1
|
Liu Z, Zhao Z, Xiao Z, Li M, Wang X, Huang Y, Li Y. Extracellular vesicles derived from bone marrow mesenchymal stem cells regulate SREBF2/HMGB1 axis by transporting miR-378a-3p to inhibit ferroptosis in intestinal ischemia-reperfusion injury. Cell Death Discov 2025; 11:223. [PMID: 40335466 PMCID: PMC12058992 DOI: 10.1038/s41420-025-02509-6] [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] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Abstract
Intestinal ischemia-reperfusion (II/R) injury represents a life-threatening and complex pathophysiological process that remains challenging to treat clinically, and emerging evidence suggests that ferroptosis plays an essential role in its pathogenesis. This study aimed to investigate whether extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) can mitigate II/R-induced ferroptosis in a murine model. Using a bioinformatics database, we initially identified genes with abnormal expression patterns in II/R injury. Then, we confirmed the association between II/R injury, ferroptosis, and the HMGB1/SREBF2 axis through in vivo and in vitro experiments. To determine the role of HMGB1 in hypoxia/reoxygenation (H/R)-induced ferroptosis in Caco-2 cells, we transfected cells with either sh-HMGB1 or control sh-NC constructs and developed an H/R model in vitro. Subsequently, we examined factors regulating HMGB1-mediated ferroptosis in Caco-2 cells and assessed the effect of BMSC-EVs on this process. To further explore the mechanism underlying the protective effects of BMSC-EVs in II/R injury, we screened for miRNAs with reduced expression during II/R and verified their involvement. Among these, miR-378a-3p was identified as a candidate for regulating ferroptosis. To confirm its functional role, we treated II/R mice with BMSC-EVs overexpressing miR-378a-3p and assessed the outcomes. Our findings revealed that HMGB1, which is a key regulatory factor of ferroptosis, was significantly upregulated during II/R injury, and its knockdown alleviated H/R-induced ferroptosis in Caco-2 cells. We also found that SREBF2 directly regulates HMGB1 expression to promote H/R-induced ferroptosis in vitro. Importantly, BMSC-EVs alleviated II/R injury by suppressing ferroptosis in Caco-2 cells, and mechanistically, miR-378a-3p, a miRNA derived from BMSC-EVs, inhibited II/R-induced ferroptosis by modulating the SREBF2/HMGB1 axis. In conclusion, BMSC-EVs may exert protective effects against II/R injury by delivering miR-378a-3p, which regulates the SREBF2/HMGB1 axis to suppress ferroptosis, providing important insights into the pathological mechanisms underlying II/R injury and potential therapeutic strategies for its management.
Collapse
Affiliation(s)
- Zan Liu
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, College of Pharmacy, Changsha Medical University, Changsha, PR China
| | - Zitong Zhao
- Center of Reproductive Medicine, Changsha Hospital for Maternal and Child Health Care of Hunan Normal University, Changsha, PR China
| | - Zhenghui Xiao
- Emergency center of Hunan Children's Hospital, Changsha, Hunan, PR China
| | - Ming Li
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China
| | - Xiyang Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yan Huang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, PR China.
- Hunan Provincial Key Laboratory of Neurorestoration, Changsha, Hunan, PR China.
| | - Yong Li
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China.
| |
Collapse
|
2
|
Liu L, Zhang H, Zheng Y, Gao T, Zheng C, Zhang K, Hou L, Liu L. High-dimensional mediation analysis for longitudinal mediators and survival outcomes. Brief Bioinform 2025; 26:bbaf206. [PMID: 40350699 PMCID: PMC12066418 DOI: 10.1093/bib/bbaf206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/17/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Mediation analysis with high-dimensional mediators is crucial for identifying epigenetic pathways linking environmental exposures to health outcomes. However, high-dimensional mediation analysis methods for longitudinal mediators and a survival outcome remain underdeveloped. This study fills that gap by introducing a method that captures mediation effects over time using multivariate, longitudinally measured time-varying mediators. Our approach uses a longitudinal mixed effects model to examine the relationship between the exposure and the mediating process. We connect the mediating process to the survival outcome using a Cox proportional hazards model with time-varying mediators. To handle high-dimensional data, we first employ a mediation-based sure independence screening method for dimension reduction. A Lasso inference procedure is further utilized to identify significant time-varying mediators. We adopt a joint significance test to accurately control the family wise error rate in testing high-dimensional mediation hypotheses. Simulation studies and an analysis of the Coronary Artery Risk Development in Young Adults Study demonstrate the utility and validity of our method.
Collapse
Affiliation(s)
- Lili Liu
- Center for Biostatistics and Data Science, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, MO 63110, United States
| | - Haixiang Zhang
- Center for Applied Mathematics, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Drive, Chicago, IL 60611, United States
| | - Tao Gao
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Drive, Chicago, IL 60611, United States
| | - Cheng Zheng
- Department of Biostatistics, University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE 68198, United States
| | - Kai Zhang
- Department of Population and Community Health, College of Public Health, The University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107, United States
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Drive, Chicago, IL 60611, United States
| | - Lei Liu
- Center for Biostatistics and Data Science, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, MO 63110, United States
| |
Collapse
|
3
|
Liu W, Liu J, Wang G, Cheng W, Gong H, Song Y, Song M, Zhuge Y, Li Y, Liu J. Down-regulation of histone deacetylase 2 attenuates ventricular arrhythmias in a mouse model of cardiac hypertrophy through up-regulation of Kv channel-interacting protein 2 expression. Cardiovasc Res 2025; 121:424-439. [PMID: 39870585 DOI: 10.1093/cvr/cvaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 07/24/2024] [Accepted: 10/17/2024] [Indexed: 01/29/2025] Open
Abstract
AIMS Decrease in repolarizing K+ currents, particularly the fast component of transient outward K+ current (Ito,f), prolongs action potential duration (APD) and predisposes the heart to ventricular arrhythmia during cardiac hypertrophy. Histone deacetylases (HDACs) have been suggested to participate in the development of cardiac hypertrophy, and Class I HDAC inhibition has been found to attenuate pathological remodelling. This study investigated the potential therapeutic effects of HDAC2 on ventricular arrhythmia in pressure overload-induced cardiac hypertrophy. METHODS AND RESULTS An in vivo cardiac hypertrophic model was produced by performing transverse aortic constriction (TAC) surgery and an in vitro cardiomyocyte hypertrophy model by stimulating neonatal rat ventricular myocytes (NRVMs) with phenylephrine (PE). HDAC2 expression was up-regulated in TAC mouse hearts and in PE-stimulated cardiomyocytes. Susceptibility to ventricular arrhythmia was increased in TAC mice, while Ito,f was decreased and APD was prolonged in TAC cardiomyocytes. Heart-specific knockdown (HKD) of HDAC2 by RNA interference increased Ito,f, shortened APD, and decreased susceptibility to ventricular arrhythmia. Concomitantly, HKD increased the expression of the obligatory β sub-unit of Ito,f, Kv channel-interacting protein 2 (KChIP2), which is down-regulated in hypertrophic hearts. The effects of HKD on KChIP2 expression, Ito,f and APD were also observed in PE-stimulated cardiomyocytes. Mechanistically, HKD increased H3K4me3 abundance and H3K4me3 enrichment at the Kcnip2 promoter in cardiomyocytes. HKD also decreased the expression of KDM5, the H3K4me3 demethylase, which resulted in H3K4me3 up-regulation. While investigating the regulatory mechanisms underlying the effect of HDAC2 on KDM5 stability, we identified CNOT4 as the active KDM5 ubiquitinase in cardiomyocytes. HKD increased CNOT4 expression and CNOT4-KDM5 interactions and thus enhanced the polyubiquitinated degradation of KDM5. CONCLUSION HDAC2 inhibition serves as a novel therapeutic strategy for preventing cardiac hypertrophy-associated electrophysiological remodelling. Furthermore, we identified a novel signalling pathway of CNOT4-mediated KDM5 degradation contributing to the up-regulation of H3K4me3-mediated KChIP2 expression in response to HDAC2 inhibition.
Collapse
MESH Headings
- Animals
- Histone Deacetylase 2/genetics
- Histone Deacetylase 2/metabolism
- Disease Models, Animal
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Down-Regulation
- Up-Regulation
- Male
- Arrhythmias, Cardiac/enzymology
- Arrhythmias, Cardiac/prevention & control
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/etiology
- Action Potentials
- Kv Channel-Interacting Proteins/metabolism
- Kv Channel-Interacting Proteins/genetics
- Cardiomegaly/enzymology
- Cardiomegaly/physiopathology
- Cardiomegaly/genetics
- Cardiomegaly/complications
- Mice, Inbred C57BL
- Cells, Cultured
- Rats
- Mice
- Signal Transduction
- Ventricular Remodeling
Collapse
Affiliation(s)
- Wenjuan Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jianping Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Gang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Wanwen Cheng
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Haochen Gong
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Yujuan Song
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Ming Song
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Yixin Zhuge
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Ying Li
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jie Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
4
|
Shi L, Wei J, Shen Y, Zhang X, Li L, Deng Y. Prognostic significance of miR-378 in cancers: a meta-analysis. BMC Cancer 2025; 25:428. [PMID: 40065304 PMCID: PMC11892288 DOI: 10.1186/s12885-025-13619-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The prognostic significance of miR-378 in cancers remains controversial. We carried out the meta-analysis to clarify the issue. METHODS Related researches were obtained from PubMed, Web of Science and EMBASE. The search was conducted up until 10 September 2023. Hazard ratios (HRs) or odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to access the correlation between miR-378 and survival outcomes and clinicopathological features. RESULTS 16 articles were included in the meta-analysis. The pooled analysis showed that low miR-378 expression predicted poor overall survival (OS) (HR: 1.73, 95% CI: 1.09-2.74). No significant difference was discovered between low miR-378 expression and disease-free survival/ recurrence-free survival (DFS/RFS) (HR: 1.43; 95% CI: 0.79-2.59). Subgroup analysis revealed that the low miR-378 expression had better predictive value for CRC (HR: 2.97, 95% CI: 1.27-6.96), GC (HR: 2.12, 95% CI: 1.23-3.67) and glioma (HR: 3.87, 95% CI: 1.52-9.81). In addition, low miR-378 expression had positive correlation with lymph node metastasis (yes vs. no)(OR: 2.42, 95% CI: 1.55-2.87) and distant metastasis (yes vs. no) (OR: 1.26, 95% CI: 1.04-2.35). CONCLUSION MiR-378 could be an efficient prognostic biomarker in cancers.
Collapse
Affiliation(s)
- Lei Shi
- Department of general surgery, the Second People's Hospital of Lanzhou City, Lanzhou, 730046, China
| | - Jie Wei
- Department of health management, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, 730050, China
| | - Yankun Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Xiaoqiang Zhang
- Department of general surgery, the Second People's Hospital of Lanzhou City, Lanzhou, 730046, China
| | - Lixin Li
- Department of general surgery, the Second People's Hospital of Lanzhou City, Lanzhou, 730046, China
| | - Ying Deng
- Department of general surgery, the Second People's Hospital of Lanzhou City, Lanzhou, 730046, China.
| |
Collapse
|
5
|
Hu H, Wang X, Yu H, Wang Z. Extracellular vesicular microRNAs and cardiac hypertrophy. Front Endocrinol (Lausanne) 2025; 15:1444940. [PMID: 39850481 PMCID: PMC11753959 DOI: 10.3389/fendo.2024.1444940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Cardiac hypertrophy is an adaptive response to pressure or volume overload such as hypertension and ischemic heart diseases. Sustained cardiac hypertrophy eventually leads to heart failure. The pathophysiological alterations of hypertrophy are complex, involving both cellular and molecular systems. Understanding the molecular events that inhibit or repress cardiac hypertrophy may help identify novel therapeutic strategies. Increasing evidence has indicated that extracellular vesicle (EV)-derived microRNAs (miRNAs) play a significant role in the development and progression of cardiac hypertrophy. In this review, we briefly review recent advancements in EV research, especially on biogenesis, cargoes and its role in cardiac hypertrophy. We then describe the latest findings regarding EV-derived miRNAs, highlighting their functions and regulatory mechanisms in cardiac hypertrophy. Finally, the potential role of EV-derived miRNAs as targets in the diagnosis and treatment of cardiac hypertrophy will be discussed.
