1
|
Abdelhamid AM, Zeinelabdeen Y, Manie T, Khallaf E, Assal RA, Youness RA. miR-17-5p/STAT3/H19: A novel regulatory axis tuning ULBP2 expression in young breast cancer patients. Pathol Res Pract 2024; 263:155638. [PMID: 39388743 DOI: 10.1016/j.prp.2024.155638] [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: 08/04/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND AIM UL-16 binding protein 2 (ULBP2) is a highly altered ligand for the activating receptor, NKG2D in breast cancer (BC). However, the mechanism behind its de-regulation in BC patients remains to be explored. The sophisticated crosstalk between miR-17-5p, the lncRNA H19, and STAT3 as a possible upstream regulatory loop for ULBP2 in young BC patients and cell lines remains as an unexplored area. Therefore, this study aimed at unravelling the ncRNA circuit regulating ULBP2 in young BC patients and cell lines. PATIENTS AND METHODS A total of 30 BC patients were recruited for this study. The expression levels of miR-17-5p, lncRNA H19, and STAT3 were examined in 30 BC tissues compared to their normal counterparts. In addition, the expression signatures of those transcripts were compared in young (<40 years) and old BC (≥40 years) patients. miR-17-5p oligonucleotides, STAT3 and H19 siRNAs were transfected in MDA-MB-231 cells using HiPerfect® Transfection Reagent. miR-17-5p and the transcripts of the target genes quantified using RT-qPCR. Their relative expression was calculated using the 2-ΔΔCT method. RESULTS Through acting as a ceRNA circuit that antagonizes the function of miR-17-5p, H19 prevented the miR-17-5p-induced downregulation of STAT3; this mechanism further contributes to the pathogenesis of BC. Ectopic expression of miR-17-5p in MDA-MB-231 cells displayed its prominent role as an indirect potential activator of NK cells by significantly repressing the expression levels of the oncogenic mediator STAT3 and the oncogenic lncRNA H19 and inducing ULBP2 expression level by 3 folds in TNBC cell lines compared to mock cells. Furthermore, knocking down of STAT3 repressed the lncRNA H19 and increased ULBP2 expression levels, whereas siRNAs against H19 increased the expression levels of ULBP2. CONCLUSION This study highlighted the crosstalk between the novel regulatory network composed of miR-17-5p, H19 and STAT3, and their impact on ULBP2 in BC. Moreover, this study underscored the potential role of miR-17-5p in counteracting the immune evasion tactics, particularly the shedding of ULBP2 in young BC patients, through the modulation of the STAT3/H19/ULBP2 regulatory axis. Thus, targeting this novel regulatory network could potentially enhance our understanding and advance the future application of the innate system-mediated immunotherapy in BC.
Collapse
Affiliation(s)
- A M Abdelhamid
- Biotechnology School, Nile University, Giza 12588, Egypt
| | - Y Zeinelabdeen
- Faculty of Medical Sciences/UMCG, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, the Netherlands
| | - T Manie
- Department of Breast Surgery, National Cancer Institute, Cairo University, Cairo, Egypt
| | - E Khallaf
- Department of General Surgery, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - R A Assal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - R A Youness
- Molecular Genetics Research Team (MGRT), Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University, New Administrative Capital 11835, Egypt.
| |
Collapse
|
2
|
Tuerxun Z, He Y, Niu Y, Bao Z, Liu X, Yang Y, He P. Analysis of Differentially Expressed Murine miRNAs in Acute Myocardial Infarction and Target Genes Related to Heart Rate. Cell Biochem Biophys 2024:10.1007/s12013-024-01528-x. [PMID: 39325365 DOI: 10.1007/s12013-024-01528-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE This study aims to investigate the expression profile of miRNAs significantly dysregulated after acute myocardial infarction (AMI) and their potential targets. METHODS After the establishment of a mouse model of AMI, RNA was extracted from mouse infarcted myocardium. Paired-end sequencing was then performed using the Illumina NovaSeq 6000 system to explore the expression profile of miRNAs. Target genes of downregulated differentially expressed miRNAs (DEmiRNAs) were predicted with miRanda (version 3.3a) and TargetScan (version 6.0). Cytoscape was used to construct a DEmiRNA-mRNA regulatory network to show the regulatory relationship. RT-qPCR was performed to measure miR-142a-3p expression in H2O2-treated rat cardiomyocyte H9c2 cells and heart tissues of MI rats. Cell counting kit-8 and TUNEL assays were conducted to detect H9c2 cell viability and apoptosis. RESULTS There were 33 differentially expressed miRNAs, of which 3 were significantly upregulated and the rest 30 were significantly downregulated. Target genes of these miRNAs were identified, and their functional enrichment was analyzed using gene ontology (GO) analysis. Importantly, target genes that can regulate heart rate and their paired upstream miRNAs attracted attention. Significant expression correlation between heart rate-related targets (Epas1, Bves, Hcn4, Cacna1e, Ank2, Slc8a1, Pde4d) and paired miRNAs (miR-142a-5p, miR-7b-5p, miR-144-3p, miR-34c-5p, miR-223-3p, miR-18a-5p) in mouse myocardial tissues was identified. MiR-142a-3p was downregulated in H9c2 cells and rat infarct tissues, and overexpressing miR-142a-3p restrains H2O2-induced H9c2 cell apoptosis. CONCLUSION Cardioprotective miRNAs, such as miR-142a-3p, were identified in mouse myocardial tissues, and some specific miRNA-target pairs are associated with heart rate regulation.
