silver Nitrate on mRNA and microRNA Expression in Human Hepatocellular Carcinoma Cells (HepG2). JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:5414-5428. [PMID: 33980351 PMCID: PMC10563035 DOI: 10.1166/jnn.2021.19481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In order to understand toxicity of nano silver, human hepatocellular carcinoma (HepG2) cells were treated either with silver nitrate (AgNO₃) or with nano silver capped with glutathione (Ag-S) at various concentration. Differentially expressed genelists for mRNA and microRNA were obtained through Illumina RNA sequencing and DEseq data analyses. Both treatments showed non-linear dose response relationships for mRNA and microRNA. Gene expression analysis showed signaling pathways common to both nano Ag-S and AgNO₃, such as cell cycle regulation, DNA damage response and cancer related pathways. But, nano Ag-S caused signaling pathway changes that were not altered by AgNO₃ such as NRF2-mediated oxidative stress response inflammation, cell membrane signaling, and cell proliferation. Nano Ag-S also affected p53 signaling, survival, apoptosis, tissue repair, lipid synthesis, angiogenesis, liver fibrosis and tumor development. Several of the pathways affected by nano Ag-S are hypothesized as major contributors to nanotoxicity. MicroRNA target filter analysis revealed additional affected pathways that were not reflected in the mRNA expression response alone, including DNA damage signaling, genomic stability, ROS, cell cycle, ubiquitination, DNA methylation, cell proliferation and fibrosis for AgNO₃; and cell cycle regulation, P53 signaling, cell proliferation, survival, apoptosis, tissue repair and so on for nano Ag-S. These pathways may be mediated by microRNA repression of protein translation.Our study clearly showed that the addition of microRNA profiling increased the numbers of signaling pathways discovered that affected by the treatments on HepG2 cells and gave US a better picture of the effects of these reagents in the cells.
Collapse
Affiliation(s)
- Sheau-Fung Thai
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 TWAlexander Dr, Durham NC 27709, USA
| | - Carlton P Jones
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 TWAlexander Dr, Durham NC 27709, USA
| | - Brian L Robinette
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, 109 TWAlexander Dr, Durham NC 27709, USA
| | - Hongzu Ren
- Center for Public Health and Environmental Assessment, US Environmental Production Agency, 109 TW Alexander Dr., Durham NC 27709, USA
| | | | - Anna A Fisher
- Center for Public Health and Environmental Assessment, US Environmental Production Agency, 109 TW Alexander Dr., Durham NC 27709, USA
| | - Kirk T Kitchin
- US Environmental Protection Agency, Retired from EPA, Durham NC 27709, USA
| |
Collapse
|
15
|
MicroRNAs-The Heart of Post-Myocardial Infarction Remodeling. Diagnostics (Basel) 2021; 11:diagnostics11091675. [PMID: 34574016 PMCID: PMC8469128 DOI: 10.3390/diagnostics11091675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is one of the most frequent cardiac emergencies, with significant potential for mortality. One of the major challenges of the post-MI healing response is that replacement fibrosis could lead to left ventricular remodeling (LVR) and heart failure (HF). This process involves canonical and non-canonical transforming growth factor-beta (TGF-β) signaling pathways translating into an intricate activation of cardiac fibroblasts and disproportionate collagen synthesis. Accumulating evidence has indicated that microRNAs (miRNAs) significantly contribute to the modulation of these signaling pathways. This review summarizes the recent updates regarding the molecular mechanisms underlying the role of the over 30 miRNAs involved in post-MI LVR. In addition, we compare the contradictory roles of several multifunctional miRNAs and highlight their potential use in pressure overload and ischemia-induced fibrosis. Finally, we discuss their attractive role as prognostic biomarkers for HF, highlighting the most relevant human trials involving these miRNAs.
