1
|
Liu Y, Yuan J, Zhang Y, Ma T, Ji Q, Tian S, Liu C. Non-coding RNA as a key regulator and novel target of apoptosis in diabetic cardiomyopathy: Current status and future prospects. Cell Signal 2025; 128:111632. [PMID: 39922440 DOI: 10.1016/j.cellsig.2025.111632] [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: 10/12/2024] [Revised: 01/19/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
The occurrence of diabetic cardiomyopathy (DCM) can be independent of several risk factors such as hypertension and myocardial ischemia, which can lead to heart failure, thus seriously threatening human health and life. Sustained hyperglycemic stimulation can induce cardiomyocyte apoptosis, which is recognized as the pathological basis of DCM. It has been demonstrated that dysregulation induced by apoptosis is closely associated to progression of DCM, but mechanisms behind it requires further clarification. Currently, increasing evidence has shown that non-coding RNA (ncRNA), especially microRNA, long-chain non-coding RNA (lncRNA), and circular RNA (circRNA), play a regulative role in apoptosis, thus affecting the progression of DCM. Notably, some ncRNAs have also exhibit potential significance as biomarkers and/or therapeutic targets for patients with DCM. In this review, recent findings regarding the potential mechanisms of ncRNA in regulating apoptosis and their role in the progression of DCM were systematically summarized in this research. The conclusion reveals that ncRNA abnormalities exert a crucial role in pathological changes of DCM, which offers potential therapeutic targets for the prevention of DCM.
Collapse
Affiliation(s)
- Yicheng Liu
- College of Rehabilitation Medicine,Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Yuan
- Science and Technology Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yuhang Zhang
- College of Rehabilitation Medicine,Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ting Ma
- College of Rehabilitation Medicine,Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qianqian Ji
- Department one of Cardiovascular Disease, Tai'an Hospital of Traditional Chinese Medicine, Taian 271000, China
| | - Sheng Tian
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, PR China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chunxiao Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|
2
|
Ma YT, Laga T, Zhong CN, Zhuang BQ, Quan HL, Hong L. ANP Increases Zn 2⁺ Accumulation During Reperfusion in Ex Vivo and In Vivo Hearts. Curr Med Sci 2025; 45:35-50. [PMID: 40014195 DOI: 10.1007/s11596-025-00019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE Atrial natriuretic peptide (ANP) and Zn2⁺ have been shown to confer cardioprotection against ischemia/reperfusion (I/R) injury. Zn2⁺ alleviates myocardial hypertrophy and pulmonary hypertension by regulating ANP expression, but its precise role in ANP-mediated cardioprotection remains unclear. This study aimed to investigate whether ANP protects the heart during reperfusion by modulating Zn2⁺ levels and to explore the underlying mechanisms involved. METHODS In this study, we utilized an isolated reperfused heart model in rats, as well as wild-type (WT) and ANP knockout (ANP-/-) mouse models, for in vivo I/R experiments. For clinical investigations, plasma samples were collected from 216 patients with ischemia-related diseases. Evans blue and TTC staining, radioimmunoassay, ICP‒OES, echocardiography, Hydro-Cy3-mediated ROS detection, and Western blotting were employed to evaluate the effect of ANP on Zn2⁺ homeostasis. RESULTS Plasma ANP levels were significantly elevated in patients with ST-elevation myocardial infarction (STEMI), non-ST-elevation myocardial infarction (NSTEMI), and heart failure (HF). ANP secretion increased during reperfusion, rather than infarction, both ex vivo and in vivo, promoting Zn2⁺ accumulation in reperfused tissue. ANP and Zn2⁺ protected mitochondria and reduced infarct size; these effects were reversed by the Zn2⁺ chelator TPEN. In WT and ANP-/- mice, EF% and FS% decreased after reperfusion, with ANP-/- mice exhibiting significantly worse cardiac function. ANP pretreatment alone improved cardiac function, but combined pretreatment with ANP and TPEN decreased EF% and FS% while increasing LVID. Reperfusion increased ROS levels in both WT and ANP-/- hearts, which were reduced by ANP pretreatment. I/R injury elevated Zn2⁺ transporter 8 (ZnT8) expression, an effect that was counteracted by ANP, although this effect was reversed by TPEN. Hypoxia-inducible factor 1-alpha (HIF-1α) expression was elevated in I/R rats and ANP-/- mice, and it was inhibited by both Zn2⁺ and ANP pretreatment. However, the HIF-1α inhibitor 2-Me did not reverse the effect of ANP on ZnT8 expression. Additionally, ANP increased PI3K expression in both WT and ANP-/- I/R mice, but this effect was blocked by the PI3K inhibitor LY294002. CONCLUSIONS ANP modulates Zn2⁺ homeostasis during reperfusion injury by downregulating ZnT8 through the PI3K signalling pathway, thereby reducing myocardial I/R injury.