Collapse
Affiliation(s)
- Zulikaier Tuerxun
- Heart center of the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yuxin He
- University of Edinburgh, Edinburgh, UK
| | - Yunxia Niu
- Department of Cardiovascular Diseases, Gansu Province Hospital of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Zhen Bao
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Xuemei Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yuchun Yang
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Pengyi He
- Heart center of the Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
3
|
Jiang H, Yang J, Li T, Wang X, Fan Z, Ye Q, Du Y. JAK/STAT3 signaling in cardiac fibrosis: a promising therapeutic target. Front Pharmacol 2024; 15:1336102. [PMID: 38495094 PMCID: PMC10940489 DOI: 10.3389/fphar.2024.1336102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
Cardiac fibrosis is a serious health problem because it is a common pathological change in almost all forms of cardiovascular diseases. Cardiac fibrosis is characterized by the transdifferentiation of cardiac fibroblasts (CFs) into cardiac myofibroblasts and the excessive deposition of extracellular matrix (ECM) components produced by activated myofibroblasts, which leads to fibrotic scar formation and subsequent cardiac dysfunction. However, there are currently few effective therapeutic strategies protecting against fibrogenesis. This lack is largely because the molecular mechanisms of cardiac fibrosis remain unclear despite extensive research. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling cascade is an extensively present intracellular signal transduction pathway and can regulate a wide range of biological processes, including cell proliferation, migration, differentiation, apoptosis, and immune response. Various upstream mediators such as cytokines, growth factors and hormones can initiate signal transmission via this pathway and play corresponding regulatory roles. STAT3 is a crucial player of the JAK/STAT pathway and its activation is related to inflammation, malignant tumors and autoimmune illnesses. Recently, the JAK/STAT3 signaling has been in the spotlight for its role in the occurrence and development of cardiac fibrosis and its activation can promote the proliferation and activation of CFs and the production of ECM proteins, thus leading to cardiac fibrosis. In this manuscript, we discuss the structure, transactivation and regulation of the JAK/STAT3 signaling pathway and review recent progress on the role of this pathway in cardiac fibrosis. Moreover, we summarize the current challenges and opportunities of targeting the JAK/STAT3 signaling for the treatment of fibrosis. In summary, the information presented in this article is critical for comprehending the role of the JAK/STAT3 pathway in cardiac fibrosis, and will also contribute to future research aimed at the development of effective anti-fibrotic therapeutic strategies targeting the JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
Dang JY, Zhang W, Chu Y, Chen JH, Ji ZL, Feng P. Downregulation of salusins alleviates hypertrophic cardiomyopathy via attenuating oxidative stress and autophagy. Eur J Med Res 2024; 29:109. [PMID: 38336819 PMCID: PMC10854150 DOI: 10.1186/s40001-024-01676-z] [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: 09/13/2023] [Accepted: 01/15/2024] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Salusins, which are translated from the alternatively spliced mRNA of torsin family 2 member A (TOR2A), play a vital role in regulation of various cardiovascular diseases. However, it remains unclear precisely regarding their roles in hypertrophic cardiomyopathy (HCM). Therefore, this study was conducted to explore therapeutic effect and the underlying mechanisms of salusins on HCM. MATERIAL AND METHODS In vivo experiments, Sprague-Dawley rats were used to induce HCM model by angiotensin (Ang) II infusion for 4 weeks. The rats were randomly divided into four groups, namely, Saline + Control shRNA (n = 7), Ang II + Control shRNA (n = 8), Saline + TOR2A shRNA (n = 7), and Ang II + TOR2A shRNA groups (n = 8). After HCM induction, doppler echocardiography is recommended to evaluate heart function. In vitro experiments, primary neonatal rat cardiomyocytes (NRCMs) and cardiac fibroblasts (NRCFs) were obtained from newborn rats, and were treated with Ang II (10-6 M) for 24 h. RESULTS After treatment with Ang II, levels of salusin-α and salusin-β were elevated in serum and cardiac tissues of rats and in the neonatal rat cardiomyocytes and cardiac fibroblasts. Downregulation of salusins alleviated the Ang II-induced cardiac hypertrophy by suppressing the increased atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and beta-myosin heavy chain (β-MHC) and cardiac fibrosis by blocking collagen I, collagen III and transforming growth factor-beta (TGF-β), and it also attenuated oxidative stress by suppressing the increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels and reversing the decreased superoxide dismutase (SOD) activity and autophagy by inhibiting the increased microtubule-associated protein light chain 3B (LC3B), Beclin1, autophagy related gene (Atg) 3 and Atg5 in the cardiac tissues of Ang II-infused rats and in the Ang II-treated NRCMs. CONCLUSIONS All these findings suggest that the levels of salusins were elevated in the HCM, and targeting of salusins contributes to alleviation of cardiac hypertrophy and fibrosis probably via attenuating oxidative stress and autophagy. Accordingly, targeting of salusins may be a strategy for HCM therapy.
