1
|
Hajsadeghi S, Iranpour A, Mirshafiee S, Nekouian R, Mollababaei M, Motevalli H, Yasin Ahmadi SA, Dakkali MS. Impact of smoking on microRNAs in significant coronary artery disease. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2025; 63:49-59. [PMID: 39543851 DOI: 10.2478/rjim-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Given the importance of coronary artery disease (CAD) and the range of cardiovascular disease phenotypes in smokers, as well as the potential genetic and epigenetic factors, we were motivated to explore the impact of smoking on some selected microRNAs associated with significant CAD. METHODS A total of 60 individuals were selected in four groups including non-smoker without significant CAD (S-A-), non-smokers with significant CAD (S-A+), smokers without significant CAD (S+A-) and smokers with significant CAD (S+A+). Micro-RNA expression was investigated using real-time PCR. General linear model was used to calculate fold change (FC) considering S-A- as the reference group. RESULTS For mir-34a, down-regulation was observed in S+A- (FC =0.13, P =0.007) and S+A+ (FC =0.23, P =0.036) groups. For mir-126-3p, down-regulation was observed in S-A+ group (FC =0.05, P =0.024). For mir-199, up-regulation was observed for S+A- group (FC =9.38, P =0.007). The only significant interaction between pack-years of smoking and number of significantly narrowed vessels (≥75% stenosis) was for mir-199 which was in favor of down-regulation (P =0.006), while the main effects were in favor of up-regulation (P <0.05). CONCLUSION Mir-34a expression may be affected by smoking, whereas mir-126-3p expression may be affected by atherosclerosis, the most common reason of CAD. The significant down-regulation of mir-199 for the interaction of smoking dose and severity of CAD was a notable finding showing the harmful consequence of this interaction. Further studies are needed for this micro-RNA.
Collapse
Affiliation(s)
- Shokoufeh Hajsadeghi
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Aida Iranpour
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Mirshafiee
- Department of Cardiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Nekouian
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Mollababaei
- Pediatric Growth and Development Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Motevalli
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyyed Amir Yasin Ahmadi
- Preventive Medicine and Public Health Research Center, Psychosocial Health Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
2
|
Li Z, Xu R, Peng J, Wu T, Liu H, Wei Y. Cell-free DNA methylation signatures reflect the risk of vascular endothelial cell injury associated with traffic-related air pollution. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:177906. [PMID: 39671927 DOI: 10.1016/j.scitotenv.2024.177906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/19/2024] [Accepted: 12/01/2024] [Indexed: 12/15/2024]
Abstract
Traffic-related air pollution (TRAP) contains a plethora of hazardous pollutants, and is associated with a multitude of adverse health effects. However, up to now, no early biomarkers that can quickly reflect the health damage induced by TRAP. We conducted a randomized crossover trial in 35 healthy adults. Each subject was exposed to high- and low- levels of TRAP by walking in the park and along the side road leading to the freeway (road). The cardiopulmonary function parameters were measured before and after each walk session, and blood was collected 2 h after the two sessions. The present study revealed an intriguing phenomenon: the cardiac function was preferentially damaged by acute TRAP exposure. Then, we investigated the extent of damage to various human cells from exposure to TRAP by the technology of cell-of-origin of plasma cell-free DNA. Consequently, we found that only the cell-specific DNA methylation level of endothelial cells (EC) was elevated after exposure to TRAP (road) in comparison with the control (park), indicating that TRAP exposure primarily damages the EC. EC is an integral part of the cardiovascular system. This evidence indicated that TRAP exposure primarily damaged the cardiac function, compared with the other functions. Based on the cell-specific gene database of EC, we found higher levels of DNA methylation in the exon region after exposure to the TRAP session compared with the control session. Meanwhile, we also found that TRAP exposure could induce an elevated level of NACC2 in plasma, suggesting that plasma NACC2 could serve as a potential biomarker for damage of EC induced by TRAP exposure. This study suggests that the first target cell type damaged by TRAP is EC, and that the expression of NACC2 and its DNA methylation level in plasma may be a useful biomarker reflecting TRAP exposure-induced EC damage.
Collapse
Affiliation(s)
- Zhigang Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Rongrong Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Jianhao Peng
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Tingting Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Hao Liu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Yongjie Wei
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; College of Environmental Science and Engineering, Tongji University, Shanghai, China; Center for Global Health, School of Public Health, Nanjing Medical University, China.
| |
Collapse
|
3
|
Hon JX, Wahab NA, Karim AKA, Mokhtar NM, Mokhtar MH. Exploring the Role of MicroRNAs in Progesterone and Estrogen Receptor Expression in Endometriosis. Biomedicines 2024; 12:2218. [PMID: 39457531 PMCID: PMC11504708 DOI: 10.3390/biomedicines12102218] [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: 08/09/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Patients with endometriosis still respond poorly to progestins due to progesterone resistance associated with microRNAs (miRNAs). The aim of this study was to investigate the expression of selected miRNAs, estrogen receptor (ER)α, ERβ, progesterone receptor (PR)-A and PR-B and to determine the target genes of upregulated miRNAs in endometriosis. Methods: In this study, 18 controls, 18 eutopic and 18 ectopic samples were analysed. Profiling and validation of miRNAs associated with functions of endometriosis were performed using next-generation sequencing (NGS) and qRT-PCR. At the same time, the expression of ERα, ERβ, PR-A and PR-B was also determined using qRT-PCR. Target prediction was also performed for miR-199a-3p, miR-1-3p and miR-125b-5p using StarBase. Results: In this study, NGS identified seven significantly differentially expressed miRNAs, of which six miRNAs related to the role of endometriosis were selected for validation by qRT-PCR. The expression of miR-199a-3p, miR-1-3p, miR-146a-5p and miR-125b-5p was upregulated in the ectopic group compared to the eutopic group. Meanwhile, ERα and ERβ were significantly differentially expressed in endometriosis compared to the control group. However, the expressions of PR-A and PR-B showed no significant differences between the groups. The predicted target genes for miR-199a-3p, miR-1-3p and miR-125b-5p are SCD, TAOK1, DDIT4, LASP1, CDK6, TAGLN2, G6PD and ELOVL6. Conclusions: Our findings demonstrated that the expressions of ERα and ERβ might be regulated by miRNAs contributing to progesterone resistance, whereas the binding of miRNAs to target genes could also contribute to the pathogenesis of endometriosis. Therefore, miRNAs could be used as potential biomarkers and for targeted therapy in patients with endometriosis.
Collapse
Affiliation(s)
- Jing-Xian Hon
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (J.-X.H.)
| | - Norhazlina Abdul Wahab
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (J.-X.H.)
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Abdul Kadir Abdul Karim
- Department of Obstetrics & Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (J.-X.H.)
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (J.-X.H.)
| |
Collapse
|
4
|
Wang Y, Shi X. The potential mechanisms and treatment effects of stem cell-derived exosomes in cardiac reengineering. NANOTECHNOLOGY 2024; 35:362005. [PMID: 38834043 DOI: 10.1088/1361-6528/ad53d1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Exosomes are extracellular vesicles of diverse compositions that are secreted by numerous cell types. Exosomes contain significant bioactive components, including lipids, proteins, mRNA, and miRNA. Exosomes play an important role in regulating cellular signaling and trafficking under both normal physiological and pathological circumstances. A multitude of factors, including thermal stress, ribosomal stress, endoplasmic reticulum stress, and oxidative stress influence the concentrations of exosomal mRNA, miRNA, proteins, and lipids. It has been stated that exosomes derived from stem cells (SCs) modulate a range of stresses by preventing or fostering cell balance. Exosomes derived from SCs facilitate recovery by facilitating cross-cellular communication via the transmission of information in the form of proteins, lipids, and other components. For this reason, exosomes are used as biomarkers to diagnose a wide variety of diseases. The focus of this review is the bioengineering of artificial exosomal cargoes. This process encompasses the control and transportation of particular exosomal cargoes, including but not limited to small molecules, recombinant proteins, immune modulators, and therapeutic medications. Therapeutic approaches of this nature have the potential to deliver therapeutic medications precisely to the intended site for the cure of a variety of disorders. Notably, our attention has been directed towards the therapeutic implementations of exosomes derived from SCs in the cure of cardiovascular ailments, including but not limited to ischemic heart disease, myocardial infarction, sepsis, heart failure, cardiomyopathy, and cardiac fibrosis. In general, researchers employ two methodologies when it comes to exosomal bioengineering. This review aims to explain the function of exosomes derived from SCs in the regulation of stress and present a novel therapeutic approach for cardiovascular disorders.
Collapse
Affiliation(s)
- Yibin Wang
- Department of Cardiology, Hangzhou Ninth People's Hospital, Hangzhou 311225, People's Republic of China
| | - Xiulian Shi
- Emergency Department, Chun'an First People's Hospital, Hangzhou 311700, People's Republic of China
| |
Collapse
|
5
|
Kaur G, Pippin JA, Chang S, Redmond J, Chesi A, Wells AD, Maerz T, Grant SFA, Coleman RM, Hankenson KD, Wagley Y. Osteoporosis GWAS-implicated DNM3 locus contextually regulates osteoblastic and chondrogenic fate of mesenchymal stem/progenitor cells through oscillating miR-199a-5p levels. JBMR Plus 2024; 8:ziae051. [PMID: 38686038 PMCID: PMC11056323 DOI: 10.1093/jbmrpl/ziae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Genome wide association study (GWAS)-implicated bone mineral density (BMD) signals have been shown to localize in cis-regulatory regions of distal effector genes using 3D genomic methods. Detailed characterization of such genes can reveal novel causal genes for BMD determination. Here, we elected to characterize the "DNM3" locus on chr1q24, where the long non-coding RNA DNM3OS and the embedded microRNA MIR199A2 (miR-199a-5p) are implicated as effector genes contacted by the region harboring variation in linkage disequilibrium with BMD-associated sentinel single nucleotide polymorphism, rs12041600. During osteoblast differentiation of human mesenchymal stem/progenitor cells (hMSC), miR-199a-5p expression was temporally decreased and correlated with the induction of osteoblastic transcription factors RUNX2 and Osterix. Functional relevance of miR-199a-5p downregulation in osteoblastogenesis was investigated by introducing miR-199a-5p mimic into hMSC. Cells overexpressing miR-199a-5p depicted a cobblestone-like morphological change and failed to produce BMP2-dependent extracellular matrix mineralization. Mechanistically, a miR-199a-5p mimic modified hMSC propagated normal SMAD1/5/9 signaling and expressed osteoblastic transcription factors RUNX2 and Osterix but depicted pronounced upregulation of SOX9 and enhanced expression of essential chondrogenic genes ACAN, COMP, and COL10A1. Mineralization defects, morphological changes, and enhanced chondrogenic gene expression associated with miR-199a-5p mimic over-expression were restored with miR-199a-5p inhibitor suggesting specificity of miR-199a-5p in chondrogenic fate specification. The expression of both the DNM3OS and miR-199a-5p temporally increased and correlated with hMSC chondrogenic differentiation. Although miR-199a-5p overexpression failed to further enhance chondrogenesis, blocking miR-199a-5p activity significantly reduced chondrogenic pellet size, extracellular matrix deposition, and chondrogenic gene expression. Taken together, our results indicate that oscillating miR-199a-5p levels dictate hMSC osteoblast or chondrocyte terminal fate. Our study highlights a functional role of miR-199a-5p as a BMD effector gene at the DNM3 BMD GWAS locus, where patients with cis-regulatory genetic variation which increases miR-199a-5p expression could lead to reduced osteoblast activity.
Collapse
Affiliation(s)
- Gurcharan Kaur
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - James A Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Solomon Chang
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Justin Redmond
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Division of Diabetes and Endocrinology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Kurt D Hankenson
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| |
Collapse
|
6
|
Xu Q, Xiao Z, Yang Q, Yu T, Deng X, Chen N, Huang Y, Wang L, Guo J, Wang J. Hydrogel-based cardiac repair and regeneration function in the treatment of myocardial infarction. Mater Today Bio 2024; 25:100978. [PMID: 38434571 PMCID: PMC10907859 DOI: 10.1016/j.mtbio.2024.100978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
A life-threatening illness that poses a serious threat to human health is myocardial infarction. It may result in a significant number of myocardial cells dying, dilated left ventricles, dysfunctional heart function, and ultimately cardiac failure. Based on the development of emerging biomaterials and the lack of clinical treatment methods and cardiac donors for myocardial infarction, hydrogels with good compatibility have been gradually applied to the treatment of myocardial infarction. Specifically, based on the three processes of pathophysiology of myocardial infarction, we summarized various types of hydrogels designed for myocardial tissue engineering in recent years, including natural hydrogels, intelligent hydrogels, growth factors, stem cells, and microRNA-loaded hydrogels. In addition, we also describe the heart patch and preparation techniques that promote the repair of MI heart function. Although most of these hydrogels are still in the preclinical research stage and lack of clinical trials, they have great potential for further application in the future. It is expected that this review will improve our knowledge of and offer fresh approaches to treating myocardial infarction.