Collapse
Affiliation(s)
- Hai Hu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xiulian Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
6
|
Schoettler FI, Fatehi Hassanabad A, Jadli AS, Patel VB, Fedak PWM. Exploring the role of pericardial miRNAs and exosomes in modulating cardiac fibrosis. Cardiovasc Pathol 2024; 73:107671. [PMID: 38906439 DOI: 10.1016/j.carpath.2024.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/26/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
The potential of the pericardial space as a therapeutic delivery tool for cardiac fibrosis and heart failure (HF) treatment has yet to be elucidated. Recently, miRNAs and exosomes have been discovered to be present in human pericardial fluid (PF). Novel studies have shown characteristic human PF miRNA compositions associated with cardiac diseases and higher miRNA expressions in PF compared to peripheral blood. Five key studies found differentially expressed miRNAs in HF, angina pectoris, aortic stenosis, ventricular tachycardia, and congenital heart diseases with either atrial fibrillation or sinus rhythm. As miRNA-based therapeutics for cardiac fibrosis and HF showed promising results in several in vivo studies for multiple miRNAs, we hypothesize a potential role of miRNA-based therapeutics delivered through the pericardial cavity. This is underlined by the favorable results of the first phase 1b clinical trial in this emerging field. Presenting the first human miRNA antisense drug trial, inhibition of miR-132 by intravenous administration of a novel antisense oligonucleotide, CDR132L, established efficacy in reducing miR-132 in plasma samples in a dose-dependent manner. We screened the literature, provided an overview of the miRNAs and exosomes present in PF, and drew a connection to those miRNAs previously elucidated in cardiac fibrosis and HF. Further, we speculate about clinical implications and potential delivery methods.
Collapse
Affiliation(s)
- Friederike I Schoettler
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ali Fatehi Hassanabad
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cardiac Sciences, Section of Cardiac Surgery, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anshul S Jadli
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul W M Fedak
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cardiac Sciences, Section of Cardiac Surgery, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
7
|
Fatehi Hassanabad A, Zarzycki AN, Patel VB, Fedak PWM. Current concepts in the epigenetic regulation of cardiac fibrosis. Cardiovasc Pathol 2024; 73:107673. [PMID: 38996851 DOI: 10.1016/j.carpath.2024.107673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
Cardiac fibrosis is a significant driver of congestive heart failure, a syndrome that continues to affect a growing patient population globally. Cardiac fibrosis results from a constellation of complex processes at the transcription, receptor, and signaling axes levels. Various mediators and signaling cascades, such as the transformation growth factor-beta pathway, have been implicated in the pathophysiology of cardiac tissue fibrosis. Our understanding of these markers and pathways has improved in recent years as more advanced technologies and assays have been developed, allowing for better delineation of the crosstalk between specific factors. There is mounting evidence suggesting that epigenetic modulation plays a pivotal role in the progression of cardiac fibrosis. Transcriptional regulation of key pro- and antifibrotic pathways can accentuate or blunt the rate and extent of fibrosis at the tissue level. Exosomes, micro-RNAs, and long noncoding RNAs all belong to factors that can impact the epigenetic signature in cardiac fibrosis. Herein, we comprehensively review the latest literature about exosomes, their contents, and cardiac fibrosis. In doing so, we highlight the specific transcriptional factors with pro- or antifibrotic properties. We also assimilate the data supporting these mediators' potential utility as diagnostic or prognostic biomarkers. Finally, we offer insight into where further work can be done to fill existing gaps to translate preclinical findings better and improve clinical outcomes.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
8
|
He C, Hu C, He WZ, Sun YC, Jiang Y, Liu L, Hou J, Chen KX, Jiao YR, Huang M, Huang M, Yang M, Lu Q, Wei J, Zeng C, Lei GH, Li CJ. Macrophage-derived extracellular vesicles regulate skeletal stem/progenitor Cell lineage fate and bone deterioration in obesity. Bioact Mater 2024; 36:508-523. [PMID: 39072285 PMCID: PMC11282946 DOI: 10.1016/j.bioactmat.2024.06.035] [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: 03/19/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Obesity-induced chronic inflammation exacerbates multiple types of tissue/organ deterioration and stem cell dysfunction; however, the effects on skeletal tissue and the underlying mechanisms are still unclear. Here, we show that obesity triggers changes in the microRNA profile of macrophage-secreted extracellular vesicles, leading to a switch in skeletal stem/progenitor cell (SSPC) differentiation between osteoblasts and adipocytes and bone deterioration. Bone marrow macrophage (BMM)-secreted extracellular vesicles (BMM-EVs) from obese mice induced bone deterioration (decreased bone volume, bone microstructural deterioration, and increased adipocyte numbers) when administered to lean mice. Conversely, BMM-EVs from lean mice rejuvenated bone deterioration in obese recipients. We further screened the differentially expressed microRNAs in obese BMM-EVs and found that among the candidates, miR-140 (with the function of promoting adipogenesis) and miR-378a (with the function of enhancing osteogenesis) coordinately determine SSPC fate of osteogenic and adipogenic differentiation by targeting the Pparα-Abca1 axis. BMM miR-140 conditional knockout mice showed resistance to obesity-induced bone deterioration, while miR-140 overexpression in SSPCs led to low bone mass and marrow adiposity in lean mice. BMM miR-378a conditional depletion in mice led to obesity-like bone deterioration. More importantly, we used an SSPC-specific targeting aptamer to precisely deliver miR-378a-3p-overloaded BMM-EVs to SSPCs via an aptamer-engineered extracellular vesicle delivery system, and this approach rescued bone deterioration in obese mice. Thus, our study reveals the critical role of BMMs in mediating obesity-induced bone deterioration by transporting selective extracellular-vesicle microRNAs into SSPCs and controlling SSPC fate.
Collapse
Affiliation(s)
- Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Chen Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Wen-Zhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu-Chen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yangzi Jiang
- School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), The Chinese University of Hong Kong, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Kai-Xuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu-Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jie Wei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Guang-Hua Lei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Zhang L, Xie F, Zhang F, Lu B. The potential roles of exosomes in pathological cardiomyocyte hypertrophy mechanisms and therapy: A review. Medicine (Baltimore) 2024; 103:e37994. [PMID: 38669371 PMCID: PMC11049793 DOI: 10.1097/md.0000000000037994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Pathological cardiac hypertrophy, characterized by the enlargement of cardiac muscle cells, leads to serious cardiac conditions and stands as a major global health issue. Exosomes, comprising small lipid bilayer vesicles, are produced by various cell types and found in numerous bodily fluids. They play a pivotal role in intercellular communication by transferring bioactive cargos to recipient cells or activating signaling pathways in target cells. Exosomes from cardiomyocytes, endothelial cells, fibroblasts, and stem cells are key in regulating processes like cardiac hypertrophy, cardiomyocyte survival, apoptosis, fibrosis, and angiogenesis within the context of cardiovascular diseases. This review delves into exosomes' roles in pathological cardiac hypertrophy, first elucidating their impact on cell communication and signaling pathways. It then advances to discuss how exosomes affect key hypertrophic processes, including metabolism, fibrosis, oxidative stress, and angiogenesis. The review culminates by evaluating the potential of exosomes as biomarkers and their significance in targeted therapeutic strategies, thus emphasizing their critical role in the pathophysiology and management of cardiac hypertrophy.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Xie
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fengmei Zhang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Beiyao Lu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Nappi F. Non-Coding RNA-Targeted Therapy: A State-of-the-Art Review. Int J Mol Sci 2024; 25:3630. [PMID: 38612441 PMCID: PMC11011542 DOI: 10.3390/ijms25073630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The use of non-coding RNAs (ncRNAs) as drug targets is being researched due to their discovery and their role in disease. Targeting ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), is an attractive approach for treating various diseases, such as cardiovascular disease and cancer. This seminar discusses the current status of ncRNAs as therapeutic targets in different pathological conditions. Regarding miRNA-based drugs, this approach has made significant progress in preclinical and clinical testing for cardiovascular diseases, where the limitations of conventional pharmacotherapy are evident. The challenges of miRNA-based drugs, including specificity, delivery, and tolerability, will be discussed. New approaches to improve their success will be explored. Furthermore, it extensively discusses the potential development of targeted therapies for cardiovascular disease. Finally, this document reports on the recent advances in identifying and characterizing microRNAs, manipulating them, and translating them into clinical applications. It also addresses the challenges and perspectives towards clinical application.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
11
|
Gallo A, Agnese V, Sciacca S, Scardulla C, Cipriani M, Pilato M, Oh JK, Pasta S, Maalouf J, Conaldi PG, Bellavia D. MicroRNA-30d and -483-3p for bi-ventricular remodelling and miR-126-3p for pulmonary hypertension in advanced heart failure. ESC Heart Fail 2024; 11:155-166. [PMID: 37864482 PMCID: PMC10804158 DOI: 10.1002/ehf2.14546] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/23/2023] Open
Abstract
AIMS MicroRNAs play a role in pathogenic mechanisms leading to heart failure. We measured a panel of 754 miRNAs in the myocardial tissue and in the serum of patients with heart failure with reduced ejection fraction due to dilatative idiopathic cardiomyopathy (DCM, N = 10) or ischaemic cardiomyopathy (N = 3), referred to left ventricular assist device implant. We aim to identify circulating miRNAs with high tissue co-expression, significantly associated to echocardiographic and haemodynamic measures. METHODS AND RESULTS We have measured a panel of 754 miRNAs in the myocardial tissue [left ventricular (LV) apex] and in the serum obtained at the same time in a well selected study population of end-stage heart failure with reduced ejection fraction due to either DCM or ischaemic cardiomyopathy, referred to continuous flow left ventricular assist device implant. We observed moderate agreement for miR-30d, miR-126-3p, and miR-483-3p. MiR-30d was correlated to LV systolic as well as diastolic volumes (r = 0.78, P = 0.001 and r = 0.80, P = 0.005, respectively), while miR-126-3p was associated to mPAP and PCWP (r = -0.79, P = 0.007 and r = -0.80, P = 0.005, respectively). Finally, serum miR-483-3p had an association with right ventricular end diastolic diameter (r = -0.73, P = 0.02) and central venous pressure (CVP) (r - 0.68 p 0.03). CONCLUSIONS In patients with DCM, few miRNAs are co-expressed in serum and tissue: They are related to LV remodelling (miR-30d), post-capillary pulmonary artery pressure (miR-126-3p), and right ventricular remodelling/filling pressures (miR-483-3p). Further studies are needed to confirm their role in diagnosis, prognosis or as therapeutic targets in heart failure with reduced ejection fraction.