Collapse
Affiliation(s)
- Jing-Yi Dang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Wei Zhang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Yi Chu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Jiang-Hong Chen
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Zhao-Le Ji
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Pin Feng
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China.
| |
Collapse
|
5
|
Qian T, Wenxian T, Anbing H. β-elemene enhances cisplatin sensitivity of non-small cell lung cancer cells via the miR-17-5p/STAT3 axis. Chem Biol Drug Des 2024; 103:e14395. [PMID: 37973414 DOI: 10.1111/cbdd.14395] [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: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
In China, β-elemene, a sesquiterpene compound derived from Curcuma wenyujin, is clinically used to treat many human malignancies, including non-small cell lung cancer (NSCLC). Nonetheless, the role of β-elemene in regulating cisplatin sensitivity of NSCLC cells and the related mechanisms are not clear. This study was conducted to investigate the role of β-elemene in sensitizing NSCLC cells to cisplatin. In this work, cisplatin-resistant NSCLC cell lines were constructed. CCK-8, colony formation, and flow cytometry assays were executed to examine cell viability, growth, and apoptosis. MiR-17-5p and STAT3 expression levels in cells were detected by qRT-PCR. Western blot was executed to determine the expression levels of STAT3 and apoptosis-related proteins (Bax and Bcl-2) in the cells. Dual-luciferase reporter gene experiments were performed to verify the targeting relationship between miR-17-5p and STAT3. Herein, we report that, β-elemene inhibits the viability, and induces the apoptosis of cisplatin-resistant NSCLC cells. Additionally, β-elemene induces the upregulation miR-17-5p and downregulation of STAT3. STAT3 is validated to be a target of miR-17-5p in NSCLC cells. Additionally, the role of β-elemene to repress the viability of cisplatin-resistant NSCLC cells is partially counteracted by miR-17-5p inhibitor or STAT3 overexpression. In summary, our study suggests that β-elemene enhances cisplatin sensitivity of NSCLC cells by modulating miR-17-5p/STAT3 axis, and it may be a choice for the complementary treatment of NSCLC patients.
Collapse
Affiliation(s)
- Tian Qian
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, P.R. China
| | - Tong Wenxian
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, P.R. China
| | - He Anbing
- Department of Oncology, The Fifth Hospital of Wuhan, Wuhan, P.R. China
| |
Collapse
|
6
|
Tran T, Cruz C, Chan A, Awad S, Rajasingh J, Deth R, Gurusamy N. Mesenchymal Stem Cell-Derived Long Noncoding RNAs in Cardiac Injury and Repair. Cells 2023; 12:2268. [PMID: 37759491 PMCID: PMC10527806 DOI: 10.3390/cells12182268] [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: 07/16/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiac injury, such as myocardial infarction and heart failure, remains a significant global health burden. The limited regenerative capacity of the adult heart poses a challenge for restoring its function after injury. Mesenchymal stem cells (MSCs) have emerged as promising candidates for cardiac regeneration due to their ability to differentiate into various cell types and secrete bioactive molecules. In recent years, attention has been given to noncoding RNAs derived from MSCs, particularly long noncoding RNAs (lncRNAs), and their potential role in cardiac injury and repair. LncRNAs are RNA molecules that do not encode proteins but play critical roles in gene regulation and cellular responses including cardiac repair and regeneration. This review focused on MSC-derived lncRNAs and their implications in cardiac regeneration, including their effects on cardiac function, myocardial remodeling, cardiomyocyte injury, and angiogenesis. Understanding the molecular mechanisms of MSC-derived lncRNAs in cardiac injury and repair may contribute to the development of novel therapeutic strategies for treating cardiovascular diseases. However, further research is needed to fully elucidate the potential of MSC-derived lncRNAs and address the challenges in this field.