Collapse
Affiliation(s)
- Qiaxin Xu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, 510630, China
| | - Qianzhi Yang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Tingting Yu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Xiujiao Deng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Nenghua Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Lihong Wang
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Endocrinology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jun Guo
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
7
|
Zalivina I, Barwari T, Yin X, Langley SR, Barallobre-Barreiro J, Wakimoto H, Zampetaki A, Mayr M, Avkiran M, Eminaga S. Inhibition of miR-199a-3p in a murine hypertrophic cardiomyopathy (HCM) model attenuates fibrotic remodeling. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 6:100056. [PMID: 38143961 PMCID: PMC10739604 DOI: 10.1016/j.jmccpl.2023.100056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
Background Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disorder, characterized by cardiomyocyte hypertrophy, cardiomyocyte disarray and fibrosis, which has a prevalence of ∼1: 200-500 and predisposes individuals to heart failure and sudden death. The mechanisms through which diverse HCM-causing mutations cause cardiac dysfunction remain mostly unknown and their identification may reveal new therapeutic avenues. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and disease phenotype in various pathologies. We explored whether miRNAs could play a role in HCM pathogenesis and offer potential therapeutic targets. Methods and results Using high-throughput miRNA expression profiling and qPCR analysis in two distinct mouse models of HCM, we found that miR-199a-3p expression levels are upregulated in mutant mice compared to age- and treatment-matched wild-type mice. We also found that miR-199a-3p expression is enriched in cardiac non-myocytes compared to cardiomyocytes. When we expressed miR-199a-3p mimic in cultured murine primary cardiac fibroblasts and analyzed the conditioned media by proteomics, we found that several extracellular matrix (ECM) proteins (e.g., TSP2, FBLN3, COL11A1, LYOX) were differentially secreted (data are available via ProteomeXchange with identifier PXD042904). We confirmed our proteomics findings by qPCR analysis of selected mRNAs and demonstrated that miR-199a-3p mimic expression in cardiac fibroblasts drives upregulation of ECM gene expression, including Tsp2, Fbln3, Pcoc1, Col1a1 and Col3a1. To examine the role of miR-199a-3p in vivo, we inhibited its function using lock-nucleic acid (LNA)-based inhibitors (antimiR-199a-3p) in an HCM mouse model. Our results revealed that progression of cardiac fibrosis is attenuated when miR-199a-3p function is inhibited in mild-to-moderate HCM. Finally, guided by computational target prediction algorithms, we identified mRNAs Cd151 and Itga3 as direct targets of miR-199a-3p and have shown that miR-199a-3p mimic expression negatively regulates AKT activation in cardiac fibroblasts. Conclusions Altogether, our results suggest that miR-199a-3p may contribute to cardiac fibrosis in HCM through its actions in cardiac fibroblasts. Thus, inhibition of miR-199a-3p in mild-to-moderate HCM may offer therapeutic benefit in combination with complementary approaches that target the primary defect in cardiac myocytes.
Collapse
Affiliation(s)
- Irina Zalivina
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Temo Barwari
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Xiaoke Yin
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Sarah R. Langley
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Anna Zampetaki
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Manuel Mayr
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Metin Avkiran
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Seda Eminaga
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| |
Collapse
|
8
|
Wugeng S, Zan S, Liu Y, Bai Y, Hu A, Wei X, Guo C, Su X. MicroRNA-214-3p protects against myocardial ischemia-reperfusion injury by targeting demethylase lysine demethylase 3A. Regen Ther 2023; 23:17-24. [PMID: 37013195 PMCID: PMC10066509 DOI: 10.1016/j.reth.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/12/2023] [Accepted: 01/26/2023] [Indexed: 04/05/2023] Open
Abstract
Objective Many studies have explored the roles of microRNAs (miRs) in myocardial ischemia/reperfusion injury (MI/RI), while the function of miR-214-3p in MI/RI remained obscure. This study aims to unravel the regulatory mechanism of miR-214-3p in MI/RI via targeting histone demethylase lysine demethylase 3A (KDM3A). Methods MI/RI rat model was established by ligating the left anterior descending coronary artery. MiR-214-3p and KDM3A expression in myocardial tissues of MI/RI rats was examined. Then, the serum oxidative stress factors, inflammatory factors, pathological changes of myocardial tissues, cardiomyocyte apoptosis, and fibrosis of myocardial tissues were detected in MI/RI rats intervening with miR-214-3p or KDM3A expression. The targeting relation between miR-214-3p and KDM3A was validated. Results MiR-214-3p was low-expressed while KDM3A was high-expressed in MI/RI rat model. Up-regulated miR-214-3p or down-regulated KDM3A protected against MI/RI via mitigating serum oxidative response, reducing the levels of inflammatory factors, alleviating the pathological changes of myocardial tissues, and decreasing cardiomyocyte apoptosis and fibrosis of myocardial tissue. KDM3A amplification reversed the therapeutic effects of elevated miR-214-3p on MI/RI. KDM3A was targeted by miR-214-3p. Conclusion miR-214-3p hinders cardiomyocyte apoptosis and myocardial injury in MI/RI rats via regulating KDM3A. Thus, miR-214-3p may function as a potential candidate for MI/RI treatment.
Collapse
|
9
|
Miceli V, Bulati M, Gallo A, Iannolo G, Busà R, Conaldi PG, Zito G. Role of Mesenchymal Stem/Stromal Cells in Modulating Ischemia/Reperfusion Injury: Current State of the Art and Future Perspectives. Biomedicines 2023; 11:689. [PMID: 36979668 PMCID: PMC10045387 DOI: 10.3390/biomedicines11030689] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) is a multistep damage that occurs in several tissues when a blood flow interruption is inevitable, such as during organ surgery or transplantation. It is responsible for cell death and tissue dysfunction, thus leading, in the case of transplantation, to organ rejection. IRI takes place during reperfusion, i.e., when blood flow is restored, by activating inflammation and reactive oxygen species (ROS) production, causing mitochondrial damage and apoptosis of parenchymal cells. Unfortunately, none of the therapies currently in use are definitive, prompting the need for new therapeutic approaches. Scientific evidence has proven that mesenchymal stem/stromal cells (MSCs) can reduce inflammation and ROS, prompting this cellular therapy to also be investigated for treatment of IRI. Moreover, it has been shown that MSC therapeutic effects were mediated in part by their secretome, which appears to be involved in immune regulation and tissue repair. For these reasons, mediated MSC paracrine function might be key for injury amelioration upon IRI damage. In this review, we highlight the scientific literature on the potential beneficial use of MSCs and their products for improving IRI outcomes in different tissues/organs, focusing in particular on the paracrine effects mediated by MSCs, and on the molecular mechanisms behind these effects.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCSS ISMETT (Istituto Mediterraneo per I Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | | | | | | | | | | | - Giovanni Zito
- Research Department, IRCSS ISMETT (Istituto Mediterraneo per I Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| |
Collapse
|
10
|
Yarmohammadi F, Ebrahimian Z, Karimi G. MicroRNAs target the PI3K/Akt/p53 and the Sirt1/Nrf2 signaling pathways in doxorubicin-induced cardiotoxicity. J Biochem Mol Toxicol 2023; 37:e23261. [PMID: 36416353 DOI: 10.1002/jbt.23261] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/06/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
Doxorubicin (DOX) is used as a chemotherapeutic agent in the treatment of solid tumors. Irreversible cardiotoxicity is the major limitation in the clinical use of DOX. Several microRNAs (miRNAs) with diversified functions are identified that participate in exacerbating or suppressing DOX-induced cardiac damage. The miRNAs are small noncoding regulatory RNAs that modify the expression of the native genes. Studies have demonstrated that miRNAs by modifying the expression of proteins such as PTEN, Akt, and survivin can affect DOX-induced cardiac apoptosis. Moreover, miRNAs can modulate cardiac oxidative stress in DOX treatment through the posttranscriptional regulation of Sirt1, p66shc, and Nrf2 expressions. This manuscript has reviewed the regulation of the PI3K/Akt/p53 and the Sirt1/Nrf2 pathways by miRNAs in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zainab Ebrahimian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Bai LQ, Wang BZ, Liu QW, Li WQ, Zhou H, Yang XY. Effects of penehyclidine hydrochloride on myocardial ischaemia-reperfusion injury in rats by inhibiting TLR4/MyD88/NF-κB pathway via miR-199a-3p. Growth Factors 2022; 40:186-199. [PMID: 35984706 DOI: 10.1080/08977194.2022.2109469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
This study was to probe the role of penehyclidine hydrochloride (PHC) mediating the impact of toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor-kappa B (NF-κB) signalling pathway on myocardial ischaemia-reperfusion injury (MI/RI) in rats through miR-199a-3p. The rat MI/RI model was established through ligating left anterior descending (LAD) coronary artery. PHC was injected preoperatively into the model rats, and injected with miR-199a-3p lentiviral vector or TLR4 antagonist (TAK-242). Next, cardiac function of rats was examined by echocardiography, and rat serum indicators, oxidative stress levels and inflammatory factors were detected. HE staining was applied to detect pathological tissue structure, TUNEL staining to detect apoptosis rate, qRCR and western blot to detect miR-199a-3p and TLR4/MyD88/NF-κB expressions in rat myocardial tissues. Dual luciferase reporter experiment was conducted to confirm the relationship between miR-199a-3p and TLR4. In conclusion, PHC suppresses TLR4/MyD88/NF-κB signalling pathway through miR-199a-3p, thereby improving MI/RI in rats.
Collapse
Affiliation(s)
- Ling Qiang Bai
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Bin Zhe Wang
- Department of The First Outpatients, The 940th Hospital of Joint Logistics Support force of Chinese People's Liberation Army, Lanzhou City, China
| | - Qi Wei Liu
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Wen Qiang Li
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Hang Zhou
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| | - Xiao Yan Yang
- Department of Cardiovascular Medicine, Baoji High-Tech Hospital, Baoji City, China
| |
Collapse
|
12
|
Improvement of Myocardial Cell Injury by miR-199a-3p/mTOR Axis through Regulating Cell Apoptosis and Autophagy. J Immunol Res 2022; 2022:1642301. [PMID: 35785031 PMCID: PMC9242751 DOI: 10.1155/2022/1642301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background Myocardial ischemia-reperfusion injury (MIRI) is characterized by its high incidence rate and mortality. miR-199a-3p is thought to be strongly linked with the development of some myocardial diseases, but the influence of miR-199a-3p in MIRI remains unclear. Methods AC16 cells were used. The concentrations of mammalian target of rapamycin (mTOR), light chain 3 II/light chain 3 I, and Beclin-1 were detected with western blotting and qRT-PCR. The binding site between mTOR and miR-199a-3p was evaluated via luciferase report assay. Cell apoptosis was evaluated through flow cytometry. Results Knockdown of miR-199a-3p accelerated the myocardial cell injury after L-oxygen treatment. Increased expression of mTOR and suppressed autophagy were observed after knockdown of miR-199a-3p. Knockdown of miR-199a-3p or overexpression of mTOR greatly aggravated cell injury through inhibiting autophagy. Conclusions. This study might be helpful for the therapeutic method of MIRI through by regulating miR-199a-3p/mTOR.
Collapse
|
13
|
Platelet-Membrane-Encapsulated Carvedilol with Improved Targeting Ability for Relieving Myocardial Ischemia-Reperfusion Injury. MEMBRANES 2022; 12:membranes12060605. [PMID: 35736311 PMCID: PMC9227294 DOI: 10.3390/membranes12060605] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/26/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023]
Abstract
In recent years, cell membrane drug delivery systems have received increasing attention. However, drug-loaded membrane delivery systems targeting therapy in myocardial ischemia–reperfusion injury (MIRI) have been relatively rarely studied. The purpose of this study was to explore the protective effect of platelet-membrane-encapsulated Carvedilol on MIRI. We extracted platelets from the blood of adult SD rats and prepared platelet membrane vesicles (PMVs). Carvedilol, a nonselective β-blocker, was encapsulated into the PMVs. In order to determine the best encapsulation rate and drug-loading rate, three different concentrations of Carvedilol in low, medium, and high amounts were fused to the PMVs in different volume ratios (drugs/PMVs at 2:1, 1:1, 1:2, and 4:1) for determining the optimum concentration and volume ratio. By comparing other delivery methods, including abdominal injection and intravenous administration, the efficacy of PMVs-encapsulated drug-targeted delivery treatment was observed. The PMVs have the ability to target ischemic-damaged myocardial tissue, and the concentration and volume ratio at the optimum encapsulation rate and the drug-loading rate are 0.5 mg and 1:1. We verified that PMVs@Carvedilol had better therapeutic effects compared to other treatment groups, and immunofluorescence observation showed a significant improvement in the apoptosis indicators and infarction area of myocardial cells. Targeted administration of PMVs@Carvedilol may be a promising treatment for myocardial reperfusion injury, as it significantly improves postinjury cardiac function and increases drug utilization compared to other delivery methods.