Collapse
|
16
|
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
|
17
|
Combined detection of miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 for screening of early heart failure diseases. Biosci Rep 2021; 40:222270. [PMID: 32124924 PMCID: PMC7080642 DOI: 10.1042/bsr20191653] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 02/17/2020] [Accepted: 03/01/2020] [Indexed: 12/16/2022] Open
Abstract
The use of circulating microRNAs as biomarkers opens up new opportunities for the diagnosis of cardiovascular diseases because of their specific expression profiles. The aim of the present study was to identify circulating microRNAs in human plasma as potential biomarkers of heart failure and related diseases. We used real-time quantitative PCR to screen microRNA in plasma samples from 62 normal controls and 62 heart failure samples. We found that circulating miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 expressed differently between healthy controls and heart failure patients. Plasma levels of miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 were unaffected by hemolysis. Correlation analysis showed any two of these miRNAs possess a strong correlation, indicating a possibility of combined analysis. MiR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 could be combined in two or three or more combinations. The results suggest that miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 may be a new diagnostic biomarker for heart failure and related diseases.
Collapse
|
18
|
Cruz MS, da Silva AMG, de Souza KSC, Luchessi AD, Silbiger VN. miRNAs emerge as circulating biomarkers of post-myocardial infarction heart failure. Heart Fail Rev 2021; 25:321-329. [PMID: 31254148 DOI: 10.1007/s10741-019-09821-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Heart failure (HF) is a clinical syndrome that involves structural changes in the heart, leading to a decrease in cardiac output, mainly caused by myocardial infarction (MI), which is the most common form of cardiovascular disease worldwide. Clinical evaluation remains the most accurate diagnostic method for ischemic HF, since the known biomarkers have high cost, are difficult to use for early diagnosis, and have low specificity. This often leads to late diagnosis since only ~ 25% symptoms of HF appear after MI. Studies suggest that small non-coding RNAs (miRNAs) play an important role in the regulation of this pathophysiological process and are, therefore, important targets in the discovery of non-invasive biomarkers for HF. Thus, the aim of this review was to identify circulating miRNAs (plasma, serum, and whole blood) described for post-MI HF patients. This review covered 19 experimental studies on humans, which investigated the relationship between circulating miRNAs and the development, monitoring, or prognosis of ischemic HF. This analysis was aimed at proposing potential targets for HF and the future application of miRNAs as HF biomarkers.
Collapse
Affiliation(s)
- Marina Sampaio Cruz
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Karla Simone Costa de Souza
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - André Ducati Luchessi
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical and Toxicology Analysis, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
19
|
Lee Y, Im E. Regulation of miRNAs by Natural Antioxidants in Cardiovascular Diseases: Focus on SIRT1 and eNOS. Antioxidants (Basel) 2021; 10:antiox10030377. [PMID: 33802566 PMCID: PMC8000568 DOI: 10.3390/antiox10030377] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of morbidity and mortality worldwide. The potential benefits of natural antioxidants derived from supplemental nutrients against CVDs are well known. Remarkably, natural antioxidants exert cardioprotective effects by reducing oxidative stress, increasing vasodilation, and normalizing endothelial dysfunction. Recently, considerable evidence has highlighted an important role played by the synergistic interaction between endothelial nitric oxide synthase (eNOS) and sirtuin 1 (SIRT1) in the maintenance of endothelial function. To provide a new perspective on the role of natural antioxidants against CVDs, we focused on microRNAs (miRNAs), which are important posttranscriptional modulators in human diseases. Several miRNAs are regulated via the consumption of natural antioxidants and are related to the regulation of oxidative stress by targeting eNOS and/or SIRT1. In this review, we have discussed the specific molecular regulation of eNOS/SIRT1-related endothelial dysfunction and its contribution to CVD pathologies; furthermore, we selected nine different miRNAs that target the expression of eNOS and SIRT1 in CVDs. Additionally, we have summarized the alteration of miRNA expression and regulation of activities of miRNA through natural antioxidant consumption.