Collapse
Affiliation(s)
- Talan Tran
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328, USA
| | - Claudia Cruz
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328, USA
| | - Anthony Chan
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328, USA
| | - Salma Awad
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Richard Deth
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
7
|
Zhan C, Liu K, Zhang Y, Zhang Y, He M, Wu R, Bi C, Shen B. Myocardial infarction unveiled: Key miRNA players screened by a novel lncRNA-miRNA-mRNA network model. Comput Biol Med 2023; 160:106987. [PMID: 37141653 DOI: 10.1016/j.compbiomed.2023.106987] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is a major contributor to global mortality, and microRNAs (miRNAs) are important in its pathogenesis. Identifying blood miRNAs with clinical application potential for the early detection and treatment of MI is crucial. METHODS We obtained MI-related miRNA and miRNA microarray datasets from MI Knowledge Base (MIKB) and Gene Expression Omnibus (GEO), respectively. A new feature called target regulatory score (TRS) was proposed to characterize the RNA interaction network. MI-related miRNAs were characterized using TRS, transcription factor (TF) gene proportion (TFP), and ageing-related gene (AG) proportion (AGP) via the lncRNA-miRNA-mRNA network. A bioinformatics model was then developed to predict MI-related miRNAs, which were verified by literature and pathway enrichment analysis. RESULTS The TRS-characterized model outperformed previous methods in identifying MI-related miRNAs. MI-related miRNAs had high TRS, TFP, and AGP values, and combining the three features improved prediction accuracy to 0.743. With this method, 31 candidate MI-related miRNAs were screened from the specific-MI lncRNA-miRNA-mRNA network, associated with key MI pathways like circulatory system processes, inflammatory response, and oxygen level adaptation. Most candidate miRNAs were directly associated with MI according to literature evidence, except hsa-miR-520c-3p and hsa-miR-190b-5p. Furthermore, CAV1, PPARA and VEGFA were identified as MI key genes, and were targeted by most of the candidate miRNAs. CONCLUSIONS This study proposed a novel bioinformatics model based on multivariate biomolecular network analysis to identify putative key miRNAs of MI, which deserve further experimental and clinical validation for translational applications.
Collapse
Affiliation(s)
- Chaoying Zhan
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxin Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Yingbo Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China; Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, Hainan, China
| | - Mengqiao He
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Rongrong Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Cheng Bi
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Bairong Shen
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
8
|
Hu Y, Jiang H, Xu Y, Chen G, Fan R, Zhou Y, Liu Y, Yao Y, Liu R, Chen W, Zhang K, Chen X, Wang R, Qiu Z. Stomatin-like protein 2 deficiency exacerbates adverse cardiac remodeling. Cell Death Discov 2023; 9:63. [PMID: 36788223 PMCID: PMC9929064 DOI: 10.1038/s41420-023-01350-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Myocardial fibrosis, oxidative stress, and autophagy both play key roles in the progression of adverse cardiac remodeling. Stomatin-like protein 2 (SLP-2) is closely related to mitochondrial function, but little is known about its role and mechanism in cardiac remodeling. We developed doxorubicin (Dox), angiotensin (Ang) II, and myocardial ischemia-reperfusion (I/R) injury induced cardiac remodeling model and Dox treated H9C2 cell injury model using SLP-2 knockout (SLP-2-/-) mice and H9C2 cells with low SLP-2 expression. We first examined cardiac functional and structural changes as well as levels of oxidative stress, apoptosis and autophagy. We found that SLP-2 deficiency leads to decreased cardiac function and promotes myocardial fibrosis. After Dox and Ang II treatment, SLP-2 deficiency further aggravated myocardial fibrosis, increased myocardial oxidative stress and apoptosis, and activated autophagy by inhibiting PI3K-Akt-mTOR signaling pathway, ultimately exacerbating adverse cardiac remodeling. Similarly, SLP-2 deficiency further exacerbates adverse cardiac remodeling after myocardial I/R injury. Moreover, we extracted cardiomyocyte mitochondria for proteomic analysis, suggesting that SLP-2 deficiency may be involved in myocardial I/R injury induced adverse cardiac remodeling by influencing ubiquitination of intramitochondrial proteins. In addition, the oxidative stress, apoptosis and autophagy levels of H9C2 cells with low SLP-2 expression were further enhanced, and the PI3K-Akt-mTOR signaling pathway was further inhibited under Dox stimulation. Our results suggest that SLP-2 deficiency promotes myocardial fibrosis, disrupts normal mitochondrial function, overactivates autophagy via PI3K-Akt-mTOR signaling pathway, affects the level of ubiquitination, leads to irreversible myocardial damage, and ultimately exacerbates adverse cardiac remodeling.