Collapse
Affiliation(s)
| | | | - Sergio Sciacca
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic TransplantationIRCCS‐ISMETTPalermoItaly
| | - Cesare Scardulla
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic TransplantationIRCCS‐ISMETTPalermoItaly
| | - Manlio Cipriani
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic TransplantationIRCCS‐ISMETTPalermoItaly
| | - Michele Pilato
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic TransplantationIRCCS‐ISMETTPalermoItaly
| | - Jae K. Oh
- Department of Internal Medicine, Division of Cardiovascular DiseasesMayo Clinic and FoundationRochesterMNUSA
| | | | - Joseph Maalouf
- Department of Internal Medicine, Division of Cardiovascular DiseasesMayo Clinic and FoundationRochesterMNUSA
| | | | | |
Collapse
|
12
|
Wang G, Feng L, Liu C, Han Z, Chen X. MiR-378 Inhibits Angiotensin II-Induced Cardiomyocyte Hypertrophy by Targeting AKT2. Int Heart J 2024; 65:528-536. [PMID: 38825497 DOI: 10.1536/ihj.23-485] [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: 06/04/2024]
Abstract
Cardiomyocyte hypertrophy plays a crucial role in heart failure development, potentially leading to sudden cardiac arrest and death. Previous studies suggest that micro-ribonucleic acids (miRNAs) show promise for the early diagnosis and treatment of cardiomyocyte hypertrophy.To investigate the miR-378 expression in the cardiomyocyte hypertrophy model, reverse transcription-polymerase chain reaction (RT-qPCR), Western blot, and immunofluorescence tests were conducted in angiotensin II (Ang II)-induced H9c2 cells and Ang II-induced mouse model of cardiomyocyte hypertrophy. The functional interaction between miR-378 and AKT2 was studied by dual-luciferase reporter, RNA pull-down, Western blot, and RT-qPCR assays.The results of RT-qPCR analysis showed the downregulated expression of miR-378 in both the cell and animal models of cardiomyocyte hypertrophy. It was observed that the introduction of the miR-378 mimic inhibited the hypertrophy of cardiomyocytes induced by Ang II. Furthermore, the co-transfection of AKT2 expression vector partially mitigated the negative impact of miR-378 overexpression on Ang II-induced cardiomyocytes. Molecular investigations provided evidence that miR-378 negatively regulated AKT2 expression by interacting with the 3' untranslated region (UTR) of AKT2 mRNA.Decreased miR-378 expression and AKT2 activation are linked to Ang II-induced cardiomyocyte hypertrophy. Targeting miR-378/AKT2 axis offers therapeutic opportunity to alleviate cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Guili Wang
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital
| | - Linlin Feng
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital
| | - Chunxiang Liu
- Department of Ultrasound, Beijing Xiaotangshan Hospital
| | - Zongqiang Han
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital
| | - Xia Chen
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital
| |
Collapse
|
13
|
Yamaguchi A, Maeshige N, Noguchi H, Yan J, Ma X, Uemura M, Su D, Kondo H, Sarosiek K, Fujino H. Pulsed ultrasound promotes secretion of anti-inflammatory extracellular vesicles from skeletal myotubes via elevation of intracellular calcium level. eLife 2023; 12:RP89512. [PMID: 38054662 PMCID: PMC10699803 DOI: 10.7554/elife.89512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
The regulation of inflammatory responses is an important intervention in biological function and macrophages play an essential role during inflammation. Skeletal muscle is the largest organ in the human body and releases various factors which mediate anti-inflammatory/immune modulatory effects. Recently, the roles of extracellular vesicles (EVs) from a large variety of cells are reported. In particular, EVs released from skeletal muscle are attracting attention due to their therapeutic effects on dysfunctional organs and tissues. Also, ultrasound (US) promotes release of EVs from skeletal muscle. In this study, we investigated the output parameters and mechanisms of US-induced EV release enhancement and the potential of US-treated skeletal muscle-derived EVs in the regulation of inflammatory responses in macrophages. High-intensity US (3.0 W/cm2) irradiation increased EV secretion from C2C12 murine muscle cells via elevating intracellular Ca2+ level without negative effects. Moreover, US-induced EVs suppressed expression levels of pro-inflammatory factors in macrophages. miRNA sequencing analysis revealed that miR-206-3p and miR-378a-3p were especially abundant in skeletal myotube-derived EVs. In this study we demonstrated that high-intensity US promotes the release of anti-inflammatory EVs from skeletal myotubes and exert anti-inflammatory effects on macrophages.
Collapse
Affiliation(s)
- Atomu Yamaguchi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hikari Noguchi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Jiawei Yan
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Xiaoqi Ma
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Mikiko Uemura
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Hiroyo Kondo
- Department of Health and Nutrition , Shubun University, Ichinomiya, Japan
| | - Kristopher Sarosiek
- John B. Little Center for Radiation Sciences, Harvard University T.H. Chan School of Public Health, Boston, United States
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
14
|
Florczyk-Soluch U, Polak K, Sabo R, Martyniak A, Stępniewski J, Dulak J. Compromised diabetic heart function is not affected by miR-378a upregulation upon hyperglycemia. Pharmacol Rep 2023; 75:1556-1570. [PMID: 37851320 PMCID: PMC10661816 DOI: 10.1007/s43440-023-00535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Cardiac-abundant microRNA-378a (miR-378a) is associated with postnatal repression of insulin-like growth factor 1 receptor (IGF-1R) controlling physiological hypertrophy and survival pathways. IGF-1/IGF-1R axis has been proposed as a therapeutic candidate against the pathophysiological progress of diabetic cardiomyopathy (DCM). We ask whether hyperglycemia-driven changes in miR-378a expression could mediate DCM progression. METHODS Diabetes mellitus was induced by streptozotocin (STZ) (55 mg/kg i.p. for 5 days) in male C57BL/6 wild type (miR-378a+/+) and miR-378a knockout (miR-378a-/-) mice. As a parallel human model, we harnessed human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM miR378a+/+ vs. hiPSC-CM miR378a-/-) subjected to high glucose (HG) treatment. RESULTS We reported miR-378a upregulation in cardiac diabetic milieu arising upon STZ administration to wild-type mice and in HG-treated hiPSC-CMs. Pro-hypertrophic IGF-1R/ERK1/2 pathway and hypertrophic marker expression were activated in miR-378a deficiency and upon STZ/HG treatment of miR-378a+/+ specimens in vivo and in vitro suggesting miR-378a-independent hyperglycemia-promoted hypertrophy. A synergistic upregulation of IGF-1R signaling in diabetic conditions was detected in miR-378a-/- hiPSC-CMs, but not in miR-378a-/- hearts that showed attenuation of this pathway, pointing to the involvement of compensatory mechanisms in the absence of miR-378a. Although STZ administration did not cause pro-inflammatory or pro-fibrotic effects that were detected in miR-378a-/- mice, the compromised diabetic heart function observed in vivo by high-resolution ultrasound imaging upon STZ treatment was not affected by miR-378a presence. CONCLUSIONS Overall, data underline the role of miR-378a in maintaining basal cardiac structural integrity while pointing to miR-378a-independent hyperglycemia-driven cardiac hypertrophy and associated dysfunction.
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Reece Sabo
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| |
Collapse
|
15
|
Poller W, Sahoo S, Hajjar R, Landmesser U, Krichevsky AM. Exploration of the Noncoding Genome for Human-Specific Therapeutic Targets-Recent Insights at Molecular and Cellular Level. Cells 2023; 12:2660. [PMID: 37998395 PMCID: PMC10670380 DOI: 10.3390/cells12222660] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
While it is well known that 98-99% of the human genome does not encode proteins, but are nevertheless transcriptionally active and give rise to a broad spectrum of noncoding RNAs [ncRNAs] with complex regulatory and structural functions, specific functions have so far been assigned to only a tiny fraction of all known transcripts. On the other hand, the striking observation of an overwhelmingly growing fraction of ncRNAs, in contrast to an only modest increase in the number of protein-coding genes, during evolution from simple organisms to humans, strongly suggests critical but so far essentially unexplored roles of the noncoding genome for human health and disease pathogenesis. Research into the vast realm of the noncoding genome during the past decades thus lead to a profoundly enhanced appreciation of the multi-level complexity of the human genome. Here, we address a few of the many huge remaining knowledge gaps and consider some newly emerging questions and concepts of research. We attempt to provide an up-to-date assessment of recent insights obtained by molecular and cell biological methods, and by the application of systems biology approaches. Specifically, we discuss current data regarding two topics of high current interest: (1) By which mechanisms could evolutionary recent ncRNAs with critical regulatory functions in a broad spectrum of cell types (neural, immune, cardiovascular) constitute novel therapeutic targets in human diseases? (2) Since noncoding genome evolution is causally linked to brain evolution, and given the profound interactions between brain and immune system, could human-specific brain-expressed ncRNAs play a direct or indirect (immune-mediated) role in human diseases? Synergistic with remarkable recent progress regarding delivery, efficacy, and safety of nucleic acid-based therapies, the ongoing large-scale exploration of the noncoding genome for human-specific therapeutic targets is encouraging to proceed with the development and clinical evaluation of novel therapeutic pathways suggested by these research fields.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department for Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum Charité (DHZC), Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany;
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 10785 Berlin, Germany
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA;
| | - Roger Hajjar
- Gene & Cell Therapy Institute, Mass General Brigham, 65 Landsdowne St, Suite 143, Cambridge, MA 02139, USA;
| | - Ulf Landmesser
- Department for Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum Charité (DHZC), Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany;
- German Center for Cardiovascular Research (DZHK), Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Anna M. Krichevsky
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
16
|
Asjad E, Dobrzynski H. MicroRNAs: Midfielders of Cardiac Health, Disease and Treatment. Int J Mol Sci 2023; 24:16207. [PMID: 38003397 PMCID: PMC10671258 DOI: 10.3390/ijms242216207] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that play a role in post-transcriptional gene regulation. It is generally accepted that their main mechanism of action is the negative regulation of gene expression, through binding to specific regions in messenger RNA (mRNA) and repressing protein translation. By interrupting protein synthesis, miRNAs can effectively turn genes off and influence many basic processes in the body, such as developmental and apoptotic behaviours of cells and cardiac organogenesis. Their importance is highlighted by inhibiting or overexpressing certain miRNAs, which will be discussed in the context of coronary artery disease, atrial fibrillation, bradycardia, and heart failure. Dysregulated levels of miRNAs in the body can exacerbate or alleviate existing disease, and their omnipresence in the body makes them reliable as quantifiable markers of disease. This review aims to provide a summary of miRNAs as biomarkers and their interactions with targets that affect cardiac health, and intersperse it with current therapeutic knowledge. It intends to succinctly inform on these topics and guide readers toward more comprehensive works if they wish to explore further through a wide-ranging citation list.