Collapse
|
14
|
Gu X, Weng R, Hou J, Liu S. Endothelial miR-199a-3p regulating cell adhesion molecules by targeting mTOR signaling during inflammation. Eur J Pharmacol 2022; 925:174984. [PMID: 35489420 DOI: 10.1016/j.ejphar.2022.174984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Adherence of monocytes to endothelial cells is the initial stage for development of coronary artery disease (CAD). MiRNAs have been reported to participate in this process by regulating the expression of cell adhesion molecules. This study aimed to explore the function of miR-199a-3p in endothelial inflammation and adhesion. METHODS We assessed the expression of miR-199a-3p in CAD patients and ApoE-/- mice. The relationship between miR-199a-3p level and endothelial inflammation and adhesion was examined. ELISA was used to test the level of IL-6 and IL-8. Dual luciferase reporter assay was used to evaluate the binding between miR-199a-3p and mTOR. RESULTS A decreased expression of miR-199a-3p was observed in the PBMCs and plasma of CAD patients, aorta of ApoE-/- mice and inflammatory HUVECs. MiR-199a-3p significantly suppressed the expression levels of pro-inflammatory cytokine (IL-6, IL-8), endothelial adhesion molecules (ICAM-1, VCAM-1) and monocyte-endothelial cells interaction. MiR-199a-3p directly targeted and repressed mTOR, and its suppression effect on ICAM-1 and VCAM-1 was abolished by mTOR inhibitor rapamycin, and rescued by mTOR activator MHY1485. Overexpression of miR-199a-3p promoted autophagy in HUVECs and inhibiting autophagy by chloroquine attenuated the effect of miR-199a-3p on ICAM-1 and VCAM-1 expression. Inhibition of autophagy promoted endothelial adhesion molecule expression and monocyte-EC interaction. CONCLUSIONS Our results suggested that miR-199a-3p suppressed endothelial inflammation and adhesion by targeting mTOR signaling and increasing autophagy. Our findings point to an important role for miR-199a-3p in the early stage of cardiovascular disease.
Collapse
Affiliation(s)
- Xiaodong Gu
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, PR China; Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, PR China
| | - Ruiqiang Weng
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, PR China; Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, PR China
| | - Jingyuan Hou
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, PR China; Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, PR China.
| | - Sudong Liu
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, PR China; Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, PR China.
| |
Collapse
|
15
|
Protective Effect of Cardiomyocyte-Specific Prolyl-4-Hydroxylase 2 Inhibition on Ischemic Injury in a Mouse MI Model. J Am Coll Surg 2022; 235:240-254. [PMID: 35758926 DOI: 10.1097/xcs.0000000000000241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Our earlier studies showed that inhibiting prolyl-4-hydroxylase enzymes (PHD-1 and PHD-3) improves angiogenesis, heart function, and limb perfusion in mouse models via stabilizing hypoxia-inducible transcription factor-alpha (HIF-1α). The present study explored the effects of the prolyl-4-hydroxylase enzyme, PHD-2, on ischemic heart failure using cardiac-specific PHD-2 gene knockout (KO) mice (PHD2-/-). STUDY DESIGN Adult wild-type (WT) and PHD2-/- mice, 8-12 weeks old, were subjected to myocardial infarction (MI) by irreversibly ligating the left anterior descending (LAD) coronary artery. All sham group mice underwent surgery without LAD ligation. Animals were divided into four groups 1) Wild-type Sham (WTS); Wild-Type myocardial infarction (WTMI); 3) PHD2KO Sham (PHD2-/-S); 4) PHD2KO myocardial infarction (PHD2-/-MI). Left ventricular tissue samples collected at various time points following surgery were used for microRNA expression profiling, Western blotting, immunohistochemical, and echocardiographic analysis. RESULTS Volcano plot analysis revealed 19 differentially expressed miRNAs in the PHD2-/-MI compared to the WTMI group. Target analysis using Ingenuity Pathway Analysis showed several differentially regulated miRNAs targeting key signaling pathways such as Akt, VEGF, Ang-1, PTEN, apoptosis, and hypoxia pathways. Compared to the WTMI group, Western blot analysis showed increased HIF-1α, VEGF, phospho-AKT, and β-catenin expression and reduced Bax expression for the PHD2-/-MI group post-MI. Echocardiographic analysis showed preserved heart functions, and picrosirius red staining revealed decreased fibrosis in PHD2-/-MI compared to the WTMI group. CONCLUSION PHD2 inhibition showed preserved heart function, enhanced angiogenic factor expression, and decreased apoptotic markers after MI. Overall, PHD2 gene inhibition is a promising candidate for managing cardiovascular diseases.
Collapse
|
16
|
Mesenchymal stem cell-derived exosomal microRNA-182-5p alleviates myocardial ischemia/reperfusion injury by targeting GSDMD in mice. Cell Death Dis 2022; 8:202. [PMID: 35422485 PMCID: PMC9010441 DOI: 10.1038/s41420-022-00909-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 01/03/2023]
Abstract
Recent evidence indicates that exosomes derived from mesenchymal stem cells (MSCs) confer protective effects against myocardial ischemia/reperfusion (I/R) injury. Exosomes are carriers of potentially protective endogenous molecules, including microRNAs (miRNAs/miRs). The current study set out to test the effects of transferring miR-182-5p from MSC-derived exosomes into myocardial cells on myocardial I/R injury. First, an I/R mouse model was developed by left anterior descending coronary artery occlusion, and myocardial cells were exposed to hypoxia/reoxygenation (H/R) for in vitro I/R model establishment. Loss- and gain-of-function experiments of miR-182-5p and GSDMD were conducted to explore the effects of miR-182-5p via MSC-derived exosomes on cell pyroptosis and viability. GSDMD was robustly expressed in I/R-injured myocardial tissues and H/R-exposed myocardial cells. GSDMD upregulation promoted H/R-induced myocardial cell pyroptosis and reduced viability, corresponding to increased lactate dehydrogenase release, reactive oxygen species production, and pyroptosis. A luciferase assay demonstrated GSDMD as a target of miR-182-5p. In addition, exosomal miR-182-5p was found to diminish GSDMD-dependent cell pyroptosis and inflammation induced by H/R. Furthermore, MSC-derived exosomes carrying miR-182-5p improved cardiac function and reduced myocardial infarction, accompanied with reduced inflammation and cell pyroptosis in vivo. Taken together, our findings suggest a cardioprotective effect of exosomal miR-182-5p against myocardial I/R injury, shedding light on an attractive therapeutic strategy.
Collapse
|
17
|
Wang M, Yan L, Li Q, Yang Y, Turrentine M, March K, Wang IW. Mesenchymal stem cell secretions improve donor heart function following ex vivo cold storage. J Thorac Cardiovasc Surg 2022; 163:e277-e292. [PMID: 32981709 PMCID: PMC7921217 DOI: 10.1016/j.jtcvs.2020.08.095] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Heart transplantation is the gold standard of treatments for end-stage heart failure, but its use is limited by extreme shortage of donor organs. The time "window" between procurement and transplantation sets the stage for myocardial ischemia/reperfusion injury, which constrains the maximal storage time and lowers use of donor organs. Given mesenchymal stem cell (MSC)-derived paracrine protection, we aimed to evaluate the efficacy of MSC-conditioned medium (CM) and extracellular vesicles (EVs) when added to ex vivo preservation solution on ameliorating ischemia/reperfusion-induced myocardial damage in donor hearts. METHODS Mouse donor hearts were stored at 0°C-4°C of <1-hour cold ischemia (<1hr-I), 6hr-I + vehicle, 6hr-I + MSC-CM, 6hr-I + MSC-EVs, and 6hr-I + MSC-CM from MSCs treated with exosome release inhibitor. The hearts were then heterotopically implanted into recipient mice. At 24 hours postsurgery, myocardial function was evaluated. Heart tissue was collected for analysis of histology, apoptotic cell death, microRNA (miR)-199a-3p expression, and myocardial cytokine production. RESULTS Six-hour cold ischemia significantly impaired myocardial function, increased cell death, and reduced miR-199a-3p in implanted hearts versus <1hr-I. MSC-CM or MSC-EVs in preservation solution reversed the detrimental effects of prolong cold ischemia on donor hearts. Exosome-depleted MSC-CM partially abolished MSC secretome-mediated cardioprotection in implanted hearts. MiR-199a-3p was highly enriched in MSC-EVs. MSC-CM and MSC-EVs increased cold ischemia-downregulated miR-199a-3p in donor hearts, whereas exosome-depletion neutralized this effect. CONCLUSIONS MSC-CM and MSC-EVs confer improved myocardial preservation in donor hearts during prolonged cold static storage and MSC-EVs can be used for intercellular transport of miRNAs in heart transplantation.
Collapse
Affiliation(s)
- Meijing Wang
- Division of Cardiothoracic Surgery, Department of Surgery, IU School of Medicine, Indianapolis, Ind.
| | - Liangliang Yan
- Division of Cardiothoracic Surgery, Department of Surgery, IU School of Medicine, Indianapolis, Ind; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fujian, China
| | - Qianzhen Li
- Division of Cardiothoracic Surgery, Department of Surgery, IU School of Medicine, Indianapolis, Ind; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fujian, China; Division of Cardiovascular Medicine, Department of Medicine, IU School of Medicine, Indianapolis, Ind
| | - Yang Yang
- Division of Cardiothoracic Surgery, Department of Surgery, IU School of Medicine, Indianapolis, Ind
| | - Mark Turrentine
- Division of Cardiothoracic Surgery, Department of Surgery, IU School of Medicine, Indianapolis, Ind
| | - Keith March
- Division of Cardiovascular Medicine, Department of Medicine, IU School of Medicine, Indianapolis, Ind; Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, Fla
| | - I-Wen Wang
- Division of Cardiothoracic Surgery, Department of Surgery, IU School of Medicine, Indianapolis, Ind; Methodist Hospital, IU Health, IU School of Medicine, Indianapolis, Ind.
| |
Collapse
|
18
|
Gustafson D, Ngai M, Wu R, Hou H, Schoffel AC, Erice C, Mandla S, Billia F, Wilson MD, Radisic M, Fan E, Trahtemberg U, Baker A, McIntosh C, Fan CPS, Dos Santos CC, Kain KC, Hanneman K, Thavendiranathan P, Fish JE, Howe KL. Cardiovascular signatures of COVID-19 predict mortality and identify barrier stabilizing therapies. EBioMedicine 2022; 78:103982. [PMID: 35405523 PMCID: PMC8989492 DOI: 10.1016/j.ebiom.2022.103982] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial cell (EC) activation, endotheliitis, vascular permeability, and thrombosis have been observed in patients with severe coronavirus disease 2019 (COVID-19), indicating that the vasculature is affected during the acute stages of SARS-CoV-2 infection. It remains unknown whether circulating vascular markers are sufficient to predict clinical outcomes, are unique to COVID-19, and if vascular permeability can be therapeutically targeted. Methods Prospectively evaluating the prevalence of circulating inflammatory, cardiac, and EC activation markers as well as developing a microRNA atlas in 241 unvaccinated patients with suspected SARS-CoV-2 infection allowed for prognostic value assessment using a Random Forest model machine learning approach. Subsequent ex vivo experiments assessed EC permeability responses to patient plasma and were used to uncover modulated gene regulatory networks from which rational therapeutic design was inferred. Findings Multiple inflammatory and EC activation biomarkers were associated with mortality in COVID-19 patients and in severity-matched SARS-CoV-2-negative patients, while dysregulation of specific microRNAs at presentation was specific for poor COVID-19-related outcomes and revealed disease-relevant pathways. Integrating the datasets using a machine learning approach further enhanced clinical risk prediction for in-hospital mortality. Exposure of ECs to COVID-19 patient plasma resulted in severity-specific gene expression responses and EC barrier dysfunction, which was ameliorated using angiopoietin-1 mimetic or recombinant Slit2-N. Interpretation Integration of multi-omics data identified microRNA and vascular biomarkers prognostic of in-hospital mortality in COVID-19 patients and revealed that vascular stabilizing therapies should be explored as a treatment for endothelial dysfunction in COVID-19, and other severe diseases where endothelial dysfunction has a central role in pathogenesis. Funding Information This work was directly supported by grant funding from the Ted Rogers Center for Heart Research, Toronto, Ontario, Canada and the Peter Munk Cardiac Center, Toronto, Ontario, Canada.