Collapse
Affiliation(s)
- Yuntao Hu
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Hongwei Jiang
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Yueyue Xu
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Ganyi Chen
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Rui Fan
- grid.263826.b0000 0004 1761 0489School of Medicine, Southeast University, Jiangsu, China
| | - Yifei Zhou
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Yafeng Liu
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Yiwei Yao
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Renjie Liu
- grid.263826.b0000 0004 1761 0489School of Medicine, Southeast University, Jiangsu, China
| | - Wen Chen
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Ke Zhang
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Changzhou Second People’s Hospital, Nanjing Medical University, Jiangsu, China
| | - Xin Chen
- grid.89957.3a0000 0000 9255 8984Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Rui Wang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China.
| | - Zhibing Qiu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China.
| |
Collapse
|
9
|
Srivastava S, Garg I, Singh Y, Meena R, Ghosh N, Kumari B, Kumar V, Eslavath MR, Singh S, Dogra V, Bargotya M, Bhattar S, Gupta U, Jain S, Hussain J, Varshney R, Ganju L. Evaluation of altered miRNA expression pattern to predict COVID-19 severity. Heliyon 2023; 9:e13388. [PMID: 36743852 PMCID: PMC9889280 DOI: 10.1016/j.heliyon.2023.e13388] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Outbreak of COVID-19 pandemic in December 2019 affected millions of people globally. After substantial research, several biomarkers for COVID-19 have been validated however no specific and reliable biomarker for the prognosis of patients with COVID-19 infection exists. Present study was designed to identify specific biomarkers to predict COVID-19 severity and tool for formulating treatment. A small cohort of subjects (n = 43) were enrolled and categorized in four study groups; Dead (n = 16), Severe (n = 10) and Moderate (n = 7) patients and healthy controls (n = 10). Small RNA sequencing was done on Illumina platform after isolation of microRNA from peripheral blood. Differential expression (DE) of miRNA (patients groups compared to control) revealed 118 down-regulated and 103 up-regulated known miRNAs with fold change (FC) expression ≥2 folds and p ≤ 0.05. DE miRNAs were then subjected to functional enrichment and network analysis. Bioinformatic analysis resulted in 31 miRNAs (24 Down-regulated; 7 up-regulated) significantly associated with COVID-19 having AUC>0.8 obtained from ROC curve. Seventeen out of 31 DE miRNAs have been linked to COVID-19 in previous studies. Three miRNAs, hsa-miR-147b-5p and hsa-miR-107 (down-regulated) and hsa-miR-1299 (up-regulated) showed significant unique DE in Dead patients. Another set of 4 miRNAs, hsa-miR-224-5p (down-regulated) and hsa-miR-4659b-3p, hsa-miR-495-3p and hsa-miR-335-3p were differentially up-regulated uniquely in Severe patients. Members of three miRNA families, hsa-miR-20, hsa-miR-32 and hsa-miR-548 were significantly down-regulated in all patients group in comparison to healthy controls. Thus a distinct miRNA expression profile was observed in Dead, Severe and Moderate COVID-19 patients. Present study suggests a panel of miRNAs which identified in COVID-19 patients and could be utilized as potential diagnostic biomarkers for predicting COVID-19 severity.