Collapse
Affiliation(s)
- Yu-Ting Ma
- Department of Physiology and Pathophysiology, School of Medicine, Yanbian University, Yanji, 133002, China
| | - Tong Laga
- Department of General Practice, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Chong-Ning Zhong
- Department of Physiology and Pathophysiology, School of Medicine, Yanbian University, Yanji, 133002, China
| | - Bing-Qi Zhuang
- Department of Physiology and Pathophysiology, School of Medicine, Yanbian University, Yanji, 133002, China
| | - Hai-Lian Quan
- Department of Physiology and Pathophysiology, School of Medicine, Yanbian University, Yanji, 133002, China.
| | - Lan Hong
- Department of Physiology and Pathophysiology, School of Medicine, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
3
|
Al-Awar A, Hussain S. Interplay of Reactive Oxygen Species (ROS) and Epigenetic Remodelling in Cardiovascular Diseases Pathogenesis: A Contemporary Perspective. FRONT BIOSCI-LANDMRK 2024; 29:398. [PMID: 39614429 DOI: 10.31083/j.fbl2911398] [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: 05/24/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) continue to be the leading cause of mortality worldwide, necessitating the development of novel therapies. Despite therapeutic advancements, the underlying mechanisms remain elusive. Reactive oxygen species (ROS) show detrimental effects at high concentrations but act as essential signalling molecules at physiological levels, playing a critical role in the pathophysiology of CVD. However, the link between pathologically elevated ROS and CVDs pathogenesis remains poorly understood. Recent research has highlighted the remodelling of the epigenetic landscape as a crucial factor in CVD pathologies. Epigenetic changes encompass alterations in DNA methylation, post-translational histone modifications, adenosine triphosphate (ATP)-dependent chromatin modifications, and noncoding RNA transcripts. Unravelling the intricate link between ROS and epigenetic changes in CVD is challenging due to the complexity of epigenetic signals in gene regulation. This review aims to provide insights into the role of ROS in modulating the epigenetic landscape within the cardiovascular system. Understanding these interactions may offer novel therapeutic strategies for managing CVD by targeting ROS-induced epigenetic changes. It has been widely accepted that epigenetic modifications are established during development and remain fixed once the lineage-specific gene expression pattern is achieved. However, emerging evidence has unveiled its remarkable dynamism. Consequently, it is now increasingly recognized that epigenetic modifications may serve as a crucial link between ROS and the underlying mechanisms implicated in CVD.
Collapse
Affiliation(s)
- Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, 41345 Gothenburg, Sweden
| | - Shafaat Hussain
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, 41345 Gothenburg, Sweden
| |
Collapse
|
4
|
Gonzalez-Candia A, Figueroa EG, Krause BJ. Pharmacological and molecular mechanisms of miRNA-based therapies for targeting cardiovascular dysfunction. Biochem Pharmacol 2024; 228:116318. [PMID: 38801924 DOI: 10.1016/j.bcp.2024.116318] [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: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Advances in understanding gene expression regulation through epigenetic mechanisms have contributed to elucidating the regulatory mechanisms of noncoding RNAs as pharmacological targets in several diseases. MicroRNAs (miRs) are a class of evolutionarily conserved, short, noncoding RNAs regulating in a concerted manner gene expression at the post-transcriptional level by targeting specific sequences of the 3'-untranslated region of mRNA. Conversely, mechanisms of cardiovascular disease (CVD) remain largely elusive due to their life-course origins, multifactorial pathophysiology, and co-morbidities. In this regard, CVD treatment with conventional medications results in therapeutic failure due to progressive resistance to monotherapy, which overlooks the multiple factors involved, and reduced adherence to poly-pharmacology approaches. Consequently, considering its role in regulating complete gene pathways, miR-based drugs have appreciably progressed into preclinical and clinical testing. This review summarizes the current knowledge about the mechanisms of miRs in cardiovascular disease, focusing specifically on describing how clinical chemistry and physics have improved the stability of the miR molecule. In addition, a comprehensive review of the main miRs involved in cardiovascular disease and the clinical trials in which these molecules are used as active pharmacological molecules is provided.