Collapse
Affiliation(s)
- Dakota Gustafson
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Michelle Ngai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Ruilin Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Huayun Hou
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | | | - Clara Erice
- Johns Hopkins School of Medicine, Baltimore, USA
| | - Serena Mandla
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Michael D Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Eddy Fan
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Interdepartmental Division of Critical Care and Institute of Medical Sciences, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Uriel Trahtemberg
- Keenan Research Center for Biomedical Research, Unity Health Toronto, Toronto, Canada; Critical Care Department, Galilee Medical Center, Nahariya, Israel
| | - Andrew Baker
- Interdepartmental Division of Critical Care and Institute of Medical Sciences, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Critical Care Department, Galilee Medical Center, Nahariya, Israel
| | - Chris McIntosh
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada; Techna Institute, University Health Network, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Vector Institute, University of Toronto, Toronto, Canada
| | - Chun-Po S Fan
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care and Institute of Medical Sciences, University of Toronto, Toronto, Canada; Keenan Research Center for Biomedical Research, Unity Health Toronto, Toronto, Canada
| | - Kevin C Kain
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Kate Hanneman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada
| | - Paaladinesh Thavendiranathan
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada; Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, Toronto, Canada
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada.
| | - Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, Canada.
| |
Collapse
|
19
|
Aonuma T, Moukette B, Kawaguchi S, Barupala NP, Sepúlveda MN, Frick K, Tang Y, Guglin M, Raman SV, Cai C, Liangpunsakul S, Nakagawa S, Kim IM. MiR-150 Attenuates Maladaptive Cardiac Remodeling Mediated by Long Noncoding RNA MIAT and Directly Represses Profibrotic Hoxa4. Circ Heart Fail 2022; 15:e008686. [PMID: 35000421 PMCID: PMC9018469 DOI: 10.1161/circheartfailure.121.008686] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND MicroRNA-150 (miR-150) plays a protective role in heart failure (HF). Long noncoding RNA, myocardial infarction-associated transcript (MIAT) regulates miR-150 function in vitro by direct interaction. Concurrent with miR-150 downregulation, MIAT is upregulated in failing hearts, and gain-of-function single-nucleotide polymorphisms in MIAT are associated with increased risk of myocardial infarction (MI) in humans. Despite the correlative relationship between MIAT and miR-150 in HF, their in vivo functional relationship has never been established, and molecular mechanisms by which these 2 noncoding RNAs regulate cardiac protection remain elusive. METHODS We use MIAT KO (knockout), Hoxa4 (homeobox a4) KO, MIAT TG (transgenic), and miR-150 TG mice. We also develop DTG (double TG) mice overexpressing MIAT and miR-150. We then use a mouse model of MI followed by cardiac functional, structural, and mechanistic studies by echocardiography, immunohistochemistry, transcriptome profiling, Western blotting, and quantitative real-time reverse transcription-polymerase chain reaction. Moreover, we perform expression analyses in hearts from patients with HF. Lastly, we investigate cardiac fibroblast activation using primary adult human cardiac fibroblasts and in vitro assays to define the conserved MIAT/miR-150/HOXA4 axis. RESULTS Using novel mouse models, we demonstrate that genetic overexpression of MIAT worsens cardiac remodeling, while genetic deletion of MIAT protects hearts against MI. Importantly, miR-150 overexpression attenuates the detrimental post-MI effects caused by MIAT. Genome-wide transcriptomic analysis of MIAT null mouse hearts identifies Hoxa4 as a novel downstream target of the MIAT/miR-150 axis. Hoxa4 is upregulated in cardiac fibroblasts isolated from ischemic myocardium and subjected to hypoxia/reoxygenation. HOXA4 is also upregulated in patients with HF. Moreover, Hoxa4 deficiency in mice protects the heart from MI. Lastly, protective actions of cardiac fibroblast miR-150 are partially attributed to the direct and functional repression of profibrotic Hoxa4. CONCLUSIONS Our findings delineate a pivotal functional interaction among MIAT, miR-150, and Hoxa4 as a novel regulatory mechanism pertinent to ischemic HF.
Collapse
Affiliation(s)
- Tatsuya Aonuma
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bruno Moukette
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Satoshi Kawaguchi
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nipuni P. Barupala
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marisa N. Sepúlveda
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kyle Frick
- Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Maya Guglin
- Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Subha V. Raman
- Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chenleng Cai
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA;,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Il-man Kim
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA;,Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA;,Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA;,Address for correspondence: Il-man Kim, PhD, Associate Professor, Department of Anatomy, Cell Biology and Physiology, Wells Center for Pediatric Research, Krannert Institute of Cardiology, Indiana University School of Medicine, 635 Barnhill Drive, MS 346A, Indianapolis, IN 46202, USA, , Phone: 317-278-2086
| |
Collapse
|
20
|
Yan W, Feng Y, Lei Z, Kuang W, Long C. MicroRNA-214-3p Ameliorates LPS-Induced Cardiomyocyte Injury by Inhibiting Cathepsin B. Folia Biol (Praha) 2022; 68:78-85. [PMID: 36384265 DOI: 10.14712/fb2022068020078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Myocardial injury is a common complication of sepsis. MicroRNA (miRNA) miR-214-3p is protective against myocardial injury caused by sepsis, but its mechanism in lipopolysaccharide (LPS)- induced cardiomyocyte injury is still unclear. An AC16 cell injury model was induced by LPS treatment. Cell Counting Kit-8 and flow cytometry assay showed decreased cell viability and increased apoptosis in LPS-treated AC16 cells. The levels of caspase- 3, Bax, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), myosin 6 (Myh6), myosin 7 (Myh7), reactive oxygen species (ROS), and malondialdehyde (MDA) were increased in LPS-treated AC16 cells, but the levels of Bcl-2 and superoxide dismutase (SOD) were decreased. MiR-214-3p was down-regulated and cathepsin B (CTSB) was upregulated in LPS-treated AC16 cells. At the same time, miR-214-3p could target CTSB and reduce its expression. We also found that a miR-214-3p mimic or CTSB silencing could significantly reduce LPSinduced apoptosis, decrease ROS, MDA, caspase-3, and Bax and increase SOD and Bcl-2. CTSB silencing could significantly reduce ANP, BNP, Myh6, and Myh7 in LPS-treated AC16 cells. The effects of CTSB silencing were reversed by a miR-214-3p inhibitor. In summary, miR-214-3p could inhibit LPSinduced myocardial injury by targeting CTSB, which provides a new idea for myocardial damage caused by sepsis.
Collapse
Affiliation(s)
- W Yan
- The First Affiliated Hospital, Department of Cardiovascular Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Y Feng
- The First Affiliated Hospital, Department of Cardiovascular Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Z Lei
- The First Affiliated Hospital, Department of Cardiovascular Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - W Kuang
- The First Affiliated Hospital, Department of Cardiovascular Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - C Long
- The First Affiliated Hospital, Department of Cardiovascular Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
21
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
22
|
Yan G, Wang J, Fang Z, Yan S, Zhang Y. MiR-26a-5p Targets WNT5A to Protect Cardiomyocytes from Injury Due to Hypoxia/Reoxygenation Through the Wnt/β-catenin Signaling Pathway. Int Heart J 2021; 62:1145-1152. [PMID: 34544974 DOI: 10.1536/ihj.21-054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study aimed to investigate the effect and mechanism of miR-26a-5p on cardiomyocyte injury induced by hypoxia/reoxygenation (H/R).After construction of an H/R model in rat cardiomyocyte H9c2 cells, miR-26a-5p in the cells was interfered with (cells transfected with miR-26a-5p inhibitor) or overexpressed (cells transfected with a miR-26a-5p mimics). The viability and the apoptosis rate of cells in each group were detected using CCK-8 and flow cytometry; the relationship between miR-26a-5p and WNT5A was verified by a dual-luciferase reporter assay; the expression of miR-26a-5p, WNT5A, cleavedcaspase3 and Wnt/β-catenin signaling pathway-related proteins in each group was detected using qRT-PCR or Western blot; LDH release, SOD, and GSH-PX activities in each group were detected by kit.In the H/R group, the expression level of miR-26a-5p was significantly decreased, whereas the expression level of WNT5A was significantly increased. The activity of the Wnt/β-catenin signaling pathway was up-regulated; the level of LDH released was significantly increased; and activities of SOD and GSH-PX were significantly decreased. The aforementioned changes resulted in decreased cell activity and increased apoptosis rate. The overexpression of miR-26a-5p could reduce the expression level of WNT5A, the activity of the Wnt/β-catenin signaling pathway, and the apoptosis rate and restore the cell viability.These results suggest that miR-26a-5p can target WNT5A and thus, inhibit the Wnt/β-catenin signaling pathway activity, inhibiting H/R-induced cardiomyocyte injury and apoptosis.
Collapse
Affiliation(s)
- Guohui Yan
- Department of Ultrasound, Zhongshan Hospital Xiamen University.,Department of Medicine, Fujian Medical University
| | - Jiajia Wang
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University
| | - Zanxi Fang
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University
| | - Shuidi Yan
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University
| | - Yang Zhang
- Department of Medicine, Fujian Medical University.,Center of Clinical Laboratory, Zhongshan Hospital Xiamen University
| |
Collapse
|
23
|
Aonuma T, Moukette B, Kawaguchi S, Barupala NP, Sepulveda MN, Corr C, Tang Y, Liangpunsakul S, Payne RM, Willis MS, Kim IM. Cardiomyocyte microRNA-150 confers cardiac protection and directly represses pro-apoptotic small proline-rich protein 1A. JCI Insight 2021; 6:e150405. [PMID: 34403363 PMCID: PMC8492334 DOI: 10.1172/jci.insight.150405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
MicroRNA-150 (miR-150) is downregulated in patients with multiple cardiovascular diseases and in diverse mouse models of heart failure (HF). miR-150 is significantly associated with HF severity and outcome in humans. We previously reported that miR-150 is activated by β-blocker carvedilol (Carv) and plays a protective role in the heart using a systemic miR-150 KO mouse model. However, mechanisms that regulate cell-specific miR-150 expression and function in HF are unknown. Here, we demonstrate that potentially novel conditional cardiomyocyte–specific (CM-specific) miR-150 KO (miR-150 cKO) in mice worsens maladaptive cardiac remodeling after myocardial infarction (MI). Genome-wide transcriptomic analysis in miR-150 cKO mouse hearts identifies small proline–rich protein 1a (Sprr1a) as a potentially novel target of miR-150. Our studies further reveal that Sprr1a expression is upregulated in CMs isolated from ischemic myocardium and subjected to simulated ischemia/reperfusion, while its expression is downregulated in hearts and CMs by Carv. We also show that left ventricular SPRR1A is upregulated in patients with HF and that Sprr1a knockdown in mice prevents maladaptive post-MI remodeling. Lastly, protective roles of CM miR-150 are, in part, attributed to the direct and functional repression of proapoptotic Sprr1a. Our findings suggest a crucial role for the miR-150/SPRR1A axis in regulating CM function post-MI.
Collapse
Affiliation(s)
- Tatsuya Aonuma
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Bruno Moukette
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Satoshi Kawaguchi
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Nipuni P Barupala
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Marisa N Sepulveda
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, United States of America
| | - Christopher Corr
- Department of Medicine, Indiana University School of Medicine, Indianapolis, United States of America
| | - Yaoliang Tang
- Department of Medicine, Augusta University, Augusta, United States of America
| | - Suthat Liangpunsakul
- Department of Medicine, Indiana University School of Medicine, Indianapolis, United States of America
| | - R Mark Payne
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, United States of America
| | - Monte S Willis
- Department of Medicine, Indiana University School of Medicine, Indianapolis, United States of America
| | - Il-Man Kim
- Indiana University School of Medicine, Indianapolis, United States of America
| |
Collapse
|
24
|
Moukette B, Barupala NP, Aonuma T, Sepulveda M, Kawaguchi S, Kim IM. Interactions between noncoding RNAs as epigenetic regulatory mechanisms in cardiovascular diseases. Methods Cell Biol 2021; 166:309-348. [PMID: 34752338 DOI: 10.1016/bs.mcb.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cardiovascular diseases (CVDs) represent the foremost cause of mortality in the United States and worldwide. It is estimated that CVDs account for approximately 17.8 million deaths each year. Despite the advances made in understanding cellular mechanisms and gene mutations governing the pathophysiology of CVDs, they remain a significant cause of mortality and morbidity. A major segment of mammalian genomes encodes for genes that are not further translated into proteins. The roles of the majority of such noncoding ribonucleic acids (RNAs) have been puzzling for a long time. However, it is becoming increasingly clear that noncoding RNAs (ncRNAs) are dynamically expressed in different cell types and have a comprehensive selection of regulatory roles at almost every step involved in DNAs, RNAs and proteins. Indeed, ncRNAs regulate gene expression through epigenetic interactions, through direct binding to target sequences, or by acting as competing endogenous RNAs. The profusion of ncRNAs in the cardiovascular system suggests that they may modulate complex regulatory networks that govern cardiac physiology and pathology. In this review, we summarize various functions of ncRNAs and highlight the recent literature on interactions between ncRNAs with an emphasis on cardiovascular disease regulation. Furthermore, as the broad-spectrum of ncRNAs potentially establishes new avenues for therapeutic development targeting CVDs, we discuss the innovative prospects of ncRNAs as therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bruno Moukette
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nipuni P Barupala
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tatsuya Aonuma
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Marisa Sepulveda
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Satoshi Kawaguchi
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Il-Man Kim
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States; Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
25
|
Huang P, Bai L, Liu L, Fu J, Wu K, Liu H, Liu Y, Qi B, Qi B. Redd1 knockdown prevents doxorubicin-induced cardiac senescence. Aging (Albany NY) 2021; 13:13788-13806. [PMID: 33962393 PMCID: PMC8202877 DOI: 10.18632/aging.202972] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Regulated in development and DNA damage response-1 (Redd1) is a stress-response gene that is transcriptionally induced by diverse stressful stimuli to influence cellular growth and survival. Although evidence suggests that aging may drive Redd1 expression in skeletal muscles, the expression patterns and functions of Redd1 in senescent cardiomyocytes remain unspecified. To address this issue, in vitro and in vivo models of cardiomyocyte senescence were established by administration of doxorubicin (Dox). Redd1 overexpression and knockdown was achieved in cultured H9c2 cardiomyocytes and mouse tissues using, respectively, lentivirals and adeno-associated virus 9 (AAV9) vectors. In the hearts of both aged (24 months old) and Dox-treated mice, as well as in Dox-exposed H9c2 cardiomyocytes, high Redd1 expression accompanied the increase in both cellular senescence markers (p16INK4a and p21) and pro-inflammatory cytokine expression indicative of a stress-associated secretory phenotype (SASP). Notably, Redd1 overexpression accentuated, whereas Redd1 silencing markedly attenuated, Dox-induced cardiomyocyte senescence features both in vitro and in vivo. Notably, AAV9-shRNA-mediated Redd1 silencing significantly alleviated Dox-induced cardiac dysfunction. Moreover, through pharmacological inhibition, immunofluorescence, and western blotting, signaling pathway analyses indicated that Redd1 promotes cardiomyocyte senescence as a downstream effector of p38 MAPK to promote NF-kB signaling via p65 phosphorylation and nuclear translocation.