Collapse
Affiliation(s)
- Emman Asjad
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| | - Halina Dobrzynski
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
- Department of Anatomy, Jagiellonian University Medical College, 31-034 Krakow, Poland
| |
Collapse
|
17
|
Nappi F, Avtaar Singh SS, Jitendra V, Alzamil A, Schoell T. The Roles of microRNAs in the Cardiovascular System. Int J Mol Sci 2023; 24:14277. [PMID: 37762578 PMCID: PMC10531750 DOI: 10.3390/ijms241814277] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The discovery of miRNAs and their role in disease represent a significant breakthrough that has stimulated and propelled research on miRNAs as targets for diagnosis and therapy. Cardiovascular disease is an area where the restrictions of early diagnosis and conventional pharmacotherapy are evident and deserve attention. Therefore, miRNA-based drugs have significant potential for development. Research and its application can make considerable progress, as seen in preclinical and clinical trials. The use of miRNAs is still experimental but has a promising role in diagnosing and predicting a variety of acute coronary syndrome presentations. Its use, either alone or in combination with currently available biomarkers, might be adopted soon, particularly if there is diagnostic ambiguity. In this review, we examine the current understanding of miRNAs as possible targets for diagnosis and treatment in the cardiovascular system. We report on recent advances in recognising and characterising miRNAs with a focus on clinical translation. The latest challenges and perspectives towards clinical application are discussed.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (T.S.)
| | | | - Vikram Jitendra
- Department of Cardiothoracic Surgery, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK;
| | - Almothana Alzamil
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (T.S.)
| | - Thibaut Schoell
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (T.S.)
| |
Collapse
|
18
|
Wang H, Shi J, Wang J, Hu Y. MicroRNA‑378: An important player in cardiovascular diseases (Review). Mol Med Rep 2023; 28:172. [PMID: 37503766 PMCID: PMC10436248 DOI: 10.3892/mmr.2023.13059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/31/2023] [Indexed: 07/29/2023] Open
Abstract
Cardiovascular disease (CVD) is a common chronic clinical condition and is the main cause of death in humans worldwide. Understanding the genetic and molecular mechanisms involved in the development of CVD is essential to develop effective prevention strategies and therapeutic measures. An increasing number of CVD‑related genetic studies have been conducted, including those on the potential roles of microRNAs (miRs). These studies have demonstrated that miR‑378 is involved in the pathological processes of CVD, including those of myocardial infarction, heart failure and coronary heart disease. Despite the potential importance of miR‑378 CVD, a comprehensive summary of the related literature is lacking. Thus, the present review aimed to summarize the findings of previous studies on the roles and mechanisms of miR‑378 in a variety of CVDs and provide an up‑to date basis for further r research targeting the prevention and treatment of CVDs.
Collapse
Affiliation(s)
- Huan Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Jingjing Shi
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Jiuchong Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| |
Collapse
|
19
|
Li K, Ma L, Lu Z, Yan L, Chen W, Wang B, Xu H, Asemi Z. Apoptosis and heart failure: The role of non-coding RNAs and exosomal non-coding RNAs. Pathol Res Pract 2023; 248:154669. [PMID: 37422971 DOI: 10.1016/j.prp.2023.154669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/01/2023] [Accepted: 07/02/2023] [Indexed: 07/11/2023]
Abstract
Heart failure is a condition that affects the cardio vascular system and occurs if the heart cannot adequately pump the oxygen and blood to the body. Myocardial infarction, reperfusion injury, and this disease is the only a few examples of the numerous cardiovascular illnesses that are impacted by the closely controlled cell deletion process known as apoptosis. Attention has been paid to the creation of alternative diagnostic and treatment modalities for the condition. Recent evidences have shown that some non-coding RNAs (ncRNAs) influence the stability of proteins, control of transcription factors, and HF apoptosis through a variety of methods. Exosomes make a significant paracrine contribution to the regulation of illnesses as well as to the communication between nearby and distant organs. However, it has not yet been determined whether exosomes regulate the cardiomyocyte-tumor cell interaction in ischemia HF to limit the vulnerability of malignancy to ferroptosis. Here, we list the numerous ncRNAs in HF that are connected to apoptosis. In addition, we emphasize the significance of exosomal ncRNAs in the HF.
Collapse
Affiliation(s)
- Ketao Li
- Department of cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310022, China
| | - Liping Ma
- Department of cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310022, China
| | - Zhiwei Lu
- Hangzhou Heyunjia Hospital, Hangzhou, Zhe'jiang 310000, China
| | - Laixing Yan
- Department of cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310022, China
| | - Wan Chen
- Department of Cardiology, Jiulongpo First People's Hospital, Chongqing 400051, China
| | - Bing Wang
- Department of cardiology, Zouping People's Hospital, Zouping, Shandong 256299, China
| | - Huiju Xu
- Department of cardiology, Hangzhou Mingzhou Hospital, Hangzhou, Zhe'jiang 311215, China.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
20
|
Singh DD, Kim Y, Choi SA, Han I, Yadav DK. Clinical Significance of MicroRNAs, Long Non-Coding RNAs, and CircRNAs in Cardiovascular Diseases. Cells 2023; 12:1629. [PMID: 37371099 DOI: 10.3390/cells12121629] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Based on recent research, the non-coding genome is essential for controlling genes and genetic programming during development, as well as for health and cardiovascular diseases (CVDs). The microRNAs (miRNAs), lncRNAs (long ncRNAs), and circRNAs (circular RNAs) with significant regulatory and structural roles make up approximately 99% of the human genome, which does not contain proteins. Non-coding RNAs (ncRNA) have been discovered to be essential novel regulators of cardiovascular risk factors and cellular processes, making them significant prospects for advanced diagnostics and prognosis evaluation. Cases of CVDs are rising due to limitations in the current therapeutic approach; most of the treatment options are based on the coding transcripts that encode proteins. Recently, various investigations have shown the role of nc-RNA in the early diagnosis and treatment of CVDs. Furthermore, the development of novel diagnoses and treatments based on miRNAs, lncRNAs, and circRNAs could be more helpful in the clinical management of patients with CVDs. CVDs are classified into various types of heart diseases, including cardiac hypertrophy (CH), heart failure (HF), rheumatic heart disease (RHD), acute coronary syndrome (ACS), myocardial infarction (MI), atherosclerosis (AS), myocardial fibrosis (MF), arrhythmia (ARR), and pulmonary arterial hypertension (PAH). Here, we discuss the biological and clinical importance of miRNAs, lncRNAs, and circRNAs and their expression profiles and manipulation of non-coding transcripts in CVDs, which will deliver an in-depth knowledge of the role of ncRNAs in CVDs for progressing new clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, India
| | - Youngsun Kim
- Department of Obstetrics and Gynecology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul 08826, Republic of Korea
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Biodisplay, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Dharmendra Kumar Yadav
- Department of Pharmacy, Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Incheon 21924, Republic of Korea
| |
Collapse
|
21
|
Nardin M, Verdoia M, Laera N, Cao D, De Luca G. New Insights into Pathophysiology and New Risk Factors for ACS. J Clin Med 2023; 12:jcm12082883. [PMID: 37109221 PMCID: PMC10146393 DOI: 10.3390/jcm12082883] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiovascular disease still represents the main cause of mortality worldwide. Despite huge improvements, atherosclerosis persists as the principal pathological condition, both in stable and acute presentation. Specifically, acute coronary syndromes have received substantial research and clinical attention in recent years, contributing to improve overall patients' outcome. The identification of different evolution patterns of the atherosclerotic plaque and coronary artery disease has suggested the potential need of different treatment approaches, according to the mechanisms and molecular elements involved. In addition to traditional risk factors, the finer portrayal of other metabolic and lipid-related mediators has led to higher and deep knowledge of atherosclerosis, providing potential new targets for clinical management of the patients. Finally, the impressive advances in genetics and non-coding RNAs have opened a wide field of research both on pathophysiology and the therapeutic side that are extensively under investigation.
Collapse
Affiliation(s)
- Matteo Nardin
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
- Third Medicine Division, Department of Medicine, ASST Spedali Civili, 25123 Brescia, Italy
| | - Monica Verdoia
- Division of Cardiology, Ospedale degli Infermi, ASL Biella, 13900 Biella, Italy
- Department of Translational Medicine, Eastern Piedmont University, 13100 Novara, Italy
| | - Nicola Laera
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Davide Cao
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU "Policlinico G. Martino", Department of Clinical and Experimental Medicine, University of Messina, 98166 Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant'Ambrogio, 20161 Milan, Italy
| |
Collapse
|
22
|
Hong S, Zhu XY, Jiang Y, Zhang L, Tang H, Jordan KL, Saadiq IM, Huang W, Lerman A, Eirin A, Lerman LO. Autologous Extracellular Vesicles Attenuate Cardiac Injury in Experimental Atherosclerotic Renovascular Disease More Effectively Than Their Parent Mesenchymal Stem/Stromal Cells. Stem Cell Rev Rep 2023; 19:700-712. [PMID: 36344721 PMCID: PMC10073252 DOI: 10.1007/s12015-022-10473-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Abstract
Atherosclerotic renovascular disease (RVD) leads to hypertension, chronic kidney disease (CKD), and heart disease. Intrarenal delivery of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (EVs) attenuate renal injury and suppress release of inflammatory cytokines in porcine RVD. We hypothesized that this strategy would also be useful for cardioprotection. Pigs with renovascular hypertension and metabolic syndrome were studied 4 weeks after treatment with a single intrarenal infusion of autologous MSCs, EVs, or vehicle. Cardiac structure and function were assessed in vivo, and myocardial remodeling and expression of the pro-fibrotic factor growth factor receptor-bound protein-2 (Grb2) were measured ex-vivo. Inflammatory cytokine levels were measured in the systemic circulation and myocardial tissue. Blood pressure was elevated in all RVD groups, but serum creatinine increased in RVD and decreased in both RVD + MSCs and RVD + EVs. RVD-induced diastolic dysfunction (lower E/A ratio) was normalized in both MSCs- and EVs- treated pigs. Intrarenal delivery of MSCs and EVs also attenuated RVD-induced myocardial fibrosis, collagen deposition, and Grb2 expression, yet EVs restored capillary density and inflammation more effectively than MSCs. These observations suggest that autologous EVs attenuate cardiac injury in experimental RVD more effectively than their parent MSCs.
Collapse
Affiliation(s)
- Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Yamei Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lei Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
23
|
Gao R, Li X. Extracellular Vesicles and Pathological Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:17-31. [PMID: 37603270 DOI: 10.1007/978-981-99-1443-2_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Pathological cardiac hypertrophy is a well-recognized risk factor for cardiovascular diseases (CVDs). Although lots of efforts have been made to illustrate the underlying molecular mechanisms, many issues remain undiscovered. Recently, intercellular communication by delivering small molecules between different cell types in the progression of cardiac hypertrophy has been reported, including bioactive nucleic acids or proteins. These extracellular vesicles (EVs) may act in an autocrine or paracrine manner between cardiomyocytes and noncardiomyocytes to provoke or inhibit cardiac remodeling and hypertrophy. Besides, EVs can be used as novel diagnostic or prognostic biomarkers in cardiac hypertrophy and also may serve as potential therapeutic targets due to its biocompatible nature and low immunogenicity. In this chapter, we will first summarize the current knowledge about EVs from different cells in pathological cardiac hypertrophy. Then, we will focus on the value of EVs as therapeutic agents and biomarkers for pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Rongrong Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Martyniak A, Jeż M, Dulak J, Stępniewski J. Adaptation of cardiomyogenesis to the generation and maturation of cardiomyocytes from human pluripotent stem cells. IUBMB Life 2023; 75:8-29. [PMID: 36263833 DOI: 10.1002/iub.2685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022]
Abstract
The advent of methods for efficient generation and cardiac differentiation of pluripotent stem cells opened new avenues for disease modelling, drug testing, and cell therapies of the heart. However, cardiomyocytes (CM) obtained from such cells demonstrate an immature, foetal-like phenotype that involves spontaneous contractions, irregular morphology, expression of embryonic isoforms of sarcomere components, and low level of ion channels. These and other features may affect cellular response to putative therapeutic compounds and the efficient integration into the host myocardium after in vivo delivery. Therefore, novel strategies to increase the maturity of pluripotent stem cell-derived CM are of utmost importance. Several approaches have already been developed relying on molecular changes that occur during foetal and postnatal maturation of the heart, its electromechanical activity, and the cellular composition. As a better understanding of these determinants may facilitate the generation of efficient protocols for in vitro acquisition of an adult-like phenotype by immature CM, this review summarizes the most important molecular factors that govern CM during embryonic development, postnatal changes that trigger heart maturation, as well as protocols that are currently used to generate mature pluripotent stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mateusz Jeż
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
25
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
26
|
Minciunescu A, Genovese L, deFilippi C. Cardiovascular Alterations and Structural Changes in the Setting of Chronic Kidney Disease: a Review of Cardiorenal Syndrome Type 4. SN COMPREHENSIVE CLINICAL MEDICINE 2022; 5:15. [PMID: 36530959 PMCID: PMC9734879 DOI: 10.1007/s42399-022-01347-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 06/17/2023]
Abstract
Cardiovascular and renal physiology are interrelated. More than a decade ago this was codified in guidelines defining the five subtypes of the cardiorenal syndrome. Morbidity and mortality for those with the cardiorenal syndrome is high compared to demographically matched individuals without cardiorenal disease, acute or chronic. The focus of this review will be the epidemiology, the impact of chronic kidney disease on cardiac structure and function, and associated clinical symptoms, outcomes, and potential treatments for patients with chronic reno-cardiac syndrome, or cardiorenal syndrome type 4. Cardiac structural changes can be profound and are described in detail both at a cellular and physiologic level. Integrating therapies for the treatment of causative or resulting comorbidities may ultimately slow progression of both cardiac and renal disease as well as minimize symptoms and death.