Collapse
Affiliation(s)
- Swati Srivastava
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India,Corresponding author
| | - Iti Garg
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India,Corresponding author
| | - Yamini Singh
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Ramesh Meena
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Nilanjana Ghosh
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Babita Kumari
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Vinay Kumar
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Malleswara Rao Eslavath
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Sayar Singh
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Vikas Dogra
- Pulmonary Medicine, Rajiv Gandhi Super Speciality Hospital (RGSSH), Delhi, India
| | - Mona Bargotya
- Pulmonary Medicine, Rajiv Gandhi Super Speciality Hospital (RGSSH), Delhi, India
| | - Sonali Bhattar
- Pulmonary Medicine, Rajiv Gandhi Super Speciality Hospital (RGSSH), Delhi, India
| | - Utkarsh Gupta
- Pulmonary Medicine, Rajiv Gandhi Super Speciality Hospital (RGSSH), Delhi, India
| | - Shruti Jain
- Pulmonary Medicine, Rajiv Gandhi Super Speciality Hospital (RGSSH), Delhi, India
| | - Javid Hussain
- Pulmonary Medicine, Rajiv Gandhi Super Speciality Hospital (RGSSH), Delhi, India
| | - Rajeev Varshney
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Lilly Ganju
- Genomics Division, Defence Institute of Physiology and Allied Science (DIPAS), Lucknow Road, Timarpur, Delhi, 110054, India
| |
Collapse
|
10
|
Amorfrutin B Protects Mouse Brain Neurons from Hypoxia/Ischemia by Inhibiting Apoptosis and Autophagy Processes Through Gene Methylation- and miRNA-Dependent Regulation. Mol Neurobiol 2023; 60:576-595. [PMID: 36324052 PMCID: PMC9849175 DOI: 10.1007/s12035-022-03087-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Amorfrutin B is a selective modulator of the PPARγ receptor, which has recently been identified as an effective neuroprotective compound that protects brain neurons from hypoxic and ischemic damage. Our study demonstrated for the first time that a 6-h delayed post-treatment with amorfrutin B prevented hypoxia/ischemia-induced neuronal apoptosis in terms of the loss of mitochondrial membrane potential, heterochromatin foci formation, and expression of specific genes and proteins. The expression of all studied apoptosis-related factors was decreased in response to amorfrutin B, both during hypoxia and ischemia, except for the expression of anti-apoptotic BCL2, which was increased. After post-treatment with amorfrutin B, the methylation rate of the pro-apoptotic Bax gene was inversely correlated with the protein level, which explained the decrease in the BAX/BCL2 ratio as a result of Bax hypermethylation. The mechanisms of the protective action of amorfrutin B also involved the inhibition of autophagy, as evidenced by diminished autophagolysosome formation and the loss of neuroprotective properties of amorfrutin B after the silencing of Becn1 and/or Atg7. Although post-treatment with amorfrutin B reduced the expression levels of Becn1, Nup62, and Ambra1 during hypoxia, it stimulated Atg5 and the protein levels of MAP1LC3B and AMBRA1 during ischemia, supporting the ambiguous role of autophagy in the development of brain pathologies. Furthermore, amorfrutin B affected the expression levels of apoptosis-focused and autophagy-related miRNAs, and many of these miRNAs were oppositely regulated by amorfrutin B and hypoxia/ischemia. The results strongly support the position of amorfrutin B among the most promising anti-stroke and wide-window therapeutics.
Collapse
|
11
|
López-Cepeda L, Castro JD, Aristizábal-Pachón AF, González-Giraldo Y, Pinzón A, Puentes-Rozo PJ, González J. Modulation of Small RNA Signatures by Astrocytes on Early Neurodegeneration Stages; Implications for Biomarker Discovery. Life (Basel) 2022; 12:1720. [PMID: 36362875 PMCID: PMC9696502 DOI: 10.3390/life12111720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/01/2022] [Accepted: 10/12/2022] [Indexed: 04/04/2024] Open
Abstract
Diagnosis of neurodegenerative disease (NDD) is complex, therefore simpler, less invasive, more accurate biomarkers are needed. small non-coding RNA (sncRNA) dysregulates in NDDs and sncRNA signatures have been explored for the diagnosis of NDDs, however, the performance of previous biomarkers is still better. Astrocyte dysfunction promotes neurodegeneration and thus derived scnRNA signatures could provide a more precise way to identify of changes related to NDD course and pathogenesis, and it could be useful for the dissection of mechanistic insights operating in NDD. Often sncRNA are transported outside the cell by the action of secreted particles such as extracellular vesicles (EV), which protect sncRNA from degradation. Furthermore, EV associated sncRNA can cross the BBB to be found in easier to obtain peripheral samples, EVs also inherit cell-specific surface markers that can be used for the identification of Astrocyte Derived Extracellular Vesicles (ADEVs) in a peripheral sample. By the study of the sncRNA transported in ADEVs it is possible to identify astrocyte specific sncRNA signatures that could show astrocyte dysfunction in a more simpler manner than previous methods. However, sncRNA signatures in ADEV are not a copy of intracellular transcriptome and methodological aspects such as the yield of sncRNA produced in ADEV or the variable amount of ADEV captured after separation protocols must be considered. Here we review the role as signaling molecules of ADEV derived sncRNA dysregulated in conditions associated with risk of neurodegeneration, providing an explanation of why to choose ADEV for the identification of astrocyte-specific transcriptome. Finally, we discuss possible limitations of this approach and the need to improve the detection limits of sncRNA for the use of ADEV derived sncRNA signatures.