Collapse
Affiliation(s)
- Pianpian Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- Department of Geriatrics, Wuhan No.1 Hospital, Wuhan, Hubei 430022, China
| | - Lijuan Bai
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lihua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jun Fu
- Department of Radiology, Wuhan No.1 Hospital, Wuhan, Hubei 430022, China
| | - Kefei Wu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hongxia Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Benming Qi
- Department of Otorhinolaryngology, First People’s Hospital of Yunnan Province, Kunming, Yunnan 650000, China
| | - Benling Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
26
|
Liu H, Sun S, Liu B. Smurf2 exerts neuroprotective effects on cerebral ischemic injury. J Biol Chem 2021; 297:100537. [PMID: 33722608 PMCID: PMC8363835 DOI: 10.1016/j.jbc.2021.100537] [Citation(s) in RCA: 10] [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: 07/13/2020] [Revised: 02/20/2021] [Accepted: 03/10/2021] [Indexed: 11/17/2022] Open
Abstract
The present study aimed to explore specific mechanisms involved in mediating the neuroprotective effects of Smad ubiquitination regulatory factor 2 (Smurf2) in cerebral ischemic injury. A middle cerebral artery occlusion (MCAO) mouse model and an oxygen–glucose deprivation (OGD)–treated neuron model were developed. The expression of Smurf2, Yin Yang 1 (YY1), hypoxia-inducible factor-1 alpha (HIF1α), and DNA damage–inducible transcript 4 gene (DDIT4) was analyzed. Thereafter, the expression of Smurf2, YY1, HIF1α, and DDIT4 was altered in the MCAO mice and OGD-treated neurons. Apoptosis in tissues and cerebral infarction were assessed. In neurons, the expression of apoptosis-related proteins, viability, and apoptosis were assessed, followed by evaluation of lactate dehydrogenase leakage rate. The interaction between Smurf2 and YY1 was analyzed by coimmunoprecipitation assay and that between YY1 ubiquitination by in vivo ubiquitination experiment. The results showed downregulation of Smurf2 and upregulation of YY1, HIF1α, and DDIT4 in both MCAO mice and OGD-treated neurons. Smurf2 elevated YY1 ubiquitination and degradation, and YY1 increased HIF1α expression to promote DDIT4 in neurons. Overexpressed Smurf2 or downregulated YY1, HIF1α, or DDIT4 reduced the volume of cerebral infarction and apoptosis in MCAO mice, while enhancing cell viability and reducing apoptosis and lactate dehydrogenase leakage in OGD-treated neurons. In summary, our findings elucidated a neuroprotective role of Smurf2 in cerebral ischemic injury via inactivation of the YY1/HIF1α/DDIT4 axis.
Collapse
Affiliation(s)
- Haibin Liu
- Department of Paediatrics, Linyi People's Hospital, Linyi, China
| | - Shengtao Sun
- Department of Paediatrics, Linyi People's Hospital, Linyi, China
| | - Bing Liu
- Department of Paediatrics, Linyi People's Hospital, Linyi, China.
| |
Collapse
|
27
|
Tian X, Hu Y, Liu Y, Yang Z, Xie H, Zhou L, Zheng S. Circular RNA Microarray Analyses in Hepatic Ischemia-Reperfusion Injury With Ischemic Preconditioning Prevention. Front Med (Lausanne) 2021; 8:626948. [PMID: 33763433 PMCID: PMC7982475 DOI: 10.3389/fmed.2021.626948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic preconditioning (IPC) represents an effective intervention to relieve hepatic ischemia-reperfusion injury (IRI). Systematic detection of circRNA expression revealing the protection effect of IPC still remains to be elucidated. Here, we applied a microarray to detect circRNA and mRNA expression in ischemic liver with and without IPC (n = 3 in each group). Compared with the sham group, there were 39 circRNAs and 432 mRNAs increased and 38 circRNAs and 254 mRNAs decreased (fold change ≥1.5, P < 0.05) in the group of hepatic IRI. As the result of IPC intervention, 43 circRNAs and 64 mRNAs were increased, and 7 circRNAs and 31 mRNAs were decreased in the IPC group when compared with IRI. We then identified circRNA_017753 as the most possible target that may closely relate to IPC protective signaling and predicted Jade1 as the target related to circRNA_017753. Three possible circRNA-miRNA-mRNA axes were constructed that may play a vital role in protective mechanisms in IPC. The study for the first time systematically detects the dysregulated circRNAs and mRNAs in response to hepatic IRI and IPC intervention. Our profile and bioinformatic analysis provide numerous novel clues to understanding the pathophysiologic mechanism of IPC protection against hepatic IRI.
Collapse
Affiliation(s)
- Xinyao Tian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, National Health Commission of PRC, Hangzhou, China
| | - Yan Hu
- Department of Pharmacy, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuanxing Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhe Yang
- Department of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Haiyang Xie
- Key Laboratory of Combined Multi-organ Transplantation, National Health Commission of PRC, Hangzhou, China
| | - Lin Zhou
- Key Laboratory of Combined Multi-organ Transplantation, National Health Commission of PRC, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, National Health Commission of PRC, Hangzhou, China.,Department of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan (Hangzhou) Hospital, Hangzhou, China
| |
Collapse
|
28
|
Li T, Zhang S, Chen F, Hu J, Yuan S, Li C, Wang X, Zhang W, Tang R. Formononetin ameliorates the drug resistance of Taxol resistant triple negative breast cancer by inhibiting autophagy. Am J Transl Res 2021; 13:497-514. [PMID: 33594306 PMCID: PMC7868832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
Characterized by autophagy-associated protein disorders, autophagy participates in Taxol resistance in triple negative breast cancer (TNBC). As an evolutionarily conserved serine/threonine protein kinase with complex signaling pathway, mammalian target of rapamycin (mTOR) can regulate various cellular functions by phosphorylation of its downstream target proteins after activation. A large number of references have demonstrated that mTOR signaling pathway is related to autophagy and apoptosis. Formononetin (FMNT) has anticancer properties against breast, prostate and colon cancers. This study aimed to explore the regulatory effect of FMNT/miR-199a-3p/mTOR pathway on Taxol resistance and autophagy in breast cancer (BC). MiR-199a-3p, mTOR, LC3 and other autophagy related proteins were detected in Taxol sensitive and Taxol resistant TNBC cell lines, which were MDA-MB-231 and MDA-MB-231/Taxol, respectively. Cell viability and toxicity were determined by CCK-8 and MTT assay, respectively. The therapeutic effect of FMNT was evaluated in xenotransplantation model of nude mice. MiR-199a-3p was more highly expressed in MDA-MB-231/Taxol than in MDA-MB-231, while mTOR and p-mTOR decreased in MDA-MB-231/Taxol in comparison with MDA-MB-231, and autophagy activation and drug resistance were enhanced. In MDA-MB-231/Taxol cell line, the role of FMNT was verified to inhibit high miR-199a-3p expression. In addition, the combination therapy of FMNT and Taxol was found to be more effective in inhibiting autophagy and drug resistance. Moreover, mTOR was the target of miR-199a-3p, which was confirmed by dual luciferase reporter (DLR) gene assay. Oral administration of FMNT reduced tumor volume after MDA-MB-231/Taxol injection in vivo. Moreover, oral administration of FMNT and Taxol suppressed autophagy and Taxol resistance by restoring mTOR protein level to that of the parent MDA-MB-231, suggesting that miR-199a-3p can severe as a new target to overcome Taxol resistance in TNBC.
Collapse
Affiliation(s)
- Tian Li
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Shiyi Zhang
- The Second School of Clinical Medicine of Guangdong Medical UniversityDongguan 523000, China
| | - Fengsong Chen
- Nantong Haimen People’s HospitalNantong 226100, China
| | - Jun Hu
- Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Shuai Yuan
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Chaoran Li
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Xiaoting Wang
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Weihong Zhang
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Runwei Tang
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| |
Collapse
|
29
|
Liu B, Wang B, Zhang X, Lock R, Nash T, Vunjak-Novakovic G. Cell type-specific microRNA therapies for myocardial infarction. Sci Transl Med 2021; 13:eabd0914. [PMID: 33568517 PMCID: PMC8848299 DOI: 10.1126/scitranslmed.abd0914] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
Current interventions fail to recover injured myocardium after infarction and prompt the need for development of cardioprotective strategies. Of increasing interest is the therapeutic use of microRNAs to control gene expression through specific targeting of mRNAs. In this Review, we discuss current microRNA-based therapeutic strategies, describing the outcomes and limitations of key microRNAs with a focus on target cell types and molecular pathways. Last, we offer a perspective on the outlook of microRNA therapies for myocardial infarction, highlighting the outstanding challenges and emerging strategies.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bryan Wang
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor Nash
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| |
Collapse
|
30
|
Zeng Z, Ma H, Chen J, Huang N, Zhang Y, Su Y, Zhang H. Knockdown of miR-1275 protects against cardiomyocytes injury through promoting neuromedin U type 1 receptor. Cell Cycle 2020; 19:3639-3649. [PMID: 33323026 DOI: 10.1080/15384101.2020.1860310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to assess the role of miR-1275 in cardiac ischemia reperfusion injury. H9 human embryonic stem cell (hESC)-derived cardiomyocytes stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were used to simulate myocardial injury in vitro. miR-1275 expression levels in cells were measured by RT-qPCR. The release of lactate dehydrogenase (LDH) and creatine kinase (CK) was examined through LDH and CK ELISA kits. Cell apoptosis was detected through flow cytometry. A Fura-2 Calcium Flux Assay Kit and a Fluo-4 assay kit were used to determine the Ca2+ concentration. Expression levels of proteins were tested by Western blotting. The binding effect of miR-1275 and neuromedin U type 1 receptor (NMUR1) was detected by dual-luciferase activity assay. The results showed that miR-1275 was upregulated in OGD/R-stimulated cardiomyocytes. Inhibition of miR-1275 suppressed the increased activity of LDH and CK, cell apoptosis, reactive oxygen species (ROS) production, intracellular Ca2+ concentration and sarcoplasmic reticulum (SR) Ca2+ leak induced by OGD/R treatment in cardiomyocytes. miR-1275 directly targets 3'UTR of NMUR1 and negatively regulates NMUR1 expression. Silence of NMUR1 abolished the protecting effect of the miR-1275 antagomir on myocardial OGD/R injury. Our study indicated that the miR-1275 antagomir protects cardiomyocytes from OGD/R injury through the promotion of NMUR1.
Collapse
Affiliation(s)
- Zhu Zeng
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Haixin Ma
- Medical Department, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Jing Chen
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Nina Huang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Yudan Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Yufei Su
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Huifang Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| |
Collapse
|
31
|
Li S, Xie Y, Yang B, Huang S, Zhang Y, Jia Z, Ding G, Zhang A. MicroRNA-214 targets COX-2 to antagonize indoxyl sulfate (IS)-induced endothelial cell apoptosis. Apoptosis 2020; 25:92-104. [PMID: 31820187 DOI: 10.1007/s10495-019-01582-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cardiovascular disease (CVD) serves as the major cause of mortality in chronic kidney disease (CKD) patients. The injury of endothelium associated with the long-term challenge of uremic toxins including the toxic indoxyl sulfate (IS) is one of key pathological factors leading to CVD. However, the mechanisms of uremic toxins, especially the IS, resulting in endothelial injury, remain unclear. miR-214 was reported to contribute to the pathogenesis of cardiovascular diseases, while its role in IS-induced endothelial cell apoptosis is unknown. In this study, we investigated the role of microRNA-214 (miR-214) in IS-induced endothelial cell apoptosis and the underlying mechanisms using mouse aortic endothelial cells (MAECs). Following IS treatment, miR-214 was significantly downregulated in MAECs in line with enhanced cell apoptosis. Meanwhile, COX-2 was upregulated at both mRNA and protein levels along with increased secretion of PGE2 in medium. To define the role of miR-214 in IS-induced endothelial cell apoptosis, we modulated miR-214 level in MAECs and found that overexpression of miR-214 markedly attenuated endothelial cell apoptosis, while antagonism of miR-214 deteriorated cell death after IS challenge. Further analyses confirmed that COX-2 is a target gene of miR-214, and the inhibition of COX-2 by a specific COX-2 inhibitor NS-398 strikingly attenuated IS-induced endothelial cell apoptosis along with a significant blockade of PGE2 secretion. In conclusion, this study demonstrated an important role of miR-214 in protecting against endothelial cell damage induced by IS possibly by direct downregulation of COX-2/PGE2 axis.