Collapse
Affiliation(s)
| | - Eunok Im
- Correspondence: ; Tel.: +82-51-510-2812; Fax: +82-51-513-6754
| |
Collapse
|
20
|
Murugesan S, Saravanakumar L, Powell MF, Rajasekaran NS, Kannappan R, Berkowitz DE. Role of exosomal microRNA signatures: An emerging factor in preeclampsia-mediated cardiovascular disease. Placenta 2021; 103:226-231. [PMID: 33171429 PMCID: PMC8278543 DOI: 10.1016/j.placenta.2020.10.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022]
Abstract
Preeclampsia (PE) and vascular dysfunction are major causes of maternal and neonatal morbidity and mortality. Although extensively studied, the complete understanding of the pathophysiology behind PE remains unclear. Current reports indicate that exosomes are essential mediators in PE-related cardiovascular disease (CVDs). Exosomes are synthesized from multivesicular bodies (MVB) and contain functionally active microRNAs miRNAs). These miRNAs have been shown to mediate physiological and pathological functions through autocrine, paracrine, and endocrine signaling mechanisms. The role of miRNAs in pregnant women with PE has been studied extensively. However, little is known about the effect of exosomal miRNAs (exomiR) in PE. This paper will review and discuss the existing evidence for exomiR function in PE and highlight the need for future studies to explore the role that exomiR signatures have in cardiovascular dysfunction associated with PE.
Collapse
Affiliation(s)
- Saravanakumar Murugesan
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Lakshmi Saravanakumar
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Mark F Powell
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Namakkal Soorappan Rajasekaran
- Cardiac Aging and Redox Signaling Laboratory, Center for Free Radical Biology, Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL, USA
| | - Ramaswamy Kannappan
- Department of Biomedical Engineering, School of Engineering and Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Dan E Berkowitz
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
21
|
Mujalli A, Banaganapalli B, Alrayes NM, Shaik NA, Elango R, Al-Aama JY. Myocardial infarction biomarker discovery with integrated gene expression, pathways and biological networks analysis. Genomics 2020; 112:5072-5085. [PMID: 32920122 DOI: 10.1016/j.ygeno.2020.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/22/2020] [Accepted: 09/03/2020] [Indexed: 01/04/2023]
Abstract
Myocardial infarction (MI) is the most prevalent coronary heart disease caused by the complex molecular interactions between multiple genes and environment. Here, we aim to identify potential biomarkers for the disease development and for prognosis of MI. We have used gene expression dataset (GSE66360) generated from 51 healthy controls and 49 patients experiencing acute MI and analyzed the differentially expressed genes (DEGs), protein-protein interactions (PPI), gene network-clusters to annotate the candidate pathways relevant to MI pathogenesis. Bioinformatic analysis revealed 810 DEGs. Their functional annotations have captured several MI targeting biological processes and pathways like immune response, inflammation and platelets degranulation. PPI network identify seventeen hub and bottleneck genes, whose involvement in MI was further confirmed by DisGeNET database. OpenTarget Platform reveal unique bottleneck genes as potential target for MI. Our findings identify several potential biomarkers associated with early stage MI providing a new insight into molecular mechanism underlying the disease.
Collapse
Affiliation(s)
- Abdulrahman Mujalli
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Babajan Banaganapalli
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nuha Mohammad Alrayes
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor A Shaik
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jumana Y Al-Aama
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia; Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
22
|
Oxidative Stress in Cardiovascular Diseases. Antioxidants (Basel) 2020; 9:antiox9090864. [PMID: 32937950 PMCID: PMC7554855 DOI: 10.3390/antiox9090864] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are subcellular messengers in signal transductions pathways with both beneficial and deleterious roles. ROS are generated as a by-product of mitochondrial respiration or metabolism or by specific enzymes such as superoxide dismutases, glutathione peroxidase, catalase, peroxiredoxins, and myeloperoxidases. Under physiological conditions, the low levels of ROS production are equivalent to their detoxification, playing a major role in cellular signaling and function. In pathological situations, particularly atherosclerosis or hypertension, the release of ROS exceeds endogenous antioxidant capacity, leading to cell death. At cardiovascular levels, oxidative stress is highly implicated in myocardial infarction, ischemia/reperfusion, or heart failure. Here, we will first detail the physiological role of low ROS production in the heart and the vessels. Indeed, ROS are able to regulate multiple cardiovascular functions, such as cell proliferation, migration, and death. Second, we will investigate the implication of oxidative stress in cardiovascular diseases. Then, we will focus on ROS produced by NAPDH oxidase or during endothelial or mitochondrial dysfunction. Given the importance of oxidative stress at the cardiovascular level, antioxidant therapies could be a real benefit. In the last part of this review, we will detail the new therapeutic strategies potentially involved in cardiovascular protection and currently under study.