Collapse
Affiliation(s)
- Leonardo López-Cepeda
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Juan David Castro
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | | | - Yeimy González-Giraldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Pedro J. Puentes-Rozo
- Grupo de Neurociencias del Caribe, Unidad de Neurociencias Cognitivas, Universidad Simón Bolívar, Barranquilla 080002, Colombia
- Grupo de Neurociencias del Caribe, Universidad del Atlántico, Barranquilla 080007, Colombia
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| |
Collapse
|
12
|
Ortuño-Sahagún D, Enterría-Rosales J, Izquierdo V, Griñán-Ferré C, Pallàs M, González-Castillo C. The Role of the miR-17-92 Cluster in Autophagy and Atherosclerosis Supports Its Link to Lysosomal Storage Diseases. Cells 2022; 11:cells11192991. [PMID: 36230953 PMCID: PMC9564236 DOI: 10.3390/cells11192991] [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: 08/15/2022] [Revised: 09/08/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Establishing the role of non-coding RNA (ncRNA), especially microRNAs (miRNAs), in the regulation of cell function constitutes a current research challenge. Two to six miRNAs can act in clusters; particularly, the miR-17-92 family, composed of miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, and miR-92a is well-characterized. This cluster functions during embryonic development in cell differentiation, growth, development, and morphogenesis and is an established oncogenic cluster. However, its role in the regulation of cellular metabolism, mainly in lipid metabolism and autophagy, has received less attention. Here, we argue that the miR-17-92 cluster is highly relevant for these two processes, and thus, could be involved in the study of pathologies derived from lysosomal deficiencies. Lysosomes are related to both processes, as they control cholesterol flux and regulate autophagy. Accordingly, we compiled, analyzed, and discussed current evidence that highlights the cluster's fundamental role in regulating cellular energetic metabolism (mainly lipid and cholesterol flux) and atherosclerosis, as well as its critical participation in autophagy regulation. Because these processes are closely related to lysosomes, we also provide experimental data from the literature to support our proposal that the miR-17-92 cluster could be involved in the pathogenesis and effects of lysosomal storage diseases (LSD).
Collapse
Affiliation(s)
- Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Correspondence: (D.O.-S.); (C.G.-C.)
| | - Julia Enterría-Rosales
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan 45201, Jalisco, Mexico
| | - Vanesa Izquierdo
- Pharmacology and Toxicology Section and Institute of Neuroscience, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08007 Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology and Toxicology Section and Institute of Neuroscience, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08007 Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology and Toxicology Section and Institute of Neuroscience, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08007 Barcelona, Spain
| | - Celia González-Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan 45201, Jalisco, Mexico
- Correspondence: (D.O.-S.); (C.G.-C.)
| |
Collapse
|
13
|
Wu L, Li Z, Li Y. The crosstalk between STAT3 and microRNA in cardiac diseases and protection. Front Cardiovasc Med 2022; 9:986423. [PMID: 36148063 PMCID: PMC9485608 DOI: 10.3389/fcvm.2022.986423] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), an important transcription factor and signaling molecule, play an important role in cardiac disease and protection. As a transcription factor, STAT3 upregulates anti-oxidative and anti-apoptotic genes but suppresses anti-inflammatory and anti-fibrotic genes in cardiac disease and protection. As a signaling molecule, STAT3 is the downstream or upstream of other molecules for signaling transduction, also activated in cardiac disease and protection. MicroRNAs (miRNAs) are endogenous short non-coding RNAs that regulate mRNA expression at the transcriptional level and prevent protein translation. Recently, STAT3 is reported to be not only the target of miRNA but also the inhibitor or inducer of miRNA to modify the mRNA expression profiles in cardiomyocytes resulting in different effects on cardiac disease and protection. We summarize the current knowledge on STAT3 regulation of individual miRNAs and the modulation of STAT3 by miRNAs in cardiac diseases and protection.