Collapse
Affiliation(s)
- Shuzhen Li
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Yifan Xie
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Bingyu Yang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Guixia Ding
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, People's Republic of China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
32
|
Shirazi-Tehrani E, Firouzabadi N, Tamaddon G, Bahramali E, Vafadar A. Carvedilol Alters Circulating MiR-1 and MiR-214 in Heart Failure. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:375-383. [PMID: 32943906 PMCID: PMC7481348 DOI: 10.2147/pgpm.s263740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/11/2020] [Indexed: 02/01/2023]
Abstract
Introduction MicroRNAs (miRNAs) are recognized as major contributors in various cardiovascular diseases, such as heart failure (HF). These small noncoding RNAs that posttranscriptionally control target genes are involved in regulating different pathophysiological processes including cardiac proliferation, ifferentiation, hypertrophy, and fibrosis. Although carvedilol, a β-adrenergic blocker, and a drug of choice in HF produce cytoprotective actions against cardiomyocyte hypertrophy, the mechanisms are poorly understood. Here we proposed that the expression of hypertrophic-specific miRNAs (miR-1, miR-133, miR-208, and miR-214) might be linked to beneficial effects of carvedilol. Methods The levels of four hypertrophic-specific miRNAs were measured in the sera of 35 patients with systolic HF receiving carvedilol (treated) and 20 HF patients not receiving any β-blockers (untreated) as well as 17 nonHF individuals (healthy) using quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Systolic HF was defined as left ventricular ejection fraction <50% by transthoracic echocardiography. Results We demonstrated that miR-1 and miR-214 were significantly upregulated in the treated group compared to the untreated group (P=0.014 and 5.3-fold, 0.033 and 4.2-fold, respectively). However, miR-133 and miR-208 did not show significant difference in expression between these two study groups. MiR-1 was significantly downregulated in the untreated group compared with healthy individuals (P=0.019 and 0.14-fold). Conclusion In conclusion, it might be postulated that one of the mechanisms by which carvedilol may exert its cardioprotective effects can be through increasing miR-1 and miR-214 expressions which may also serve as a potential therapeutic target in patients with systolic HF in future.
Collapse
Affiliation(s)
- Elham Shirazi-Tehrani
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Gholamhossein Tamaddon
- Department of Medical Biotechnology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Bahramali
- Digestive Disease Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Asma Vafadar
- Department of Medical Biotechnology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
33
|
Samidurai A, Xi L, Salloum FN, Das A, Kukreja RC. PDE5 inhibitor sildenafil attenuates cardiac microRNA 214 upregulation and pro-apoptotic signaling after chronic alcohol ingestion in mice. Mol Cell Biochem 2020; 471:189-201. [PMID: 32535704 PMCID: PMC10801845 DOI: 10.1007/s11010-020-03779-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
Abusive chronic alcohol consumption can cause metabolic and functional derangements in the heart and is a risk factor for development of non-ischemic cardiomyopathy. microRNA 214 (miR-214) is a molecular sensor of stress signals that negatively impacts cell survival. Considering cardioprotective and microRNA modulatory effects of sildenafil, a phosphodiesterase 5 (PDE5) inhibitor, we investigated the impact of chronic alcohol consumption on cardiac expression of miR-214 and its anti-apoptotic protein target, Bcl-2 and whether sildenafil attenuates such changes. Adult male FVB mice received unlimited access to either normal liquid diet (control), alcohol diet (35% daily calories intake), or alcohol + sildenafil (1 mg/kg/day, p.o.) for 14 weeks (n = 6-7/group). The alcohol-fed groups with or without sildenafil had increased total diet consumption and lower body weight as compared with controls. Echocardiography-assessed left ventricular function was unaltered by 14-week alcohol intake. Alcohol-fed group had 2.6-fold increase in miR-214 and significant decrease in Bcl-2 expression, along with enhanced phosphorylation of ERK1/2 and cleavage of PARP (marker of apoptotic DNA damage) in the heart. Co-ingestion with sildenafil blunted the alcohol-induced increase in miR-214, ERK1/2 phosphorylation, and maintained Bcl-2 and decreased PARP cleavage levels. In conclusion, chronic alcohol consumption triggers miR-214-mediated pro-apoptotic signaling in the heart, which was prevented by co-treatment with sildenafil. Thus, PDE5 inhibition may serve as a novel protective strategy against cardiac apoptosis due to chronic alcohol abuse.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| | - Lei Xi
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| | - Fadi N Salloum
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| | - Anindita Das
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| | - Rakesh C Kukreja
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA.
- Division of Cardiology, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA.
| |
Collapse
|
34
|
Okazaki Y, Chew SH, Nagai H, Yamashita Y, Ohara H, Jiang L, Akatsuka S, Takahashi T, Toyokuni S. Overexpression of miR-199/214 is a distinctive feature of iron-induced and asbestos-induced sarcomatoid mesothelioma in rats. Cancer Sci 2020; 111:2016-2027. [PMID: 32248600 PMCID: PMC7293088 DOI: 10.1111/cas.14405] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Malignant mesothelioma (MM) is one of the most lethal tumors in humans. The onset of MM is linked to exposure to asbestos, which generates reactive oxygen species (ROS). ROS are believed to be derived from the frustrated phagocytosis and the iron in asbestos. To explore the pathogenesis of MM, peritoneal MM was induced in rats by the repeated intraperitoneal injection of iron saccharate and nitrilotriacetate. In the present study, we used microarray techniques to screen the microRNA (miR) expression profiles of these MM. We observed that the histological subtype impacted the hierarchical clustering of miR expression profiles and determined that miR-199/214 is a distinctive feature of iron saccharate-induced sarcomatoid mesothelioma (SM). Twist1, a transcriptional regulator of the epithelial-mesenchymal transition, has been shown to activate miR-199/214 transcription; thus, the expression level of Twist1 was examined in iron-induced and asbestos-induced mesotheliomas in rats. Twist1 was exclusively expressed in iron saccharate-induced SM but not in the epithelioid subtype. The Twist1-miR-199/214 axis is activated in iron saccharate-induced and asbestos-induced SM. The expression levels of miR-214 and Twist1 were correlated in an asbestos-induced MM cell line, suggesting that the Twist1-miR-199/214 axis is preserved. MeT5A, an immortalized human mesothelial cell line, was used for the functional analysis of miR. The overexpression of miR-199/214 promoted cellular proliferation, mobility and phosphorylation of Akt and ERK in MeT5A cells. These results indicate that miR-199/214 may affect the aggressive biological behavior of SM.
Collapse
Affiliation(s)
- Yasumasa Okazaki
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Shan Hwu Chew
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Hirotaka Nagai
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoriko Yamashita
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Hiroki Ohara
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Li Jiang
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinya Akatsuka
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| | - Takashi Takahashi
- Division of Molecular CarcinogenesisNagoya University Graduate School of MedicineNagoyaJapan
- Aichi Cancer Center Research InstituteNagoyaJapan
| | - Shinya Toyokuni
- Department of Pathology and Biological ResponsesNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
35
|
Caparosa EM, Sedgewick AJ, Zenonos G, Zhao Y, Carlisle DL, Stefaneanu L, Jankowitz BT, Gardner P, Chang YF, Lariviere WR, LaFramboise WA, Benos PV, Friedlander RM. Regional Molecular Signature of the Symptomatic Atherosclerotic Carotid Plaque. Neurosurgery 2020; 85:E284-E293. [PMID: 30335165 DOI: 10.1093/neuros/nyy470] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Many studies have explored molecular markers of carotid plaque development and vulnerability to rupture, usually having examined whole carotid plaques. However, there are regional differences in plaque morphology and known shear-related mechanisms in areas surrounding the lipid core. OBJECTIVE To determine whether there are regional differences in protein expression along the long axis of the carotid plaque and how that might produce gaps in our understanding of the carotid plaque molecular signature. METHODS Levels of 7 inflammatory cytokines (IL-1β, IL-6, IL-8, IL-10, IL-12 p70, IFN-γ, and TNF-α) and caspase-3 were analyzed in prebifurcation, bifurcation, and postbifurcation segments of internal carotid plaques surgically removed from symptomatic and asymptomatic patients. Expression profiles of miRNAs and mRNAs were determined with microarrays for the rupture-prone postbifurcation segment for comparison with published whole plaque results. RESULTS Expression levels of all proteins examined, except IL-10, were lowest in the prebifurcation segment and significantly higher in the postbifurcation segment. Patient group differences in protein expression were observed for the prebifurcation segment; however, no significant differences were observed in the postbifurcation segment between symptomatic and asymptomatic patients. Expression profiles from postbifurcation carotid plaques identified 4 novel high priority miRNAs differentially expressed between patient groups (miR-214, miR-484, miR-942, and miR-1287) and 3 high-confidence miRNA:mRNA targets, including miR-214:APOD, miR-484:DACH1, and miR-942:GPR56. CONCLUSION The results demonstrate regional differences in protein expression for the first time and show that focus on the rupture-prone postbifurcation region leads to prioritization for further study of novel miRNA gene regulation mechanisms.
Collapse
Affiliation(s)
- Ellen M Caparosa
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew J Sedgewick
- Joint Carnegie-Mellon -University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Georgios Zenonos
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yin Zhao
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Diane L Carlisle
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lucia Stefaneanu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian T Jankowitz
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Paul Gardner
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yue-Fang Chang
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William R Lariviere
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Joint Carnegie-Mellon -University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, Pennsylvania
| | - Robert M Friedlander
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Lu Y, Xi J, Zhang Y, Li C, Chen W, Hu X, Zhang M, Zhang F, Wei H, Li Z, Wang Z. MicroRNA-214-5p protects against myocardial ischemia reperfusion injury through targeting the FAS ligand. Arch Med Sci 2020; 16:1119-1129. [PMID: 32864001 PMCID: PMC7444694 DOI: 10.5114/aoms.2019.85405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/16/2019] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION MicroRNAs (miRNAs) are considered as crucial modulators in myocardial ischemia and reperfusion (I/R) injury. The present study aimed to investigate the expression and biological functions of miR-214-5p via targeting Fas ligand (FASLG) in I/R injury. MATERIAL AND METHODS Lactate dehydrogenase, casein kinase, malondialdehyde assay, reactive oxygen species (ROS) detection and cell apoptosis analysis measured cell damage and cell apoptosis in H9c2 cells under hypoxia/reperfusion (H/R) treatment. Bioinformatics and dual luciferase reporter assays demonstrated the molecular mechanism of miR-214-5p in cardiac cells. 2,3,5-Triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (HE) staining and adenovirus injection were performed in I/R treated mice. RESULTS The expression of miR-214-5p was decreased in H/R injured H9c2 cells compared with control cells (p < 0.001). Overexpression of miR-214-5p reduced cell damage and apoptosis in H9c2 cells under H/R treatment (p < 0.001). Further study revealed that FASLG was a target of miR-214-5p. Enhanced expression of FASLG attenuated the protective function of miR-214-5p in H9c2 cells subjected to H/R injury (P < 0.001). Moreover, the elevated expression of miR-214-5p by adenovirus injection protected cardiac cells from I/R injury in mice (n = 6/per group). CONCLUSIONS We found that miR-214-5p exerted a protective role in I/R injured cardiac cells by direct targeting FASLG in vitro and in vivo.
Collapse
Affiliation(s)
- Yuan Lu
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jue Xi
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yao Zhang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chenzong Li
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wensu Chen
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaoqin Hu
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Min Zhang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fengyun Zhang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Wei
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhi Li
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhirong Wang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
37
|
Huang P, Fu J, Chen L, Ju C, Wu K, Liu H, Liu Y, Qi B, Qi B, Liu L. Redd1 protects against post‑infarction cardiac dysfunction by targeting apoptosis and autophagy. Int J Mol Med 2019; 44:2065-2076. [PMID: 31638187 PMCID: PMC6844599 DOI: 10.3892/ijmm.2019.4366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Post-infarction remodeling is accompanied and influenced by perturbations in the mammalian target of rapamycin (mTOR) signaling. Regulated in development and DNA damage response-1 (Redd1) has been reported to be involved in DNA repair and modulation of mTOR activity. However, little is known about the role of Redd1 in the heart. In the present study the potential contribution of Redd1 overex-pression to the chronic phase of heart failure after myocardial infarction (MI) was explored and the mechanisms underlying Redd1 actions were determined. Redd1 was downregulated in the mouse heart subjected to MI surgery. To determine the role of Redd1 in the process of MI, adeno-associated virus 9 mediated overexpression of Redd1 was used to enhance Redd1 content in cardiomyocytes. Redd1 overexpression improved left ventricular dysfunction and reduced the expansion index. Additionally, Redd1 overexpression resulted in suppressed myocardial apoptosis and improved autophagy. Furthermore, the studies revealed that Redd1 overexpression could inhibit the phosphorylation of mTOR and its downstream effectors P70/S6 kinase and 4EBP1. In conclusion, this study demonstrated that Redd1 overexpression protects against the development and persistence of heart failure post MI by reducing apoptosis and enhancing autophagy via the mTOR signaling pathway. The present study clearly demonstrated that Redd1 is a therapeutic target in the development of heart failure after MI.