Collapse
|
23
|
Jusic A, Salgado-Somoza A, Paes AB, Stefanizzi FM, Martínez-Alarcón N, Pinet F, Martelli F, Devaux Y, Robinson EL, Novella S. Approaching Sex Differences in Cardiovascular Non-Coding RNA Research. Int J Mol Sci 2020; 21:E4890. [PMID: 32664454 PMCID: PMC7402336 DOI: 10.3390/ijms21144890] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the biggest cause of sickness and mortality worldwide in both males and females. Clinical statistics demonstrate clear sex differences in risk, prevalence, mortality rates, and response to treatment for different entities of CVD. The reason for this remains poorly understood. Non-coding RNAs (ncRNAs) are emerging as key mediators and biomarkers of CVD. Similarly, current knowledge on differential regulation, expression, and pathology-associated function of ncRNAs between sexes is minimal. Here, we provide a state-of-the-art overview of what is known on sex differences in ncRNA research in CVD as well as discussing the contributing biological factors to this sex dimorphism including genetic and epigenetic factors and sex hormone regulation of transcription. We then focus on the experimental models of CVD and their use in translational ncRNA research in the cardiovascular field. In particular, we want to highlight the importance of considering sex of the cellular and pre-clinical models in clinical studies in ncRNA research and to carefully consider the appropriate experimental models most applicable to human patient populations. Moreover, we aim to identify sex-specific targets for treatment and diagnosis for the biggest socioeconomic health problem globally.
Collapse
Affiliation(s)
- Amela Jusic
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Tuzla, 75000 Tuzla, Bosnia and Herzegovina;
| | - Antonio Salgado-Somoza
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (A.S.-S.); (F.M.S.); (Y.D.)
| | - Ana B. Paes
- INCLIVA Biomedical Research Institute, Menéndez Pelayo 4 Accesorio, 46010 Valencia, Spain; (A.B.P.); (N.M.-A.)
| | - Francesca Maria Stefanizzi
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (A.S.-S.); (F.M.S.); (Y.D.)
| | - Núria Martínez-Alarcón
- INCLIVA Biomedical Research Institute, Menéndez Pelayo 4 Accesorio, 46010 Valencia, Spain; (A.B.P.); (N.M.-A.)
| | - Florence Pinet
- INSERM, CHU Lille, Institut Pasteur de Lille, University of Lille, U1167 F-59000 Lille, France;
| | - Fabio Martelli
- Molecular Cardiology Laboratory, Policlinico San Donato IRCCS, San Donato Milanese, 20097 Milan, Italy;
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (A.S.-S.); (F.M.S.); (Y.D.)
| | - Emma Louise Robinson
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Susana Novella
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, and INCLIVA Biomedical Research Institute, Menéndez Pelayo 4 Accesorio, 46010 Valencia, Spain
| |
Collapse
|
24
|
Meiri E, Volinsky N, Dromi N, Kredo-Russo S, Benjamin H, Tabak S, Marmor H, Motin M, Lebanony D, Lithwick-Yanai G, Kadosh E, Kreader C, Grosman-Rimon L, Amir O. Differential expression of microRNA in serum fractions and association of Argonaute 1 microRNAs with heart failure. J Cell Mol Med 2020; 24:6586-6595. [PMID: 32400052 PMCID: PMC7299714 DOI: 10.1111/jcmm.15306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/01/2019] [Accepted: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
The serum or plasma microRNA (miRNA) molecules have been suggested as diagnostic and prognostic biomarkers, in various pathological conditions. However, these molecules are also found in different serum fractions, such as exosomes and Argonaute (Ago) protein complexes. Ago1 is the predominant Ago protein expressed in heart tissue. The objective of the study was to examine the hypothesis that Ago1‐associated miRNAs may be more relevant to cardiac disease and heart failure compared with the serum. In total, 84 miRNA molecules were screened for their expression in the whole serum, exosomes and Ago1, and Ago2 complexes. Ago1‐bound miR‐222‐3p, miR‐497‐5p and miR‐21‐5p were significantly higher, and let‐7a‐5p was significantly lower in HF patients compared with healthy controls, whereas no such difference was observed for those markers in the serum samples among the groups. A combination of these 4 miRNAs into an Ago1‐HF score provided a ROC curve with an AUC of 1, demonstrating clear discrimination between heart failure patients and healthy individuals. Ago1 fraction might be a better and more specific platform for identifying HF‐related miRNAs compared with the whole serum.