Collapse
Affiliation(s)
- Alejandro Gonzalez-Candia
- Laboratory of Fetal Neuroprogramming (www.neurofetal-lab.cl), Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile
| | - Esteban G Figueroa
- Laboratory of Fetal Neuroprogramming (www.neurofetal-lab.cl), Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile
| | - Bernardo J Krause
- Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile.
| |
Collapse
|
5
|
Chen G, Zou J, He Q, Xia S, Xiao Q, Du R, Zhou S, Zhang C, Wang N, Feng Y. The Role of Non-Coding RNAs in Regulating Cachexia Muscle Atrophy. Cells 2024; 13:1620. [PMID: 39404384 PMCID: PMC11482569 DOI: 10.3390/cells13191620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Cachexia is a late consequence of various diseases that is characterized by systemic muscle loss, with or without fat loss, leading to significant mortality. Multiple signaling pathways and molecules that increase catabolism, decrease anabolism, and interfere with muscle regeneration are activated. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play vital roles in cachexia muscle atrophy. This review mainly provides the mechanisms of specific ncRNAs to regulate muscle loss during cachexia and discusses the role of ncRNAs in cachectic biomarkers and novel therapeutic strategies that could offer new insights for clinical practice.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| | - Jiayi Zou
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (Q.H.)
| | - Qianhua He
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (Q.H.)
| | - Shuyi Xia
- Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
| | - Qili Xiao
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Q.X.); (S.Z.)
| | - Ruoxi Du
- Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
| | - Shengmei Zhou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Q.X.); (S.Z.)
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| |
Collapse
|
6
|
Silva ED, Pereira-Sousa D, Ribeiro-Costa F, Cerqueira R, Enguita FJ, Gomes RN, Dias-Ferreira J, Pereira C, Castanheira A, Pinto-do-Ó P, Leite-Moreira AF, Nascimento DS. Pericardial Fluid Accumulates microRNAs That Regulate Heart Fibrosis after Myocardial Infarction. Int J Mol Sci 2024; 25:8329. [PMID: 39125899 PMCID: PMC11313565 DOI: 10.3390/ijms25158329] [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: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Pericardial fluid (PF) has been suggested as a reservoir of molecular targets that can be modulated for efficient repair after myocardial infarction (MI). Here, we set out to address the content of this biofluid after MI, namely in terms of microRNAs (miRs) that are important modulators of the cardiac pathological response. PF was collected during coronary artery bypass grafting (CABG) from two MI cohorts, patients with non-ST-segment elevation MI (NSTEMI) and patients with ST-segment elevation MI (STEMI), and a control group composed of patients with stable angina and without previous history of MI. The PF miR content was analyzed by small RNA sequencing, and its biological effect was assessed on human cardiac fibroblasts. PF accumulates fibrotic and inflammatory molecules in STEMI patients, namely causing the soluble suppression of tumorigenicity 2 (ST-2), which inversely correlates with the left ventricle ejection fraction. Although the PF of the three patient groups induce similar levels of fibroblast-to-myofibroblast activation in vitro, RNA sequencing revealed that PF from STEMI patients is particularly enriched not only in pro-fibrotic miRs but also anti-fibrotic miRs. Among those, miR-22-3p was herein found to inhibit TGF-β-induced human cardiac fibroblast activation in vitro. PF constitutes an attractive source for screening diagnostic/prognostic miRs and for unveiling novel therapeutic targets in cardiac fibrosis.