Collapse
Affiliation(s)
- Pianpian Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jun Fu
- Department of Radiology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Long Chen
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chenhui Ju
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kefei Wu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hongxia Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Benming Qi
- Department of Otorhinolaryngology, First People's Hospital of Yunnan Province, Kunming, Yunnan 650000, P.R. China
| | - Benling Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lihua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
38
|
Yang H, Qin X, Wang H, Zhao X, Liu Y, Wo HT, Liu C, Nishiga M, Chen H, Ge J, Sayed N, Abilez OJ, Ding D, Heilshorn SC, Li K. An in Vivo miRNA Delivery System for Restoring Infarcted Myocardium. ACS NANO 2019; 13:9880-9894. [PMID: 31149806 PMCID: PMC7930012 DOI: 10.1021/acsnano.9b03343] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
A major challenge in myocardial infarction (MI)-related heart failure treatment using microRNA is the efficient and sustainable delivery of miRNAs into myocardium to achieve functional improvement through stimulation of intrinsic myocardial restoration. In this study, we established an in vivo delivery system using polymeric nanoparticles to carry miRNA (miNPs) for localized delivery within a shear-thinning injectable hydrogel. The miNPs triggered proliferation of human embryonic stem cell-derived cardiomyocytes and endothelial cells (hESC-CMs and hESC-ECs) and promoted angiogenesis in hypoxic conditions, showing significantly lower cytotoxicity than Lipofectamine. Furthermore, one injected dose of hydrogel/miNP in MI rats demonstrated significantly improved cardiac functions: increased ejection fraction from 45% to 64%, reduced scar size from 20% to 10%, and doubled capillary density in the border zone compared to the control group at 4 weeks. As such, our results indicate that this injectable hydrogel/miNP composite can deliver miRNA to restore injured myocardium efficiently and safely.
Collapse
Affiliation(s)
- Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
- Corresponding Authors.,
| | - Xulei Qin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Huiyuan Wang
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Xin Zhao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Yonggang Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Hung-Ta Wo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Masataka Nishiga
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Haodong Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jing Ge
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Oscar J. Abilez
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Kai Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States
- Corresponding Authors.,
| |
Collapse
|
39
|
Du Y, Yang X, Gong Q, Xu Z, Cheng Y, Su G. Inhibitor of growth 4 affects hypoxia-induced migration and angiogenesis regulation in retinal pigment epithelial cells. J Cell Physiol 2019; 234:15243-15256. [PMID: 30667053 DOI: 10.1002/jcp.28170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Inhibitor of growth 4 (ING4), a potential tumor suppressor, is implicated in cell migration and angiogenesis. However, its effects on diabetic retinopathy (DR) have not been elucidated. In this study, we aimed to evaluate ING4 expression in normal and diabetic rats and clarify its effects on hypoxia-induced dysfunction in human retinal pigment epithelial (ARPE-19) cells. A Type 1 diabetic model was generated by injecting rats intraperitoneally with streptozotocin and then killed them 4, 8, or 12 weeks later. ING4 expression in retinal tissue was detected using western blot analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and immunohistochemistry assays. After transfection with an ING4 overexpression lentiviral vector or small interfering RNA (siRNA), ARPE-19 migration under hypoxia was tested using wound healing and transwell assays. The angiogenic effect of conditioned medium (CM) from ARPE-19 cells was examined by assessing human retinal endothelial cell (HREC) capillary tube formation. Additionally, western blot analysis and RT-qPCR were performed to investigate the signaling pathways in which ING4, specificity protein 1 (Sp1), matrix metalloproteinase 2 (MMP-2), MMP-9, and vascular endothelial growth factor A (VEGF-A) were involved. Here, we found that ING4 expression was significantly reduced in the diabetic rats' retinal tissue. Silencing ING4 aggravated hypoxia-induced ARPE-19 cell migration. CM collected from ING4 siRNA-transfected ARPE-19 cells under hypoxia promoted HREC angiogenesis. These effects were reversed by ING4 overexpression. Furthermore, ING4 suppressed MMP-2, MMP-9, and VEGF-A expression in an Sp1-dependent manner in hypoxia-conditioned ARPE-19 cells. Overall, our results provide valuable mechanistic insights into the protective effects of ING4 on hypoxia-induced migration and angiogenesis regulation in ARPE-19 cells. Restoring ING4 may be a novel strategy for treating DR.
Collapse
Affiliation(s)
- Yang Du
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyue Yang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Qiaoyun Gong
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zhixiang Xu
- Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yan Cheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
40
|
Joris V, Gomez EL, Menchi L, Lobysheva I, Di Mauro V, Esfahani H, Condorelli G, Balligand JL, Catalucci D, Dessy C. MicroRNA-199a-3p and MicroRNA-199a-5p Take Part to a Redundant Network of Regulation of the NOS (NO Synthase)/NO Pathway in the Endothelium. Arterioscler Thromb Vasc Biol 2019; 38:2345-2357. [PMID: 29976767 DOI: 10.1161/atvbaha.118.311145] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective- Members of the microRNA (miR)-199a family, namely miR-199a-5p and miR-199a-3p, have been recently identified as potential regulators of cardiac homeostasis. Also, upregulation of miR-199a expression in cardiomyocytes was reported to influence endothelial cells. Whether miR-199a is expressed by endothelial cells and, if so, whether it directly regulates endothelial function remains unknown. We investigate the implication of miR-199a products on endothelial function by focusing on the NOS (nitric oxide synthase)/NO pathway. Approach and Results- Bovine aortic endothelial cells were transfected with specific miRNA inhibitors (locked-nucleic acids), and potential molecular targets identified with prediction algorithms were evaluated by Western blot or immunofluorescence. Ex vivo experiments were performed with mice treated with antagomiRs targeting miR-199a-3p or -5p. Isolated vessels and blood were used for electron paramagnetic resonance or myograph experiments. eNOS (endothelial NO synthase) activity (through phosphorylations Ser1177/Thr495) is increased by miR-199a-3p/-5p inhibition through an upregulation of the PI3K (phosphoinositide 3-kinase)/Akt (protein kinase B) and calcineurin pathways. SOD1 (superoxide dismutase 1) and PRDX1 (peroxiredoxin 1) upregulation was also observed in locked-nucleic acid-treated cells. Moreover, miR-199a-5p controls angiogenesis and VEGFA (vascular endothelial growth factor A) production and upregulation of NO-dependent relaxation were observed in vessels from antagomiR-treated mice. This was correlated with increased circulated hemoglobin-NO levels and decreased superoxide production. Angiotensin infusion for 2 weeks also revealed an upregulation of miR-199a-3p/-5p in vascular tissues. Conclusions- Our study reveals that miR-199a-3p and miR-199a-5p participate in a redundant network of regulation of the NOS/NO pathway in the endothelium. We highlighted that inhibition of miR-199a-3p and -5p independently increases NO bioavailability by promoting eNOS activity and reducing its degradation, thereby supporting VEGF-induced endothelial tubulogenesis and modulating vessel contractile tone.
Collapse
Affiliation(s)
- Virginie Joris
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Elvira Leon Gomez
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Lisa Menchi
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | | | - Vittoria Di Mauro
- Humanitas University, Rozzano (Milan), Italy (V.D.M., G.C.).,Humanitas#8232, Clinical and Research Center, Rozzano (Milan), Italy (V.D.M., G.C., D.C.).,Institute of Genetics and Biomedical Research, Milan Unit, National Research Council, Italy (V.D.M., G.C., D.C.)
| | - Hrag Esfahani
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Gianluigi Condorelli
- Humanitas University, Rozzano (Milan), Italy (V.D.M., G.C.).,Humanitas#8232, Clinical and Research Center, Rozzano (Milan), Italy (V.D.M., G.C., D.C.).,Institute of Genetics and Biomedical Research, Milan Unit, National Research Council, Italy (V.D.M., G.C., D.C.)
| | - Jean-Luc Balligand
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Daniele Catalucci
- Humanitas#8232, Clinical and Research Center, Rozzano (Milan), Italy (V.D.M., G.C., D.C.).,Institute of Genetics and Biomedical Research, Milan Unit, National Research Council, Italy (V.D.M., G.C., D.C.)
| | - Chantal Dessy
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| |
Collapse
|
41
|
Refaie MMM, El-Hussieny M, Bayoumi AMA, Shehata S. Mechanisms mediating the cardioprotective effect of carvedilol in cadmium induced cardiotoxicity. Role of eNOS and HO1/Nrf2 pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 70:103198. [PMID: 31154273 DOI: 10.1016/j.etap.2019.103198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/30/2019] [Accepted: 05/24/2019] [Indexed: 06/09/2023]
Abstract
Cadmium (Cd) is a highly toxic heavy metal with several harmful effects including cardiotoxicity. For the first time, we aimed to evaluate the possible cardioprotective effect of carvedilol (CAR) in Cd induced cardiotoxicity and study the mechanisms involved in such protection including endothelial nitric oxide synthase (eNOS) and HO1/Nrf2 pathway. CAR (1,10 mg/kg/d) was administered orally for 4 weeks with Cd induced cardiac injury (3 mg/kg/d) orally for 4 weeks. We measured cardiac enzymes, mean arterial pressure changes, heme oxygenase-1 (HO1) and total antioxidant capacity (TAC). Moreover; cardiac tissue malondialdehyde (MDA), tumor necrosis factor alpha (TNFα), western blotting of caspase3 and eNOS levels and histopathology were evaluated. Immunoexpression of eNOS in cardiac tissue, gene expression changes of HO1, and nuclear factor erythroid 2-related factor 2 (Nrf2) using real time polymerase chain reactions (rtPCR) were detected. Our results showed that CAR could significantly decrease Cd induced cardiotoxicity.
Collapse
Affiliation(s)
- Marwa M M Refaie
- Department of Pharmacology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt.
| | - Maram El-Hussieny
- Department of Pathology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt
| | - Asmaa M A Bayoumi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, 61511, El-Minia, Egypt
| | - Sayed Shehata
- Department of Cardiology, Faculty of Medicine, Minia University, 61511, El-Minia, Egypt
| |
Collapse
|
42
|
Zhu G, Pei L, Lin F, Yin H, Li X, He W, Liu N, Gou X. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. J Cell Physiol 2019; 234:23736-23749. [PMID: 31180587 DOI: 10.1002/jcp.28941] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 02/05/2023]
Abstract
Renal ischemia/reperfusion (I/R) injury is the main reason for acute kidney injury (AKI) and is closely related to high morbidity and mortality. In this study, we found that exosomes from human-bone-marrow-derived mesenchymal stem cells (hBMSC-Exos) play a protective role in hypoxia/reoxygenation (H/R) injury. hBMSC-Exos were enriched in miR-199a-3p, and hBMSC-Exo treatment increased the expression level of miR-199a-3p in renal cells. We further explored the function of miR-199a-3p on H/R injury. miR-199a-3p was knocked down in hBMSCs with a miR-199a-3p inhibitor. HK-2 cells cocultured with miR-199a-3p-knockdown hBMSCs were more susceptible to H/R injury and showed more apoptosis than those cocultured with hBMSCs or miR-199a-3p-overexpressing hBMSCs. Meanwhile, we found that HK-2 cells exposed to H/R treatment incubated with hBMSC-Exos decreased semaphorin 3A (Sema3A) and activated the protein kinase B (AKT) and extracellular-signal-regulated kinase (ERK) pathways. However, HK-2 cells cocultured with miR-199a-3p-knockdown hBMSCs restored Sema3A expression and blocked the activation of the AKT and ERK pathways. Moreover, knocking down Sema3A could reactivate the AKT and ERK pathways suppressed by a miR-199a-3p inhibitor. In vivo, we injected hBMSC-Exos into mice suffering from I/R injury; this treatment induced functional recovery and histologic protection and reduced cleaved caspase-3 and Sema3A expression levels, as shown by immunohistochemistry. On the whole, this study demonstrated an antiapoptotic effect of hBMSC-Exos, which protected against I/R injury, via delivering miR-199a-3p to renal cells, downregulating Sema3A expression and thereby activating the AKT and ERK pathways. These findings reveal a novel mechanism of AKI treated with hBMSC-Exos and provide a therapeutic method for kidney diseases.