Collapse
Affiliation(s)
- Eti Meiri
- Rosetta Genomics Ltd, Rehovot, Israel
| | - Natalia Volinsky
- Division of Cardiovascular Medicine, Baruch Padeh Medical Center, Poriya, Israel.,The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Nir Dromi
- Rosetta Genomics Ltd, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | - Liza Grosman-Rimon
- Division of Cardiovascular Medicine, Baruch Padeh Medical Center, Poriya, Israel.,The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Offer Amir
- Division of Cardiovascular Medicine, Baruch Padeh Medical Center, Poriya, Israel.,The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| |
Collapse
|
25
|
Kalinina EV, Ivanova-Radkevich VI, Chernov NN. Role of MicroRNAs in the Regulation of Redox-Dependent Processes. BIOCHEMISTRY (MOSCOW) 2019; 84:1233-1246. [DOI: 10.1134/s0006297919110026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
26
|
Soler-Botija C, Gálvez-Montón C, Bayés-Genís A. Epigenetic Biomarkers in Cardiovascular Diseases. Front Genet 2019; 10:950. [PMID: 31649728 PMCID: PMC6795132 DOI: 10.3389/fgene.2019.00950] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are the number one cause of death worldwide and greatly impact quality of life and medical costs. Enormous effort has been made in research to obtain new tools for efficient and quick diagnosis and predicting the prognosis of these diseases. Discoveries of epigenetic mechanisms have related several pathologies, including cardiovascular diseases, to epigenetic dysregulation. This has implications on disease progression and is the basis for new preventive strategies. Advances in methodology and big data analysis have identified novel mechanisms and targets involved in numerous diseases, allowing more individualized epigenetic maps for personalized diagnosis and treatment. This paves the way for what is called pharmacoepigenetics, which predicts the drug response and develops a tailored therapy based on differences in the epigenetic basis of each patient. Similarly, epigenetic biomarkers have emerged as a promising instrument for the consistent diagnosis and prognosis of cardiovascular diseases. Their good accessibility and feasible methods of detection make them suitable for use in clinical practice. However, multicenter studies with a large sample population are required to determine with certainty which epigenetic biomarkers are reliable for clinical routine. Therefore, this review focuses on current discoveries regarding epigenetic biomarkers and its controversy aiming to improve the diagnosis, prognosis, and therapy in cardiovascular patients.
Collapse
Affiliation(s)
- Carolina Soler-Botija
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayés-Genís
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiology Service, HUGTiP, Badalona, Spain
- Department of Medicine, Barcelona Autonomous University (UAB), Badalona, Spain
| |
Collapse
|
27
|
Baćević M, Brković B, Lambert F, Djukić L, Petrović N, Roganović J. Leukocyte- and platelet-rich fibrin as graft material improves microRNA-21 expression and decreases oxidative stress in the calvarial defects of diabetic rabbits. Arch Oral Biol 2019; 102:231-237. [PMID: 31082699 DOI: 10.1016/j.archoralbio.2019.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Leukocyte- and platelet-rich fibrin (L-PRF) represents a natural, low-cost product which may promote tissue healing by mechanisms not fully elucidated. Diabetes mellitus (DM) disrupts bone healing by inducing inflammation and oxidative stress (OS), mechanisms regulated by microRNAs (miRs). The aim of the present study was to investigate the microRNA-21 (miR-21) involvement in diabetic bone regeneration using L-PRF alone or in combination with a standard grafting material. DESIGN After the induction of diabetes (alloxan 100 mg/kg), four cranial osteotomies were made in diabetic (n = 12) and non-diabetic (n = 12) rabbits: one was left empty and the remaining three were grafted with L-PRF, bovine hydroxyapatite (Bio-Oss®) and L-PRF + Bio-Oss®. Two and eight weeks postoperatively, the samples were harvested for miR-21 expression (Real-time RT-PCR) and enzyme-linked immunosorbent assay analyses. RESULTS Diabetic rabbits showed decreased miR-21 and matrix metalloproteinase-9 (MMP-9) protein expression while increased malondialdehyde (MDA) levels two weeks postoperatively; however, there were no significant differences in miR-21 and MMP-9 levels between diabetic and non-diabetic rabbits in samples taken eight weeks postoperatively. Application of L-PRF and L-PRF + Bio-Oss® improved miR-21 and MMP-9 and decreased MDA levels while Bio-Oss® alone enhanced superoxide dismutase (SOD) activity levels in diabetic rabbits. CONCLUSION L-PRF alone or in combination with bovine hydroxyapatite as bone graft could be beneficial in DM since it seems to improve inflammation-modulatory miR-21 expression and decreases oxidative stress.