Collapse
Affiliation(s)
- Lan Wu
- Affiliated Zhoupu Hospital and Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Lan Wu
| | - Zhizheng Li
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yanfei Li
- Affiliated Zhoupu Hospital and Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
14
|
Santovito D, Weber C. Non-canonical features of microRNAs: paradigms emerging from cardiovascular disease. Nat Rev Cardiol 2022; 19:620-638. [PMID: 35304600 DOI: 10.1038/s41569-022-00680-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 02/08/2023]
Abstract
Research showing that microRNAs (miRNAs) are versatile regulators of gene expression has instigated tremendous interest in cardiovascular research. The overwhelming majority of studies are predicated on the dogmatic notion that miRNAs regulate the expression of specific target mRNAs by inhibiting mRNA translation or promoting mRNA decay in the RNA-induced silencing complex (RISC). These efforts mostly identified and dissected contributions of multiple regulatory networks of miRNA-target mRNAs to cardiovascular pathogenesis. However, evidence from studies in the past decade indicates that miRNAs also operate beyond this canonical paradigm, featuring non-conventional regulatory functions and cellular localizations that have a pathophysiological role in cardiovascular disease. In this Review, we highlight the functional relevance of atypical miRNA biogenesis and localization as well as RISC heterogeneity. Moreover, we delineate remarkable non-canonical examples of miRNA functionality, including direct interactions with proteins beyond the Argonaute family and their role in transcriptional regulation in the nucleus and in mitochondria. We scrutinize the relevance of non-conventional biogenesis and non-canonical functions of miRNAs in cardiovascular homeostasis and pathology, and contextualize how uncovering these non-conventional properties can expand the scope of translational research in the cardiovascular field and beyond.
Collapse
Affiliation(s)
- Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany. .,Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy.
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany. .,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
15
|
Kansakar U, Varzideh F, Mone P, Jankauskas SS, Santulli G. Functional Role of microRNAs in Regulating Cardiomyocyte Death. Cells 2022; 11:983. [PMID: 35326433 PMCID: PMC8946783 DOI: 10.3390/cells11060983] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
microRNAs (miRNA, miRs) play crucial roles in cardiovascular disease regulating numerous processes, including inflammation, cell proliferation, angiogenesis, and cell death. Herein, we present an updated and comprehensive overview of the functional involvement of miRs in the regulation of cardiomyocyte death, a central event in acute myocardial infarction, ischemia/reperfusion, and heart failure. Specifically, in this systematic review we are focusing on necrosis, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Fahimeh Varzideh
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Pasquale Mone
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Stanislovas S. Jankauskas
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Santulli
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
16
|
Deng F, Wu Z, Xu M, Xia P. YAP Activates STAT3 Signalling to Promote Colonic Epithelial Cell Proliferation in DSS-Induced Colitis and Colitis Associated Cancer. J Inflamm Res 2022; 15:5471-5482. [PMID: 36164660 PMCID: PMC9508680 DOI: 10.2147/jir.s377077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND AIMS Yes-associated protein (YAP) is a key transcriptional coactivator of cell proliferation and differentiation. In this study, we sought to identify the roles of YAP in colonic epithelial regeneration and tumourigenesis. METHODS Murine DSS-induced colitis and YAP overexpression models were constructed via lentiviral intraperitoneal injection. Stable YAP-overexpressing cells, protein immunoprecipitation, and ChIP were used to deeply explore the molecular mechanism. RESULTS We found that the expression of YAP was dramatically diminished in the colonic crypts during the acute colitis phase, while YAP was strikingly enhanced to initiate tissue repair after DSS withdrawal. Overexpressing YAP in mice drastically accelerated epithelial regeneration, presenting with more intact structural integrity and reduced inflammatory cell infiltration in the mucosa. Further mechanistic studies showed that the expression of YAP in the nucleus was significantly increased by 2 h post-DSS removal, accompanied by upregulated protein levels of activated STAT3. Overexpression of YAP (YAPWT) elevated the expression of activated STAT3 and its transcriptional targets and strengthened the proliferation and "wound healing" ability of colonic cells. However, these effects were reversed when STAT3 was silenced in YAPWT cells. Moreover, YAP could directly interact with STAT3 in the nucleus, and c-Myc and CyclinD1 were the transcriptional targets. Finally, during colitis-associated cancer (CAC), YAPWT promoted the progression of CAC, while the phosphomimetic YAP downregulated the expression of STAT3 and inhibited the development and progression of CAC. CONCLUSION YAP activates STAT3 signalling to facilitate mucosal regeneration after DSS-induced colitis. However, excessive YAP activation in the colonic epithelium promotes CAC development.
Collapse
Affiliation(s)
- Feihong Deng
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
- Correspondence: Feihong Deng, Department of Gastroenterology, the Second Xiangya Hospital of Central South University; Research Center of Digestive Disease, Central South University, Changsha, Hunan, 410011, People’s Republic of China, Email
| | - Zengrong Wu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| | - Mengmeng Xu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| | - Pianpian Xia
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| |
Collapse
|