Collapse
Affiliation(s)
- Andrei Minciunescu
- Inova Heart and Vascular Institute, 3300 Gallows Rd, Falls Church, VA 22042 USA
| | - Leonard Genovese
- Inova Heart and Vascular Institute, 3300 Gallows Rd, Falls Church, VA 22042 USA
| | | |
Collapse
|
27
|
Mathis BJ, Kato H, Hiramatsu Y. Induction of Cardiac Pathology: Endogenous versus Exogenous Nrf2 Upregulation. Cells 2022; 11:cells11233855. [PMID: 36497112 PMCID: PMC9736027 DOI: 10.3390/cells11233855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of the endogenous antioxidant response to reactive oxygen species as well as a controller of Phase II detoxification in response to xenobiotics. This amenity to specific external manipulation exploits the binding affinity of Nrf2 for its constitutive repressor and degradation facilitator Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1). Derived from both natural and synthesized origins, these compounds have been extensively tested without definitive beneficial results. Unfortunately, multiple terminated trials have shown a negative side to Nrf2 with regard to cardiac pathologies while animal-based studies have demonstrated cardiomyocyte hypertrophy and heart failure after chronic Nrf2 upregulation. Putatively based on autophagic control of Nrf2 activity-modulating upstream factors, new evidence of miRNA involvement has added complexity to this mechanism. What follows is an extensive survey of Nrf2-regulating exogenous compounds that may promote cardiomyopathy, clinical trial evidence, and a comparison to exercise-induced factors that also upregulate Nrf2 while preventing cardiac pathologies.
Collapse
Affiliation(s)
- Bryan J. Mathis
- International Medical Center, University of Tsukuba Hospital, Tsukuba 305-8576, Ibaraki, Japan
- Correspondence: ; Tel.: +81-29-853-3004
| | - Hideyuki Kato
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Yuji Hiramatsu
- International Medical Center, University of Tsukuba Hospital, Tsukuba 305-8576, Ibaraki, Japan
| |
Collapse
|
28
|
Djalinac N, Kolesnik E, Maechler H, Scheruebel-Posch S, Pelzmann B, Rainer PP, Foessl I, Wallner M, Scherr D, Heinemann A, Sedej S, Ljubojevic-Holzer S, von Lewinski D, Bisping E. miR-1183 Is a Key Marker of Remodeling upon Stretch and Tachycardia in Human Myocardium. Int J Mol Sci 2022; 23:ijms23136962. [PMID: 35805966 PMCID: PMC9266684 DOI: 10.3390/ijms23136962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Many cardiac insults causing atrial remodeling are linked to either stretch or tachycardia, but a comparative characterization of their effects on early remodeling events in human myocardium is lacking. Here, we applied isometric stretch or sustained tachycardia at 2.5 Hz in human atrial trabeculae for 6 h followed by microarray gene expression profiling. Among largely independent expression patterns, we found a small common fraction with the microRNA miR-1183 as the highest up-regulated transcript (up to 4-fold). Both, acute stretch and tachycardia induced down-regulation of the predicted miR-1183 target genes ADAM20 and PLA2G7. Furthermore, miR-1183 was also significantly up-regulated in chronically remodeled atrial samples from patients with persistent atrial fibrillation (3-fold up-regulation versus sinus rhythm samples), and in ventricular myocardium from dilative cardiomyopathy hearts (2-fold up-regulation) as compared to non-failing controls. In sum, although stretch and tachycardia show distinct transcriptomic signatures in human atrial myocardium, both cardiac insults consistently regulate the expression of miR-1183 and its downstream targets in acute and chronic remodeling. Thus, elevated expression of miR-1183 might serve as a tissue biomarker for atrial remodeling and might be of potential functional significance in cardiac disease.
Collapse
Affiliation(s)
- Natasa Djalinac
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
- Unit of Human Molecular Genetics and Functional Genomics, Department of Biology, University of Padua, 35121 Padua, Italy
| | - Ewald Kolesnik
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
| | - Heinrich Maechler
- Department of Cardiothoracic Surgery, Medical University of Graz, 8036 Graz, Austria;
| | - Susanne Scheruebel-Posch
- Gottfried Schatz Research Center, Institute of Biophysics, Medical University of Graz, 8010 Graz, Austria; (S.S.-P.); (B.P.)
| | - Brigitte Pelzmann
- Gottfried Schatz Research Center, Institute of Biophysics, Medical University of Graz, 8010 Graz, Austria; (S.S.-P.); (B.P.)
| | - Peter P. Rainer
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
- BioTechMed Graz, 8036 Graz, Austria
| | - Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8010 Graz, Austria;
| | - Markus Wallner
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Correspondence: (M.W.); (D.v.L.); Tel.: +43-316-385-31261 (M.W.); +43-316-385-80684 (D.v.L.)
| | - Daniel Scherr
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, 8010 Graz, Austria;
| | - Simon Sedej
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
- BioTechMed Graz, 8036 Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Senka Ljubojevic-Holzer
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
- BioTechMed Graz, 8036 Graz, Austria
| | - Dirk von Lewinski
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
- Correspondence: (M.W.); (D.v.L.); Tel.: +43-316-385-31261 (M.W.); +43-316-385-80684 (D.v.L.)
| | - Egbert Bisping
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036 Graz, Austria; (N.D.); (E.K.); (P.P.R.); (D.S.); (S.S.); (S.L.-H.); (E.B.)
| |
Collapse
|
29
|
Abstract
The discovery of microRNAs and their role in diseases was a breakthrough that inspired research into microRNAs as drug targets. Cardiovascular diseases are an area in which limitations of conventional pharmacotherapy are highly apparent and where microRNA-based drugs have appreciably progressed into preclinical and clinical testing. In this Review, we summarize the current state of microRNAs as therapeutic targets in the cardiovascular system. We report recent advances in the identification and characterization of microRNAs, their manipulation and clinical translation, and discuss challenges and perspectives toward clinical application.
Collapse
Affiliation(s)
- Bernhard Laggerbauer
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
30
|
Luo F, Bu H. Letter by Luo and Bu Regarding Article, "Comprehensive Proteomics Profiling Reveals Circulating Biomarkers of Hypertrophic Cardiomyopathy". Circ Heart Fail 2022; 15:e009499. [PMID: 35440167 DOI: 10.1161/circheartfailure.122.009499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Fanyan Luo
- Department of Cardiovascular Surgery and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haisong Bu
- Department of Cardiovascular Surgery and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
31
|
Non-Coding RNAs in the Therapeutic Landscape of Pathological Cardiac Hypertrophy. Cells 2022; 11:cells11111805. [PMID: 35681500 PMCID: PMC9180404 DOI: 10.3390/cells11111805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are a major health problem, and long-term survival for people diagnosed with heart failure is, still, unrealistic. Pathological cardiac hypertrophy largely contributes to morbidity and mortality, as effective therapeutic approaches are lacking. Non-coding RNAs (ncRNAs) arise as active regulators of the signaling pathways and mechanisms that govern this pathology, and their therapeutic potential has received great attention in the last decades. Preclinical studies in large animal models have been successful in ameliorating cardiac hypertrophy, and an antisense drug for the treatment of heart failure has, already, entered clinical trials. In this review, we provide an overview of the molecular mechanisms underlying cardiac hypertrophy, the involvement of ncRNAs, and the current therapeutic landscape of oligonucleotides targeting these regulators. Strategies to improve the delivery of such therapeutics and overcome the actual challenges are, also, defined and discussed. With the fast advance in the improvement of oligonucleotide drug delivery, the inclusion of ncRNAs-targeting therapies for cardiac hypertrophy seems, increasingly, a closer reality.
Collapse
|
32
|
Louro AF, Paiva MA, Oliveira MR, Kasper KA, Alves PM, Gomes‐Alves P, Serra M. Bioactivity and miRNome Profiling of Native Extracellular Vesicles in Human Induced Pluripotent Stem Cell-Cardiomyocyte Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104296. [PMID: 35322574 PMCID: PMC9130911 DOI: 10.1002/advs.202104296] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/05/2022] [Indexed: 05/17/2023]
Abstract
Extracellular vesicles (EV) are an attractive therapy to boost cardiac regeneration. Nevertheless, identification of native EV and corresponding cell platform(s) suitable for therapeutic application, is still a challenge. Here, EV are isolated from key stages of the human induced pluripotent stem cell-cardiomyocyte (hiPSC-CM) differentiation and maturation, i.e., from hiPSC (hiPSC-EV), cardiac progenitors, immature and mature cardiomyocytes, with the aim of identifying a promising cell biofactory for EV production, and pinpoint the genetic signatures of bioactive EV. EV secreted by hiPSC and cardiac derivatives show a typical size distribution profile and the expression of specific EV markers. Bioactivity assays show increased tube formation and migration in HUVEC treated with hiPSC-EV compared to EV from committed cell populations. hiPSC-EV also significantly increase cell cycle activity of hiPSC-CM. Global miRNA expression profiles, obtained by small RNA-seq analysis, corroborate an EV-miRNA pattern indicative of stem cell to cardiomyocyte specification, confirming that hiPSC-EV are enriched in pluripotency-associated miRNA with higher in vitro pro-angiogenic and pro-proliferative properties. In particular, a stemness maintenance miRNA cluster upregulated in hiPSC-EV targets the PTEN/PI3K/AKT pathway, involved in cell proliferation and survival. Overall, the findings validate hiPSC as cell biofactories for EV production for cardiac regenerative applications.