Collapse
Affiliation(s)
- Elsa D. Silva
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Daniel Pereira-Sousa
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Center for Translational Medicine (CTM), International Clinical Research Centre (ICRC), St. Anne’s Hospital, 60200 Brno, Czech Republic
- Department of Biomedical Sciences, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Francisco Ribeiro-Costa
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Rui Cerqueira
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, 4150-180 Porto, Portugal; (R.C.)
| | - Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Rita N. Gomes
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - João Dias-Ferreira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Cassilda Pereira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO)/Health Research Network (RISE-Health), ESS, Polytechnic of Porto, 4200-072 Porto, Portugal
- Chemical and Biomolecular Sciences, School of Health (ESS), Polytechnic of Porto, 4200-465 Porto, Portugal
| | - Ana Castanheira
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
- INL—International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal
| | - Perpétua Pinto-do-Ó
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Adelino F. Leite-Moreira
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, 4150-180 Porto, Portugal; (R.C.)
| | - Diana S. Nascimento
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (E.D.S.); (F.R.-C.); (R.N.G.); (J.D.-F.); (C.P.); (A.C.); (P.P.-d.-Ó.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
7
|
Giardinelli S, Meliota G, Mentino D, D’Amato G, Faienza MF. Molecular Basis of Cardiomyopathies in Type 2 Diabetes. Int J Mol Sci 2024; 25:8280. [PMID: 39125850 PMCID: PMC11313011 DOI: 10.3390/ijms25158280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Diabetic cardiomyopathy (DbCM) is a common complication in individuals with type 2 diabetes mellitus (T2DM), and its exact pathogenesis is still debated. It was hypothesized that chronic hyperglycemia and insulin resistance activate critical cellular pathways that are responsible for numerous functional and anatomical perturbations in the heart. Interstitial inflammation, oxidative stress, myocardial apoptosis, mitochondria dysfunction, defective cardiac metabolism, cardiac remodeling, hypertrophy and fibrosis with consequent impaired contractility are the most common mechanisms implicated. Epigenetic changes also have an emerging role in the regulation of these crucial pathways. The aim of this review was to highlight the increasing knowledge on the molecular mechanisms of DbCM and the new therapies targeting specific pathways.
Collapse
Affiliation(s)
- Silvia Giardinelli
- Department of Medical Sciences, Pediatrics, University of Ferrara, 44121 Ferrara, Italy;
| | - Giovanni Meliota
- Department of Pediatric Cardiology, Giovanni XXIII Pediatric Hospital, 70126 Bari, Italy;
| | - Donatella Mentino
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Gabriele D’Amato
- Neonatal Intensive Care Unit, Di Venere Hospital, 70012 Bari, Italy;
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| |
Collapse
|
8
|
Yao X, Huang X, Chen J, Lin W, Tian J. Roles of non-coding RNA in diabetic cardiomyopathy. Cardiovasc Diabetol 2024; 23:227. [PMID: 38951895 PMCID: PMC11218407 DOI: 10.1186/s12933-024-02252-9] [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: 02/28/2024] [Accepted: 04/26/2024] [Indexed: 07/03/2024] Open
Abstract
In recent years, the incidence of diabetes has been increasing rapidly, posing a serious threat to human health. Diabetic cardiomyopathy (DCM) is characterized by cardiomyocyte hypertrophy, myocardial fibrosis, apoptosis, ventricular remodeling, and cardiac dysfunction in individuals with diabetes, ultimately leading to heart failure and mortality. However, the underlying mechanisms contributing to DCM remain incompletely understood. With advancements in molecular biology technology, accumulating evidence has shown that numerous non-coding RNAs (ncRNAs) crucial roles in the development and progression of DCM. This review aims to summarize recent studies on the involvement of three types of ncRNAs (micro RNA, long ncRNA and circular RNA) in the pathophysiology of DCM, with the goal of providing innovative strategies for the prevention and treatment of DCM.
Collapse
Affiliation(s)
- Xi Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xinyue Huang
- International School of Medicine, International Institutes of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Weiqiang Lin
- International School of Medicine, International Institutes of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China.