Collapse
Affiliation(s)
- Miljana Baćević
- Dental Biomaterial Research Unit (d-BRU), Faculty of Medicine, University of Liege, Liege, Belgium
| | - Božidar Brković
- Department of Oral Surgery, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - France Lambert
- Department of Periodontology and Oral Surgery, Faculty of Medicine, University of Liege, Belgium
| | - Ljiljana Djukić
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Nina Petrović
- Department of Radiobiology and Molecular Genetics, Institute of Nuclear Sciences Vinča, University of Belgrade, Belgrade, Serbia; Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jelena Roganović
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
28
|
Essack M, Salhi A, Stanimirovic J, Tifratene F, Bin Raies A, Hungler A, Uludag M, Van Neste C, Trpkovic A, Bajic VP, Bajic VB, Isenovic ER. Literature-Based Enrichment Insights into Redox Control of Vascular Biology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1769437. [PMID: 31223421 PMCID: PMC6542245 DOI: 10.1155/2019/1769437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
In cellular physiology and signaling, reactive oxygen species (ROS) play one of the most critical roles. ROS overproduction leads to cellular oxidative stress. This may lead to an irrecoverable imbalance of redox (oxidation-reduction reaction) function that deregulates redox homeostasis, which itself could lead to several diseases including neurodegenerative disease, cardiovascular disease, and cancers. In this study, we focus on the redox effects related to vascular systems in mammals. To support research in this domain, we developed an online knowledge base, DES-RedoxVasc, which enables exploration of information contained in the biomedical scientific literature. The DES-RedoxVasc system analyzed 233399 documents consisting of PubMed abstracts and PubMed Central full-text articles related to different aspects of redox biology in vascular systems. It allows researchers to explore enriched concepts from 28 curated thematic dictionaries, as well as literature-derived potential associations of pairs of such enriched concepts, where associations themselves are statistically enriched. For example, the system allows exploration of associations of pathways, diseases, mutations, genes/proteins, miRNAs, long ncRNAs, toxins, drugs, biological processes, molecular functions, etc. that allow for insights about different aspects of redox effects and control of processes related to the vascular system. Moreover, we deliver case studies about some existing or possibly novel knowledge regarding redox of vascular biology demonstrating the usefulness of DES-RedoxVasc. DES-RedoxVasc is the first compiled knowledge base using text mining for the exploration of this topic.
Collapse
Affiliation(s)
- Magbubah Essack
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Adil Salhi
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Julijana Stanimirovic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| | - Faroug Tifratene
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Arwa Bin Raies
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Arnaud Hungler
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Mahmut Uludag
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Christophe Van Neste
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Andreja Trpkovic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| | - Vladan P. Bajic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| |
Collapse
|
29
|
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
|
30
|
Li X, Wei Y, Wang Z. microRNA-21 and hypertension. Hypertens Res 2018; 41:649-661. [PMID: 29973661 DOI: 10.1038/s41440-018-0071-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 01/11/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
Hypertension, a multifactorial disease, is a major risk factor for the development of stroke, coronary artery disease, heart failure, and chronic renal failure. However, its underlying cellular and molecular mechanisms remain largely elusive. Numerous studies have shown that microRNAs (miRNAs) are involved in a variety of cellular processes, including cellular proliferation, apoptosis, differentiation, and the development of diseases. microRNA-21 (miR-21), a conserved single-stranded non-coding RNA that is composed of approximately 22 nucleotides, is one of the most intensively studied miRNAs in recent years, and it can regulate gene expression at the post-transcriptional level. miR-21 is expressed in many kinds of tumors and in the cardiovascular system, and it plays an important role in the occurrence and development of cardiovascular diseases. In recent years, more and more evidence indicates that miR-21 plays an important role in hypertension. This article reviews the source, function, and altered levels of miR-21 in hypertension and the role of miR-21 in the pathogenesis of hypertension and target organ damage (TOD). The potential role of miR-21 as a new target for predicting and treating hypertension is also explored.