Collapse
Affiliation(s)
- Ana F. Louro
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| | - Marta A. Paiva
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| | - Marta R. Oliveira
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| | - Katharina A. Kasper
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| | - Paula M. Alves
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| | - Patrícia Gomes‐Alves
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| | - Margarida Serra
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2781‐901Portugal
- ITQB‐NOVAInstituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAv. da RepúblicaOeiras2780‐157Portugal
| |
Collapse
|
33
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
34
|
Witten A, Martens L, Schäfer AC, Troidl C, Pankuweit S, Vlacil AK, Oberoi R, Schieffer B, Grote K, Stoll M, Markus B. Monocyte subpopulation profiling indicates CDK6-derived cell differentiation and identifies subpopulation-specific miRNA expression sets in acute and stable coronary artery disease. Sci Rep 2022; 12:5589. [PMID: 35379829 PMCID: PMC8979987 DOI: 10.1038/s41598-022-08600-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Coronary artery disease (CAD) is a long-lasting inflammatory disease characterized by monocyte migration into the vessel wall leading to clinical events like myocardial infarction (MI). However, the role of monocyte subsets, especially their miRNA-driven differentiation in this scenario is still in its infancy. Here, we characterized monocyte subsets in controls and disease phenotypes of CAD and MI patients using flow cytometry and miRNA and mRNA expression profiling using RNA sequencing. We observed major differences in the miRNA profiles between the classical (CD14++CD16−) and nonclassical (CD14+CD16++) monocyte subsets irrespective of the disease phenotype suggesting the Cyclin-dependent Kinase 6 (CDK6) to be an important player in monocyte maturation. Between control and MI patients, we found a set of miRNAs to be differentially expressed in the nonclassical monocytes and targeting CCND2 (Cyclin D2) that is able to enhance myocardial repair. Interestingly, miRNAs as miR-125b playing a role in vascular calcification were differentially expressed in the classical subset in patients suffering from CAD and not MI in comparison to control samples. In conclusion, our study describes specific peculiarities of monocyte subset miRNA expression in control and diseased samples and provides basis to further functional analysis and to identify new cardiovascular disease treatment targets.
Collapse
|
35
|
Li H, Zhan J, Chen C, Wang D. MicroRNAs in cardiovascular diseases. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:140-168. [PMID: 37724243 PMCID: PMC10471109 DOI: 10.1515/mr-2021-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/29/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death and disability worldwide, despite the wide diversity of molecular targets identified and the development of therapeutic methods. MicroRNAs (miRNAs) are a class of small (about 22 nucleotides) non-coding RNAs (ncRNAs) that negatively regulate gene expression at the post-transcriptional level in the cytoplasm and play complicated roles in different CVDs. While miRNA overexpression in one type of cell protects against heart disease, it promotes cardiac dysfunction in another type of cardiac cell. Moreover, recent studies have shown that, apart from cytosolic miRNAs, subcellular miRNAs such as mitochondria- and nucleus-localized miRNAs are dysregulated in CVDs. However, the functional properties of cellular- and subcellular-localized miRNAs have not been well characterized. In this review article, by carefully revisiting animal-based miRNA studies in CVDs, we will address the regulation and functional properties of miRNAs in various CVDs. Specifically, the cell-cell crosstalk and subcellular perspective of miRNAs are highlighted. We will provide the background for attractive molecular targets that might be useful in preventing the progression of CVDs and heart failure (HF) as well as insights for future studies.
Collapse
Affiliation(s)
- Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiabing Zhan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
36
|
Luo F, Liu W, Bu H. MicroRNAs in hypertrophic cardiomyopathy: pathogenesis, diagnosis, treatment potential and roles as clinical biomarkers. Heart Fail Rev 2022; 27:2211-2221. [PMID: 35332416 DOI: 10.1007/s10741-022-10231-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 12/28/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heritable cardiomyopathy and is characterized by increased left ventricular wall thickness, but existing diagnostic and treatment approaches face limitations. MicroRNAs (miRNAs) are type of noncoding RNA molecule that plays crucial roles in the pathological process of cardiac remodelling. Accordingly, miRNAs related to HCM may represent potential novel therapeutic targets. In this review, we first discuss the different roles of miRNAs in the development of HCM. We then summarize the roles of common miRNAs as diagnostic and clinical biomarkers in HCM. Finally, we outline current and future challenges and potential new directions for miRNA-based therapeutics for HCM.
Collapse
Affiliation(s)
- Fanyan Luo
- The Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.,National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- The Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.,National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haisong Bu
- The Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China. .,National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
37
|
Siwaponanan P, Kaewkumdee P, Phromawan W, Udompunturak S, Chomanee N, Udol K, Pattanapanyasat K, Krittayaphong R. Increased expression of six-large extracellular vesicle-derived miRNAs signature for nonvalvular atrial fibrillation. J Transl Med 2022; 20:4. [PMID: 34980172 PMCID: PMC8722074 DOI: 10.1186/s12967-021-03213-6] [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: 09/21/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022] Open
Abstract
Backgrounds Non-valvular atrial fibrillation (AF) is the most common type of cardiac arrhythmia. AF is caused by electrophysiological abnormalities and alteration of atrial tissues, which leads to the generation of abnormal electrical impulses. Extracellular vesicles (EVs) are membrane-bound vesicles released by all cell types. Large EVs (lEVs) are secreted by the outward budding of the plasma membrane during cell activation or cell stress. lEVs are thought to act as vehicles for miRNAs to modulate cardiovascular function, and to be involved in the pathophysiology of cardiovascular diseases (CVDs), including AF. This study identified lEV-miRNAs that were differentially expressed between AF patients and non-AF controls. Methods lEVs were isolated by differential centrifugation and characterized by Nanoparticle Tracking Analysis (NTA), Transmission Electron Microscopy (TEM), flow cytometry and Western blot analysis. For the discovery phase, 12 AF patients and 12 non-AF controls were enrolled to determine lEV-miRNA profile using quantitative reverse transcription polymerase chain reaction array. The candidate miRNAs were confirmed their expression in a validation cohort using droplet digital PCR (30 AF, 30 controls). Bioinformatics analysis was used to predict their target genes and functional pathways. Results TEM, NTA and flow cytometry demonstrated that lEVs presented as cup shape vesicles with a size ranging from 100 to 1000 nm. AF patients had significantly higher levels of lEVs at the size of 101–200 nm than non-AF controls. Western blot analysis was used to confirm EV markers and showed the high level of cardiomyocyte expression (Caveolin-3) in lEVs from AF patients. Nineteen miRNAs were significantly higher (> twofold, p < 0.05) in AF patients compared to non-AF controls. Six highly expressed miRNAs (miR-106b-3p, miR-590-5p, miR-339-3p, miR-378-3p, miR-328-3p, and miR-532-3p) were selected to confirm their expression. Logistic regression analysis showed that increases in the levels of these 6 highly expressed miRNAs associated with AF. The possible functional roles of these lEV-miRNAs may involve in arrhythmogenesis, cell apoptosis, cell proliferation, oxygen hemostasis, and structural remodeling in AF. Conclusion Increased expression of six lEV-miRNAs reflects the pathophysiology of AF that may provide fundamental knowledge to develop the novel biomarkers for diagnosis or monitoring the patients with the high risk of AF. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03213-6.
Collapse
Affiliation(s)
- Panjaree Siwaponanan
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pontawee Kaewkumdee
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wilasinee Phromawan
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suthipol Udompunturak
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nusara Chomanee
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kamol Udol
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kovit Pattanapanyasat
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rungroj Krittayaphong
- Division of Cardiology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
38
|
Bi X, Zhang Y, Yu Y, Yuan J, Xu S, Liu F, Ye J, Liu P. MiRNA-339-5p promotes isoproterenol-induced cardiomyocyte hypertrophy by targeting VCP to activate the mTOR signaling. Cell Biol Int 2021; 46:288-299. [PMID: 34854520 DOI: 10.1002/cbin.11731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/09/2021] [Accepted: 11/27/2021] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) regulate multiple biological processes and participate in various cardiovascular diseases. This study aims to investigate the role of miR-339-5p in cardiomyocyte hypertrophy and the involved mechanism. Neonatal rat cardiomyocytes (NRCMs) were cultured and stimulated with isoproterenol (ISO). The hypertrophic responses were monitored by measuring the cell surface area and expression of hypertrophic markers including β-myosin heavy chain (β-MHC) and atrial natriuretic factor (ANF). Bioinformatic prediction tools and dual-luciferase reporter assay were performed to identify the target gene of miR-339-5p. Quantitative real-time polymerase chain reaction and western blot analysis were used to determine the levels of miR-339-5p and its downstream effectors. Our data showed that miR-339-5p was upregulated during cardiomyocyte hypertrophy triggered by ISO. MiR-339-5p overexpression resulted in enlargement of cell size and increased the levels of hypertrophic markers. In contrast, inhibition of miR-339-5p significantly attenuated ISO-induced hypertrophic responses of NRCMs. Valosin-containing protein (VCP), a suppressor of cardiac hypertrophy via inhibiting mechanistic target of rapamycin (mTOR) signaling, was validated as a target of miR-339-5p. MiR-339-5p suppressed VCP protein expression, leading to elevated phosphorylation of mTOR and ribosomal protein S6 kinase (S6K). VCP depletion activated the mTOR/S6K cascade and could compromise the anti-hypertrophic effects of miR-339-5p inhibitor. Additionally, the hypertrophic responses caused by miR-339-5p was alleviated in the presence of mTOR inhibitor rapamycin. In conclusion, our research revealed that miR-339-5p promoted ISO-induced cardiomyocyte hypertrophy by targeting VCP to activate the mTOR signaling, suggesting a promising therapeutic intervention by interfering miR-339-5p.
Collapse
Affiliation(s)
- Xueying Bi
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Yuhong Zhang
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Youhui Yu
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Jing Yuan
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Siting Xu
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Fang Liu
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Jiantao Ye
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, Sun Yat-Sen University, Guangdong, Guangzhou, China
| |
Collapse
|
39
|
MicroRNAs: From Junk RNA to Life Regulators and Their Role in Cardiovascular Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded small non-coding RNA (18–25 nucleotides) that until a few years ago were considered junk RNA. In the last twenty years, they have acquired more importance thanks to the understanding of their influence on gene expression and their role as negative regulators at post-transcriptional level, influencing the stability of messenger RNA (mRNA). Approximately 5% of the genome encodes miRNAs which are responsible for regulating numerous signaling pathways, cellular processes and cell-to-cell communication. In the cardiovascular system, miRNAs control the functions of various cells, such as cardiomyocytes, endothelial cells, smooth muscle cells and fibroblasts, playing a role in physiological and pathological processes and seeming also related to variations in contractility and hereditary cardiomyopathies. They provide a new perspective on the pathophysiology of disorders such as hypertrophy, fibrosis, arrhythmia, inflammation and atherosclerosis. MiRNAs are differentially expressed in diseased tissue and can be released into the circulation and then detected. MiRNAs have become interesting for the development of new diagnostic and therapeutic tools for various diseases, including heart disease. In this review, the concept of miRNAs and their role in cardiomyopathies will be introduced, focusing on their potential as therapeutic and diagnostic targets (as biomarkers).