| | - Jingyan Tian
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Clinical Trials Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
9
|
Tudurachi BS, Anghel L, Tudurachi A, Sascău RA, Zanfirescu RL, Stătescu C. Unraveling the Cardiac Matrix: From Diabetes to Heart Failure, Exploring Pathways and Potential Medications. Biomedicines 2024; 12:1314. [PMID: 38927520 PMCID: PMC11201699 DOI: 10.3390/biomedicines12061314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/08/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Myocardial infarction (MI) often leads to heart failure (HF) through acute or chronic maladaptive remodeling processes. This establishes coronary artery disease (CAD) and HF as significant contributors to cardiovascular illness and death. Therefore, treatment strategies for patients with CAD primarily focus on preventing MI and lessening the impact of HF after an MI event. Myocardial fibrosis, characterized by abnormal extracellular matrix (ECM) deposition, is central to cardiac remodeling. Understanding these processes is key to identifying new treatment targets. Recent studies highlight SGLT2 inhibitors (SGLT2i) and GLP-1 receptor agonists (GLP1-RAs) as favorable options in managing type 2 diabetes due to their low hypoglycemic risk and cardiovascular benefits. This review explores inflammation's role in cardiac fibrosis and evaluates emerging anti-diabetic medications' effectiveness, such as SGLT2i, GLP1-RAs, and dipeptidyl peptidase-4 inhibitors (DPP4i), in preventing fibrosis in patients with diabetes post-acute MI. Recent studies were analyzed to identify effective medications in reducing fibrosis risk in these patients. By addressing these areas, we can advance our understanding of the potential benefits of anti-diabetic medications in reducing cardiac fibrosis post-MI and improve patient outcomes in individuals with diabetes at risk of HF.
Collapse
Affiliation(s)
- Bogdan-Sorin Tudurachi
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (B.-S.T.); (R.A.S.); (C.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iasi, Romania; (A.T.); (R.-L.Z.)
| | - Larisa Anghel
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (B.-S.T.); (R.A.S.); (C.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iasi, Romania; (A.T.); (R.-L.Z.)
| | - Andreea Tudurachi
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iasi, Romania; (A.T.); (R.-L.Z.)
| | - Radu Andy Sascău
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (B.-S.T.); (R.A.S.); (C.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iasi, Romania; (A.T.); (R.-L.Z.)
| | - Răzvan-Liviu Zanfirescu
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iasi, Romania; (A.T.); (R.-L.Z.)
- Physiology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania
| | - Cristian Stătescu
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700503 Iasi, Romania; (B.-S.T.); (R.A.S.); (C.S.)
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I. M. Georgescu”, 700503 Iasi, Romania; (A.T.); (R.-L.Z.)
| |
Collapse
|
10
|
Aghaei-Zarch SM. Crosstalk between MiRNAs/lncRNAs and PI3K/AKT signaling pathway in diabetes mellitus: Mechanistic and therapeutic perspectives. Noncoding RNA Res 2024; 9:486-507. [PMID: 38511053 PMCID: PMC10950585 DOI: 10.1016/j.ncrna.2024.01.005] [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: 10/03/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 03/22/2024] Open
Abstract
Diabetes as a fastest growing diseases worldwide is characterized by elevated blood glucose levels. There's an enormous financial burden associated with this endocrine disorder, with unequal access to health care between developed and developing countries. PI3Ks (phosphoinositide 3-kinases) have been demonstrated to be crucial for glucose homeostasis, and malfunctioning of these molecules can contribute to an increase in glucose serum levels, the main pathophysiological feature of diabetes. Additionally, recent evidence suggests that miRNAs and lncRNAs are reciprocally interacting with this signaling pathway. It is therefore evident that abnormal regulation of miRNAs/lncRNAs in the lncRNAs/miRNAs/PI3K/AKT axis is related to clinicopathological characteristics and plays a crucial role in the regulation of biological processes. It has therefore been attempted in this review to describe the interaction between PI3K/AKT signaling pathway and various miRNAs/lncRNAs and their importance in DM biology. We also presented the clinical applications of PI3K/AKT-related ncRNAs/herbal medicine in patients with DM.