Collapse
Affiliation(s)
- Xiao Li
- Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, 100029, Beijing, China
| | - Yongxiang Wei
- Department of Otolaryngology Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029, Beijing, China.
| | - Zuoguang Wang
- Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, Blood Vessel Diseases, 100029, Beijing, China.
| |
Collapse
|
31
|
Radović N, Nikolić Jakoba N, Petrović N, Milosavljević A, Brković B, Roganović J. MicroRNA-146a and microRNA-155 as novel crevicular fluid biomarkers for periodontitis in non-diabetic and type 2 diabetic patients. J Clin Periodontol 2018; 45:663-671. [PMID: 29517812 DOI: 10.1111/jcpe.12888] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2018] [Indexed: 12/18/2022]
Abstract
AIM Recent studies point at the crucial role of epigenetic mechanisms in the development of multifactorial diseases such as periodontitis and diabetes mellitus (DM) type 2. In addition, circulatory microRNAs (miRs) have emerged as novel biomarkers for various diseases. Aim of this study was to investigate the levels of miR-146a and miR-155 and superoxide dismutase (SOD) activity in gingival crevicular fluid (GCF) of periodontitis patients with (CPDM) and without (CP) DM type 2 as well as in periodontally healthy, control groups (PHDM and PH, respectively). MATERIAL AND METHODS miR modulation was analysed using quantitative real-time PCR while SOD activity was measured spectrophotometrically. RESULTS The upregulation of miR-146a and miR-155 was observed in CP and CPDM patients' baseline, while the levels decreased after 6 weeks of the non-surgical therapy to the levels comparable to PH and PHDM, respectively. Expression levels of miRs positively correlated with SOD activity. Levels of miR-146a were higher in PHDM compared to PH patients. Multivariate analysis revealed that levels of miR-146a and miR-155 were significantly associated with periodontitis when adjusting for age and gender. CONCLUSIONS miR-146a and miR-155 may be considered as possible novel biomarkers for periodontitis in non-diabetic and type 2 diabetic patients.
Collapse
Affiliation(s)
- Nikola Radović
- Department of Periodontology, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Nataša Nikolić Jakoba
- Department of Periodontology, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Nina Petrović
- Department of Radiobiology and Molecular Genetics, Institute of Nuclear Sciences Vinca, University of Belgrade, Belgrade, Serbia
- Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Aleksandra Milosavljević
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Božidar Brković
- Department of Oral Surgery, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Roganović
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
32
|
Lan J, Huang Z, Han J, Shao J, Huang C. Redox regulation of microRNAs in cancer. Cancer Lett 2018; 418:250-259. [PMID: 29330105 DOI: 10.1016/j.canlet.2018.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/22/2017] [Accepted: 01/05/2018] [Indexed: 02/05/2023]
Abstract
Dysregulation of microRNAs (miRNAs) has long been implicated in tumorigenesis, whereas the underlying mechanisms remain largely unknown. Oxidative stress is a hallmark of cancer that involved in multiple pathophysiological processes, including the aberrant regulation of miRNAs. Compelling evidences have implied complicated interplay between reactive oxygen species (ROS) and miRNAs. Indeed, ROS induces carcinogenesis through either reducing or increasing the miRNA level, leading to the activation of oncogenes or silence of tumor suppressors, respectively. In turn, miRNAs target ROS productive genes or antioxidant responsive elements to affect cellular redox balance, which contributes to establishing a microenvironment favoring cancer cell growth and metastasis. Both miRNAs and ROS have been identified as potential biomarkers and therapeutic targets in human malignancies, and comprehensive understanding of the molecular events herein will facilitate the development of novel cancer therapeutic strategies.
Collapse
Affiliation(s)
- Jiang Lan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Jichun Shao
- Department of Urology, Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, Sichuan, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China.
| |
Collapse
|