Collapse
|
40
|
Han C, Yang J, Sun J, Qin G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol Ther 2021; 233:108025. [PMID: 34687770 PMCID: PMC9018895 DOI: 10.1016/j.pharmthera.2021.108025] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/15/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are lipid bilayer particles naturally released from the cell. While exosomes are formed as intraluminal vesicles (ILVs) of the multivesicular endosomes (MVEs) and released to extracellular space upon MVE-plasma membrane fusion, microvesicles are generated through direct outward budding of the plasma membrane. Exosomes and microvesicles have same membrane orientation, different yet overlapping sizes; their cargo contents are selectively packed and dependent on the source cell type and functional state. Both exosomes and microvesicles can transfer bioactive RNAs, proteins, lipids, and metabolites from donor to recipient cells and influence the biological properties of the latter. Over the last decade, their potential roles as effective inter-tissue communicators in cardiovascular physiology and pathology have been increasingly appreciated. In addition, EVs are attractive sources of biomarkers for the diagnosis and prognosis of diseases, because they acquire their complex cargoes through cellular processes intimately linked to disease pathogenesis. Furthermore, EVs obtained from various stem/progenitor cell populations have been tested as cell-free therapy in various preclinical models of cardiovascular diseases and demonstrate unequivocally encouraging benefits. Here we summarize the findings from recent research on the biological functions of EVs in the ischemic heart disease and heart failure, and their potential as novel diagnostic biomarkers and therapeutic opportunities.
Collapse
Affiliation(s)
- Chaoshan Han
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Junjie Yang
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Jiacheng Sun
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA.
| |
Collapse
|
41
|
Yang C, Wen K. Predictive value and regulatory mechanism of serum miR-499a-5p on myocardial dysfunction in sepsis. J Cardiothorac Surg 2021; 16:301. [PMID: 34654440 PMCID: PMC8518260 DOI: 10.1186/s13019-021-01679-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background This study sought to investigate the predictive value and regulatory mechanism of serum miR-499a-5p in sepsis-induced myocardial dysfunction (SIMD). Methods A total of 60 patients with sepsis and 60 healthy volunteers were enrolled in this study. The serum levels of miRNAs (miR-451, miR-378 and miR-499a-5p) were detected. Receiver operating characteristic curve and logistic regression analysis were used to evaluate the diagnostic and prognostic value of miR-499a-5p in SIMD patients. AC16 cells were used to establish SIMD model in vitro using lipopolysaccharide (LPS). An analysis was conducted for miR-499a-5p expression, cell viability, and the concentration of creatine kinase-MB isoform (CK-MB), brain natriuretic peptide (BNP), superoxide dismutase (SOD) and cytochrome C oxidase IV (COX IV). The downstream target of miR-499a-5p was verified. Results Our results revealed a poor expression of miR-499a-5p in the serum of SIMD patients, while no significant difference was evident for miR-451 and miR-378. The level of miR-499a-5p in the survival group was higher than the non-survival group. miR-499a-5p elicited good diagnostic and prognostic value for SIMD. Our findings revealed that miR-499a-5p was decreased significantly in LPS-treated cardiomyocytes. After overexpression of miR-499a-5p, the cell viability increased, and the concentrations of CK-MB and BNP were decreased, while the concentrations of SOD and COX IV were increased. EIF4E was validated as the target of miR-499a-5p. After overexpression of EIF4E, the cell viability was decreased and the concentrations of CK-MB and BNP were increased while the concentrations of SOD and COX IV were decreased. Conclusion The level of miR-499a-5p is weak in SIMD patients. miR-499a-5p has a good diagnostic and prognostic value for SIMD by inhibiting EIF4E transcription.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Critical Care Medicine, The Second Hospital of Shandong University, No. 247 Beiyuan Dajie Street, Jinan City, 250012, Shandong Province, China
| | - Kun Wen
- Department of Critical Care Medicine, The Second Hospital of Shandong University, No. 247 Beiyuan Dajie Street, Jinan City, 250012, Shandong Province, China.
| |
Collapse
|
42
|
Wang Q, Liu K, Jin C. Clinical value of microRNA-378a-3p in sepsis and its role in sepsis-induced inflammation and cardiac dysfunction. Bioengineered 2021; 12:8496-8504. [PMID: 34565302 PMCID: PMC8806767 DOI: 10.1080/21655979.2021.1985339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This study explored the clinical meaning of miR-378a-3p in sepsis and its influence on sepsis-induced inflammation and cardiac dysfunction. Serum levels of miR-378a-3p were detected by quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). The Receiver Operating Characteristic (ROC) curve was used to evaluate its diagnostic value. The effects of miR-378a-3p on inflammation and cardiac function were evaluated by monitoring left ventricular systolic pressure (LVSP), left ventricular and end-diastolic pressure (LVEDP), maximum rate of change in left ventricular pressure (± dp/dtmax) and detecting the levels of troponin I (cTnI), creatine kinase isoenzyme MB (CK-MB), tumor necrosis factor-a (TNF-a), interleukin-6 (IL-6), and interleukin-1β (IL-1β) via enzyme linked immunosorbent assay (ELISA). Serum miR-378a-3p was increased in sepsis patients and rat model. ROC curve indicated that miR-378a-3p might have diagnostic significance for sepsis miR-378a-3p antagomir improved the cardiac function by upregulating the levels of LVSP and ± dp/dtmax, and decreasing the levels of LVEDP, cTnI and CK-MB in rat model. miR-378a-3p antagomir also significantly alleviated the inflammatory responseby down-regulating the expression of TNF-a, IL-6, and IL-1β. Besides, logistics regression analysis illustrated that miR-378a-3p was an independent influencing factor for the onset of cardiac dysfunction in sepsis. miR-378a-3p has the potential as a diagnostic biomarker for sepsis and decreasing the level of miR-378a-3p had the ability to ameliorate cardiac dysfunction and inflammatory response caused by sepsis.
Collapse
Affiliation(s)
- Qing Wang
- Department Of Emergency, Emergency General Hospital, Beijing, China
| | - Kuo Liu
- Department Of Emergency, Emergency General Hospital, Beijing, China
| | - Changming Jin
- Department Of Emergency, Emergency General Hospital, Beijing, China
| |
Collapse
|
43
|
Nasser MI, Masood M, Adlat S, Gang D, Zhu S, Li G, Li N, Chen J, Zhu P. Mesenchymal stem cell-derived exosome microRNA as therapy for cardiac ischemic injury. Biomed Pharmacother 2021; 143:112118. [PMID: 34481378 DOI: 10.1016/j.biopha.2021.112118] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular diseases (CVD) are a significant cause of human health harm. In the past, stem cell therapy was reported to have functional defects, such as immune rejection, tumorigenicity, and infusion toxicity. Exosomes are extracellular vesicles with lipid bilayer membrane structure, containing proteins, lipids, mRNA, miRNA, DNA, and other molecules, which can mediate various biological functions such as immune response, inflammatory response, cell migration, and differentiation intercellular communication. Exosomal miRNAs have outstanding advantages in disease diagnosis and curative effect prediction. Likewise, paracrine factors could also mediate the main therapeutic effect of mesenchymal stem cells. Research has shown that mesenchymal stem cell-derived micro-exosomes, which may come from stem cells, accumulate in the ischemic tissue and regulate cell proliferation, apoptosis, inflammation, and angiogenesis sites of myocardial injury after being transplanted. This review reviewed the molecular mechanisms of exosomes and internal microRNAs derived from mesenchymal stem cells in cardiac ischemic injury repair.
Collapse
Affiliation(s)
- M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Muqaddas Masood
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Salah Adlat
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Deng Gang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Ge Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Nanbo Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Jimei Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
44
|
Zhang Y, Wang H, Xia Y. The expression of miR-211-5p in atherosclerosis and its influence on diagnosis and prognosis. BMC Cardiovasc Disord 2021; 21:371. [PMID: 34340677 PMCID: PMC8330028 DOI: 10.1186/s12872-021-02187-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The purpose of this study was to evaluate the diagnostic and prognostic significance of miR-211-5p in atherosclerosis (AS) by detecting the expression level in serum of patients with AS. METHODS A total of 85 healthy controls and 90 asymptomatic AS patients participated in this study. The expression level of miR-211-5p in all subjects were measured by qRT-PCR. Spearman correlation coefficient was used to evaluate the correlation of miR-211-5p with CRP and CIMT. The ROC curve was established to assess the diagnostic value of miR-211-5p in AS. The Kaplan-Meier survival curve and multivariate COX regression analysis were used to evaluate the prognostic significance of miR-211-5p in AS. RESULTS The expression levels of miR-211-5p in AS patients were significantly lower than in healthy controls (P < 0.001), and miR-211-5p showed a significant negative correlation with CRP (r = - 0.639, P < 0.001) and CIMT (r = - 0.730, P < 0.001). The AUC of the ROC curve was 0.900, the specificity and the sensitivity were 84.7% and 78.9%, respectively, which indicating that miR-211-5p had diagnostic value for AS. Survival analysis showed that patients with low miR-211-5p expression were more likely to have cardiovascular end-point events (Log rank P = 0.013). CONCLUSION Serum miR-211-5p could be used as a new biomarker for the diagnosis of AS, and the low expression of miR-211-5p is associated with the poor prognosis of AS.
Collapse
Affiliation(s)
- Yanxia Zhang
- Department of Health Comprehensive Geriatrics, Yidu Central Hospital of Weifang, No. 4138, Linglongshan Road, Weifang, 262500, Shandong, China.
| | - Huiyun Wang
- Department of Health Comprehensive Geriatrics, Yidu Central Hospital of Weifang, No. 4138, Linglongshan Road, Weifang, 262500, Shandong, China
| | - Yu Xia
- Department of Health Comprehensive Geriatrics, Yidu Central Hospital of Weifang, No. 4138, Linglongshan Road, Weifang, 262500, Shandong, China
| |
Collapse
|
45
|
Martyniak A, Andrysiak K, Motais B, Coste S, Podkalicka P, Ferdek P, Stępniewski J, Dulak J. Generation of microRNA-378a-deficient hiPSC as a novel tool to study its role in human cardiomyocytes. J Mol Cell Cardiol 2021; 160:128-141. [PMID: 34329686 DOI: 10.1016/j.yjmcc.2021.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 06/29/2021] [Accepted: 07/23/2021] [Indexed: 12/17/2022]
Abstract
microRNA-378a (miR-378a) is one of the most highly expressed microRNAs in the heart. However, its role in the human cardiac tissue has not been fully understood. It was observed that miR-378a protects cardiomyocytes from hypertrophic growth by regulation of IGF1R and the expression of downstream kinases. Increased levels of miR-378a were reported in the serum of Duchenne muscular dystrophy (DMD) patients and female carriers of DMD gene-associated mutations with developed cardiomyopathy. In order to shed more light on the role of miR-378a in human cardiomyocytes and its potential involvement in DMD-related cardiomyopathy, we generated two human induced pluripotent stem cell (hiPSC) models; one with deletion of miR-378a and the second one with deletion of DMD exon 50 leading to the DMD phenotype. Our results indicate that lack of miR-378a does not influence the pluripotency of hiPSC and their ability to differentiate into cardiomyocytes (hiPSC-CM). miR-378a-deficient hiPSC-CM exhibited, however, significantly bigger size compared to the isogenic control cells, indicating the role of this miRNA in the hypertrophic growth of human cardiomyocytes. In accordance, the level of NFATc3, phosphoAKT, phosphoERK and ERK was higher in these cells compared to the control counterparts. A similar effect was achieved by silencing miR-378a with antagomirs. Of note, the percentage of cells with nuclear localization of NFATc3 was higher in miR-378a-deficient hiPSC-CM. Analysis of electrophysiological properties and Ca2+ oscillations revealed the decrease in the spike slope velocity and lower frequency of calcium spikes in miR-378a-deficient hiPSC-CM. Interestingly, the level of miR-378a increased gradually during cardiac differentiation of hiPSC. Of note, it was low until day 15 in differentiating DMD-deficient hiPSC-CM and then rose to a similar level as in the isogenic control counterparts. In summary, our findings confirmed the utility of hiPSC-based models for deciphering the role of miR-378a in the control and diseased human cardiomyocytes.