Collapse
Affiliation(s)
- Gongmin Zhu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijiao Pei
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Lin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hubin Yin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyuan Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nian Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
43
|
Chen HP, Wen J, Tan SR, Kang LM, Zhu GC. MiR-199a-3p inhibition facilitates cardiomyocyte differentiation of embryonic stem cell through promotion of MEF2C. J Cell Physiol 2019; 234:23315-23325. [PMID: 31140610 DOI: 10.1002/jcp.28899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/26/2022]
Abstract
MicroRNAs (miRNAs) is a small molecule (19-25 nucleotide) noncoding RNA that inhibits the expression of target messenger RNA (mRNA) at the posttranscriptional level as an endogenous regulator. There is an increasing evidence that miR-199a-3p has a significant effect on the development of multiple tumors. However, the specific roles of miR-199a-3p in myocardial differentiation of embryonic stem cell still need to be investigated. Method of the hanging drop was used to build the model of cardiomyocyte differentiation of stem cell and beating rate of embryoid bodies (EBs) was calculated. The levels of intracellular MEF2C, a-MHC, GATA4, Nkx2.5, and cTnT mRNA were measured by real-time quantitative polymerase chain reaction, while the expressions of miR-199a-3p were detected simultaneously. Protein levels of MEF2C, a-MHC, GATA4, Nkx2.5, and cTnT were quantified by western blot analysis. Immunoreactivities of MEF2C and cTnT were analyzed by immunofluorescence. The interaction between miR-199a-3p and its predicted target (3'-untranslated region of MEF2C mRNA) was verified by luciferase assay. MiR-199a-3p levels increased during cardiogenesis. MiR-199a-3p inhibitor increased the beating rate of EBs and promoted expressions of cardiac-specific markers (GATA4, Nkx2.5, cTnT, and a-MHC). Notably, miR-199a-3p inhibition brought upregulation of MEF2C, which is the target of miR-199a-3p that we predicted and verified experimentally. In addition, MEF2C siRNA decreased miR-199a-3p inhibitor promoted EBs beating and attenuated miR-199a-3p inhibitor-induced cTnT and MEF2C expressions. The results above showed that MEF2C was involved in the process of promoting the differentiation of stem cells into cardiac myocytes by miR-199a-3p inhibitors.
Collapse
Affiliation(s)
- Hong-Ping Chen
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Tumor Pathogen's and Molecular Pathology, Nanchang University, Nanchang, China
| | - Jing Wen
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang, China
| | - Si-Rui Tan
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang, China
| | - Lu-Mei Kang
- Department of Animal Science, Medical College, Nanchang University, Nanchang, China
| | - Gao-Chun Zhu
- Department of Anatomy of the Human Body, Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
44
|
Chen Z, Su X, Shen Y, Jin Y, Luo T, Kim IM, Weintraub NL, Tang Y. MiR322 mediates cardioprotection against ischemia/reperfusion injury via FBXW7/notch pathway. J Mol Cell Cardiol 2019; 133:67-74. [PMID: 31150734 DOI: 10.1016/j.yjmcc.2019.05.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/04/2019] [Accepted: 05/25/2019] [Indexed: 12/26/2022]
Abstract
Myocardial ischemia/reperfusion (MI/R) causes loss of cardiomyocytes via oxidative stress-induced cardiomyocyte apoptosis. miR322, orthologous to human miR-424, was identified as an ischemia-induced angiogenic miRNA, but its cellular source and function in the setting of acute MI/R remains largely unknown. Using LacZ-tagged miR322 cluster reporter mice, we observed that vascular endothelial cells are the major cellular source of the miR322 cluster in adult hearts. Moreover, miR322 levels were significantly reduced in the heart at 24 h after MI/R injury. Intramyocardial injection of mimic-miR322 significantly diminished cardiac apoptosis (as determined by expression levels of active caspase 3 by Western blot analysis and immunostaining for TUNEL) and reduced infarct size by about 40%, in association with reduced FBXW7 and increased active Notch 1 levels in the ischemic hearts. FBXW7, which is an ubiquitin ligase that is crucial for activated Notch1 turnover, was identified as a direct target of miR322 via FBXW7 3'UTR reporter assay. Co-injection of FBXW7 plasmid with mimic-miR322 in ischemic hearts abolished the effect of mimic-miR322 to reduce apoptosis and infarct size in MI/R hearts. These data identify FBXW7 as a direct target of miR322 and suggest that miR322 could have potential therapeutic application for cardioprotection against ischemia/reperfusion-induced injury.
Collapse
Affiliation(s)
- Zixin Chen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Xuan Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yan Shen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yue Jin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Tong Luo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Il-Man Kim
- Department of Cellular & Integrative Physiology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
45
|
Huang X, Gao Y, Qin J, Lu S. miR-214 Down-Regulation Promoted Hypoxia/Reoxygenation-Induced Hepatocyte Apoptosis Through TRAF1/ASK1/JNK Pathway. Dig Dis Sci 2019; 64:1217-1225. [PMID: 30560327 DOI: 10.1007/s10620-018-5405-9] [Citation(s) in RCA: 8] [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: 10/09/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study investigated the role of miR-214 in the hepatocyte apoptosis induced by hypoxia/reoxygenation (H/R) injury. MATERIALS AND METHODS In vivo hepatic ischemia/reperfusion (HIR) injury, mice model and in vitro HR model were established. miR-214, TRAF1, ASK1, and JNK expression levels were detected by qRT-PCR and western blot. The apoptosis of mouse hepatocyte AML12 was detected by flow cytometry analysis. The interaction between miR-214 and TRAF1 was confirmed by dual-luciferase reporter gene assay. RESULTS Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were elevated in HIR injury mice compared with sham mice. miR-214 expression was down-regulated in liver tissues of HIR and H/R-induced hepatocytes, whereas TRAF1, ASK1, and JNK expressions were up-regulated in HIR and H/R groups. H/R stimulation promoted the apoptosis of hepatocytes, and miR-214 overexpression inhibited the apoptosis of hepatocytes. Besides, TRAF1 was a target of miR-214 and negatively regulated by miR-214. miR-214/TRAF1 pathway involved in the modulation of H/R-induced apoptosis of hepatocytes. In vivo study proved miR-214 reduced hepatic injury of HIR mice. CONCLUSION miR-214 overexpression reduces hepatocyte apoptosis after HIR injury through negatively regulating TRAF1/ASK1/JNK pathway.
Collapse
Affiliation(s)
- Xinli Huang
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yun Gao
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jianjie Qin
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Sen Lu
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
46
|
Bencivenga L, Liccardo D, Napolitano C, Visaggi L, Rengo G, Leosco D. β-Adrenergic Receptor Signaling and Heart Failure: From Bench to Bedside. Heart Fail Clin 2019; 15:409-419. [PMID: 31079699 DOI: 10.1016/j.hfc.2019.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite improvements in management and therapeutic approach in the last decades, heart failure is still associated with high mortality rates. The sustained enhancement in the sympathetic nervous system tone, observed in patients with heart failure, causes alteration in β-adrenergic receptor signaling and function. This latter phenomenon is the result of several heart failure-related molecular abnormalities involving adrenergic receptors, G-protein-coupled receptor kinases, and β-arrestins. This article summarizes novel encouraging preclinical strategies to reactivate β-adrenergic receptor signaling in heart failure, including pharmacologic and gene therapy approaches, and attempts to translate acquired notions into the clinical setting.
Collapse
Affiliation(s)
- Leonardo Bencivenga
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Daniela Liccardo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Carmen Napolitano
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Lucia Visaggi
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy.
| |
Collapse
|
47
|
Dehaini H, Awada H, El-Yazbi A, Zouein FA, Issa K, Eid AA, Ibrahim M, Badran A, Baydoun E, Pintus G, Eid AH. MicroRNAs as Potential Pharmaco-targets in Ischemia-Reperfusion Injury Compounded by Diabetes. Cells 2019; 8:152. [PMID: 30759843 PMCID: PMC6406262 DOI: 10.3390/cells8020152] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Ischemia-Reperfusion (I/R) injury is the tissue damage that results from re-oxygenation of ischemic tissues. There are many players that contribute to I/R injury. One of these factors is the family of microRNAs (miRNAs), which are currently being heavily studied. This review aims to critically summarize the latest papers that attributed roles of certain miRNAs in I/R injury, particularly in diabetic conditions and dissect their potential as novel pharmacologic targets in the treatment and management of diabetes. METHODS PubMed was searched for publications containing microRNA and I/R, in the absence or presence of diabetes. All papers that provided sufficient evidence linking miRNA with I/R, especially in the context of diabetes, were selected. Several miRNAs are found to be either pro-apoptotic, as in the case of miR-34a, miR-144, miR-155, and miR-200, or anti-apoptotic, as in the case of miR-210, miR-21, and miR-146a. Here, we further dissect the evidence that shows diverse cell-context dependent effects of these miRNAs, particularly in cardiomyocytes, endothelial, or leukocytes. We also provide insight into cases where the possibility of having two miRNAs working together to intensify a given response is noted. CONCLUSIONS This review arrives at the conclusion that the utilization of miRNAs as translational agents or pharmaco-targets in treating I/R injury in diabetic patients is promising and becoming increasingly clearer.
Collapse
Affiliation(s)
- Hassan Dehaini
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Hussein Awada
- Department of Biology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
- Department of Pharmacology and Toxicology, Alexandria University, Alexandria P.O. Box 21521, El-Mesallah, Egypt.
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Khodr Issa
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Maryam Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman P.O Box 961343 Amman, Jordan.
| | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar.
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar.
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar.
| |
Collapse
|
48
|
The essential role of tumor suppressor gene ING4 in various human cancers and non-neoplastic disorders. Biosci Rep 2019; 39:BSR20180773. [PMID: 30643005 PMCID: PMC6356015 DOI: 10.1042/bsr20180773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Inhibitor of growth 4 (ING4), a member of the ING family discovered in 2003, has been shown to act as a tumor suppressor and is frequently down-regulated in various human cancers. Numerous published in vivo and in vitro studies have shown that ING4 is responsible for important cancer hallmarks such as pathologic cell cycle arrest, apoptosis, autophagy, contact inhibition, and hypoxic adaptation, and also affects tumor angiogenesis, invasion, and metastasis. These characteristics are typically associated with regulation through chromatin acetylation by binding histone H3 trimethylated at lysine 4 (H3K4me3) and through transcriptional activity of transcription factor P53 and NF-κB. In addition, emerging evidence has indicated that abnormalities in ING4 expression and function play key roles in non-neoplastic disorders. Here, we provide an overview of ING4-modulated chromosome remodeling and transcriptional function, as well as the functional consequences of different genetic variants. We also present the current understanding concerning the role of ING4 in the development of neoplastic and non-neoplastic diseases. These studies offer inspiration for pursuing novel therapeutics for various cancers.
Collapse
|
49
|
Regulat-INGs in tumors and diseases: Focus on ncRNAs. Cancer Lett 2019; 447:66-74. [PMID: 30673590 DOI: 10.1016/j.canlet.2019.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/11/2022]
Abstract
ING family genes (Inhibitor of Growth) are tumor suppressor genes that play a vital role in cell homeostasis. It has been shown that their expression is lost or diminished in many cancers and other diseases. The main mechanisms by which they are regulated in oncogenesis have not yet been fully elucidated. The involvement of non-coding RNAs (ncRNAs) and in particular microRNAs (miRNAs) in post-transcriptional gene regulation is well established. miRNAs are short sequences (18-25 nucleotides) that can bind to the 3 'UTR sequence of the targeted messenger RNA (mRNA), leading to its degradation or translational repression. Interactions between the ING family and miRNAs have been described in some cancers but also in other diseases. The involvement of miRNAs in ING family regulation opens up new fields of investigation, particularly for targeted therapies. In this review, we will summarize the regulatory mechanisms at the RNA and protein level of the ING family and focus on the interactions with ncRNAs.
Collapse
|
50
|
Liang J, Zhu W, Zhang Z, Zhu J, Fu Y, Lin Q, Kuang S, Zhang M, Shan Z. [MicroRNA-199a-3p enhances expressions of fibrosis-associated genes through targeting Smad1 in mouse cardiac fibroblasts]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1203-1208. [PMID: 30377137 DOI: 10.3969/j.issn.1673-4254.2018.10.08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To investigate the role of miR-199a-3p in cardiac fibrosis and the potential target of miR-199a-3p. METHODS Cardiac fibroblasts were isolated from C57BL/6 mice and cultured. The miR-199a-3p mimic and Smad1 siRNA were transiently transfected into the cardiac fibroblasts via liposome. Dual luciferase reporter assay was performed to confirm the interaction between miR-199a-3p and the 3'-UTR of Smad1. The expressions of Smad1 and fibrosis-related genes at the mRNA and protein levels in the cells after miR-199a-3p mimic transfection were determined using RT-qPCR and Western blotting, respectively. The expressions of Smad1, Smad3 and fibrosis-related genes at the protein level in cells transfected with miR-199a-3p mimic and Smad1 siRNA were detected using Western blotting. RESULTS Over-expression of miR-199a-3p significantly increased the expression of cardiac fibrosis-related genes in cultured mouse cardiac fibroblasts. Dual luciferase reporter assay revealed the interaction of miR-199a-3p with the 3'-UTR of Smad1. The results of RT-qPCR and Western blotting confirmed that miR-199a-3p inhibited Smad1 expression at the post- transcriptional level. Transfection with miR-199a-3p mimic and siRNA-mediated Smad1 silencing consistently activated the Smad3 signaling pathway and enhanced the expressions of cardiac fibrosis-related genes in the cardiac fibroblasts. CONCLUSIONS As the target gene of miR-199a-3p, Smad1 mediates the pro-fibrotic effect of miR-199a-3p by activating the Smad3 signaling in cultured mouse cardiac fibroblasts.
Collapse
Affiliation(s)
- Jingnan Liang
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China
| | - Wensi Zhu
- Department of Pharmacy, Guangdong Women and Children's Hospital, Guangzhou 511400, China
| | - Zhuo Zhang
- Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jiening Zhu
- Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yongheng Fu
- Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qiuxiong Lin
- Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Sujuan Kuang
- Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Mengzhen Zhang
- Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhixin Shan
- School of Pharmacy, Southern Medical University, Guangzhou 510515, China.,Research Center of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|