Collapse
Affiliation(s)
- Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Benjamin Motais
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Solène Coste
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Paweł Ferdek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| |
Collapse
|
46
|
Kim JM, Byun JS, Kim J, Park MS, Hong SA, Nam TK, Choi HH, Hong S, Han SH, Jeong HB, Park KY, Kim HR. Analysis of microRNA signatures in ischemic stroke thrombus. J Neurointerv Surg 2021; 14:neurintsurg-2021-017597. [PMID: 34244338 DOI: 10.1136/neurintsurg-2021-017597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/28/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND We investigated the microRNA expression pattern from thrombus retrieved by mechanical thrombectomy in acute stroke patients to understand the stroke mechanism. METHODS This study included acute ischemic stroke patients who had undergone intra-arterial thrombectomy at Chung-Ang University Hospital in Seoul, Korea between February 2016 and March 2019. The thrombus was retrieved and stored at -70℃ after obtaining informed consent. MicroRNA microarray analysis was performed for the patients with identified stroke mechanisms including (1) large artery atherosclerosis, (2) cardioembolism with atrial fibrillation, and (3) cardioembolism with valvular heart disease. The microRNAs derived from microarray analysis were validated by quantitative real-time polymerase chain reaction (qRT-PCR) from different patient populations. The correlation analysis was performed between microRNA levels and laboratory data to understand the functional relevance of the altered microRNA. RESULTS In total, 55 thrombi were obtained from 74 patients, and the microRNAs were analyzed in 45 samples. Microarray analysis of 2578 microRNAs revealed that 50 microRNAs were significantly altered among the three groups. Validation using qRT-PCR showed that miR-378f and miR-450b-5p were significantly elevated among the cardioembolic thrombi; both microRNAs were inversely correlated with the ejection fraction from echocardiography. Thrombi from patients with early neurological deterioration exhibited higher levels of miR-93-5p and lower levels of miR-629-5p than those from neurologically stable patients. CONCLUSIONS The microRNA expression pattern can provide information regarding the mechanism of stroke by reflecting the underlying pathological status of the organ from which the thrombus was derived.
Collapse
Affiliation(s)
- Jeong-Min Kim
- Department of Neurology, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Jun-Soo Byun
- Department of Neurology, Chung-Ang University, Seoul, Seoul, Korea (the Republic of)
| | - Jiah Kim
- Department of Neurology, Samsung Medical Center, Gangnam-gu, Seoul, Korea (the Republic of)
| | - Moo-Seok Park
- Department of Neuroradiology, Ewha Women's University Hospital, Seoul, Korea (the Republic of)
| | - Soon Auck Hong
- Department of Pathology, Chung-Ang University, Seoul, Korea (the Republic of)
| | - Taek-Kyun Nam
- Department of Neurosurgery, Chung-Ang University, Seoul, Korea (the Republic of)
| | - Hyun Ho Choi
- Department of Neurosurgery, Chung-Ang University, Seoul, Korea (the Republic of)
| | - Sungguan Hong
- Department of Chemistry, Chung-Ang University, Seoul, Korea (the Republic of)
| | - Su-Hyun Han
- Department of Laboratory Medicine, Chung-Ang University, Seoul, Korea (the Republic of)
| | - Hae-Bong Jeong
- Department of Neurology, Chung-Ang University, Seoul, Seoul, Korea (the Republic of)
| | - Kwang-Yeol Park
- Department of Neurology, Chung-Ang University, Seoul, Seoul, Korea (the Republic of)
| | - Hye Ryoun Kim
- Department of Laboratory Medicine, Chung-Ang University, Seoul, Korea (the Republic of)
| |
Collapse
|
47
|
Barut Z, Cabbar AT, Yilmaz SG, Akdeniz FT, Simsek MA, Capar B, Degertekin M, Dalan AB, Yerebakan H, Isbir T. Investigation of Circulating miRNA-133, miRNA-26, and miRNA-378 as Candidate Biomarkers for Left Ventricular Hypertrophy. In Vivo 2021; 35:1605-1610. [PMID: 33910842 DOI: 10.21873/invivo.12417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Left ventricular hypertrophy (LVH) involves increased muscular mass of the left ventricle due to increased cardiomyocyte size and is caused by cardiomyopathies. Several microRNAs (miRNAs) have been implicated in processes that contribute to heart disease. This study aimed to examine miRNA-133, miRNA-26 and miRNA-378 as candidate biomarkers to define prognosis in patients with LVH. PATIENTS AND METHODS The study group consisted of 70 patients who were diagnosed with LVH and 16 unaffected individuals who served as the control group. Real-time polymerase chain reaction (RT-PCR) was used to analyze serum miRNA-133, miRNA-26, and miRNA-378 expression levels in LVH patients and the control group. Receiver operating characteristic (ROC) curve analysis was performed to assess the diagnostic capability of miRNA-378. RESULTS When crossing threshold (CT) values were compared between patient and control samples, we found that there were no statistically significant differences in miRNA-133 and miRNA-26 CT values, while the miRNA-378 expression was significantly increased in LVH patients. ROC analysis demonstrated that the expression levels of miRNA-378 (AUC=0.484, p=0.0013) were significantly different between groups. CONCLUSION We observed a statistically significant relationship between miRNA-378 expression levels and LVH, suggesting that circulating miRNA-378 may be used as a novel biomarker to distinguish patients who have LVH from those who do not.
Collapse
Affiliation(s)
- Zerrin Barut
- Department of Basic Medical Science, Faculty of Dentistry, Antalya Bilim University, Antalya, Turkey
| | - Ayca Turer Cabbar
- Department of Cardiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Seda Gulec Yilmaz
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Fatma Tuba Akdeniz
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Mustafa Aytek Simsek
- Department of Cardiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Betul Capar
- Department of Molecular Medicine, Institute of Health Sciences, Yeditepe University, Istanbul, Turkey
| | - Muzaffer Degertekin
- Department of Cardiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Altay Burak Dalan
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Halit Yerebakan
- Department of Cardiovascular Surgery, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Turgay Isbir
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey;
| |
Collapse
|
48
|
Ionescu RF, Cretoiu SM. MicroRNAs as monitoring markers for right-sided heart failure and congestive hepatopathy. J Med Life 2021; 14:142-147. [PMID: 34104236 PMCID: PMC8169151 DOI: 10.25122/jml-2021-0071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The last decades showed a worrying increase in the evolution of cardiovascular diseases towards different stages of heart failure (HF), as a stigma of the western lifestyle. MicroRNAs (miRNAs), non-coding RNAs, which are approximately 22-nucleotide long, were shown to regulate gene expression at the post-transcriptional level and play a role in the pathogenesis and progression of HF. miRNAs research is of high interest nowadays, as these molecules display mechanisms of action that can influence the course of evolution of common chronic diseases, including HF. The potential of post-transcriptional regulation by miRNAs concerning the diagnosis, management, and therapy for HF represents a new promising approach in the accurate assessment of cardiovascular diseases. This review aims to assess the current knowledge of miRNAs in cardiovascular diseases, especially right-sided heart failure and hepatomegaly. Moreover, attention is focused on their role as potential molecular biomarkers and more promising aspects involving miRNAs as future therapeutic targets in the pathophysiology of HF.
Collapse
Affiliation(s)
- Ruxandra Florentina Ionescu
- Department of Cardiology I, Central Military Emergency University Hospital Dr. Carol Davila, Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
49
|
Sileno S, Beji S, D'Agostino M, Carassiti A, Melillo G, Magenta A. microRNAs involved in psoriasis and cardiovascular diseases. VASCULAR BIOLOGY 2021; 3:R49-R68. [PMID: 34291190 PMCID: PMC8284950 DOI: 10.1530/vb-21-0007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022]
Abstract
Psoriasis is a chronic inflammatory disease involving the skin. Both genetic and environmental factors play a pathogenic role in psoriasis and contribute to the severity of the disease. Psoriasis, in fact, has been associated with different comorbidities such as diabetes, metabolic syndrome, gastrointestinal or kidney diseases, cardiovascular disease (CVD), and cerebrovascular diseases (CeVD). Indeed, life expectancy in severe psoriasis is reduced by up to 5 years due to CVD and CeVD. Moreover, patients with severe psoriasis have a higher prevalence of traditional cardiovascular (CV) risk factors, including dyslipidemia, diabetes, smoking, and hypertension. Further, systemic inflammation is associated with oxidative stress increase and induces endothelial damage and atherosclerosis progression. Different miRNA have been already described in psoriasis, both in the skin tissues and in the blood flow, to play a role in the progression of disease. In this review, we will summarize and discuss the most important miRNAs that play a role in psoriasis and are also linked to CVD.
Collapse
Affiliation(s)
- Sara Sileno
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Experimental Immunology Laboratory Via Monti di Creta, Rome, Italy
| | - Sara Beji
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Experimental Immunology Laboratory Via Monti di Creta, Rome, Italy
| | - Marco D'Agostino
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Experimental Immunology Laboratory Via Monti di Creta, Rome, Italy
| | - Alessandra Carassiti
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Experimental Immunology Laboratory Via Monti di Creta, Rome, Italy
| | - Guido Melillo
- Unit of Cardiology, IDI-IRCCS, Via Monti di Creta, Rome, Italy
| | - Alessandra Magenta
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Via Fosso del Cavaliere, Rome, Italy
| |
Collapse
|
50
|
Chen Y, Xu Y, Deng Z, Wang Y, Zheng Y, Jiang W, Jiang L. MicroRNA expression profiling involved in doxorubicin-induced cardiotoxicity using high-throughput deep-sequencing analysis. Oncol Lett 2021; 22:560. [PMID: 34093775 PMCID: PMC8170198 DOI: 10.3892/ol.2021.12821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are sensitive biomarkers and endogenous repressors of gene expression by decreasing mRNA stability and interfering with mRNA translation. Despite a number of investigations revealing the dysregulation of miRNA expression associated with cardiotoxicity induced by doxorubicin (Dox), perturbation of miRNAs directly resulting from Dox at early stage in cardiomyocytes and the target gene interaction remain largely unknown. In the present study, high-throughput deep-sequencing was used to analyze changes in global miRNA expression in H9c2 cardiomyocytes exposed to 5 µg/ml Dox for 0, 12 or 24 h. Compared with the 0-h time point, the expression levels of 386 unique miRNAs were altered. Based on miRNA expression and fold-change, the target genes of 76 selected miRNAs were further analyzed using gene interaction networks and pathway enrichment analysis. These miRNAs were involved in the regulation of different pathways, whose functions included apoptosis, cell proliferation, extracellular matrix remodeling, oxidative stress and lipid metabolism. These differentially expressed miRNAs included let-7 family, miR-29b-3p, miR-378-3/5p, miR-351-3p, miR-664-3p, miR-455-3p, miR-298-3p, miR-702-5p, miR-128-1-5p, miR-671 and miR-421-5p. The present data indicated that global wide miRNA profiling in Dox-induced cardiomyocytes may provide a novel mechanistic insight into understanding Dox-induced heart failure and cardiotoxicity, as well as novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ying Chen
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yingjie Xu
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Zhoufeng Deng
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yin Wang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Ying Zheng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| | - Weihua Jiang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Li Jiang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|