Collapse
Affiliation(s)
- Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Yuan L, Wang T, Duan J, Zhou J, Li N, Li G, Zhou H. Expression Profiles and Bioinformatic Analysis of Circular RNAs in Db/Db Mice with Cardiac Fibrosis. Diabetes Metab Syndr Obes 2024; 17:2107-2120. [PMID: 38799279 PMCID: PMC11128257 DOI: 10.2147/dmso.s465588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Cardiac fibrosis is one of the important causes of heart failure and death in diabetic cardiomyopathy (DCM) patients. Circular RNAs (circRNAs) are covalently closed RNA molecules in eukaryotes and have high stability. Their role in myocardial fibrosis with diabetic cardiomyopathy (DCM) remain to be fully elucidated. This study aimed to understand the expression profiles of circRNAs in myocardial fibrosis with DCM, exploring the possible biomarkers and therapeutic targets for DCM. Methods At 21 weeks of age, db/db mice established the type 2 DCM model measured by echocardiography, and the cardiac tissue was extracted for Hematoxylin-eosin, Masson's trichrome staining, and transmission electron microscopy. Subsequently, the expression profile of circRNAs in myocardial fibrosis of db/db mice was constructed using microarray hybridization and verified by real-time quantitative polymerase chain reaction. A circRNA-microRNA-messenger RNA coexpression network was constructed, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were done. Results Compared with normal control mice, db/db mice had 77 upregulated circRNAs and 135 downregulated circRNAs in their chromosomes (fold change ≥1.5, P ≤ 0.05). Moreover, the enrichment analysis of circRNA host genes showed that these differentially expressed circRNAs were mainly involved in mitogen-activated protein kinase signaling pathways. CircPHF20L1, circCLASP1, and circSLC8A1 were the key circRNAs. Moreover, circCLASP1/miR-182-5p/Wnt7a, circSLC8A1/miR-29b-1-5p/Col12a1, and most especially circPHF20L1/miR-29a-3p/Col6a2 might be three novel axes in the development of myocardial fibrosis in DCM. Conclusion The findings will provide some novel circRNAs and molecular pathways for the prevention or clinical treatment of DCM through intervention with specific circRNAs.
Collapse
Affiliation(s)
- Lingling Yuan
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Ting Wang
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Jinsheng Duan
- Department of Cardiology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Jing Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Na Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Guizhi Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Hong Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| |
Collapse
|
12
|
Ma X, Mei S, Wuyun Q, Zhou L, Sun D, Yan J. Epigenetics in diabetic cardiomyopathy. Clin Epigenetics 2024; 16:52. [PMID: 38581056 PMCID: PMC10996175 DOI: 10.1186/s13148-024-01667-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a critical complication that poses a significant threat to the health of patients with diabetes. The intricate pathological mechanisms of DCM cause diastolic dysfunction, followed by impaired systolic function in the late stages. Accumulating researches have revealed the association between DCM and various epigenetic regulatory mechanisms, including DNA methylation, histone modifications, non-coding RNAs, and other epigenetic molecules. Recently, a profound understanding of epigenetics in the pathophysiology of DCM has been broadened owing to advanced high-throughput technologies, which assist in developing potential therapeutic strategies. In this review, we briefly introduce the epigenetics regulation and update the relevant progress in DCM. We propose the role of epigenetic factors and non-coding RNAs (ncRNAs) as potential biomarkers and drugs in DCM diagnosis and treatment, providing a new perspective and understanding of epigenomics in DCM.
Collapse
Affiliation(s)
- Xiaozhu Ma
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Shuai Mei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Qidamugai Wuyun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Li Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dating Sun
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
13
|
Li Z, Deng X, Lan Y. Identification of a potentially functional circRNA-miRNA-mRNA regulatory network in type 2 diabetes mellitus by integrated microarray analysis. Minerva Endocrinol (Torino) 2024; 49:33-46. [PMID: 33792237 DOI: 10.23736/s2724-6507.21.03370-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) function as miRNA sponges by adsorbing microRNAs (miRNAs), thereby regulating messenger RNA (mRNA) expression. The circRNA-miRNA-mRNA regulatory network associated with type 2 diabetes mellitus (T2DM) has rarely been explored. A circRNA-miRNA-mRNA regulatory network associated with T2DM was established to help deepen our understanding of the molecular mechanism of and therapeutic targets for T2DM. METHODS Differentially expressed circRNAs (DEcircRNAs), miRNAs (DEmiRNAs), and mRNAs (DEmRNAs) were derived from the Gene Expression Omnibus (GEO) microarray datasets GSE114248, GSE51674 and GSE95849, respectively. A circRNA-miRNA-mRNA regulatory network associated with T2DM and its subnetwork were constructed. The hub genes were screened using a protein-protein interaction (PPI) network. Finally, a hub gene-related network was constructed. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed. RESULTS The circRNA-miRNA-mRNA network included 9 circRNAs, 24 miRNAs and 320 mRNAs. When four key circRNAs (circMYO9B, circGRAMD1B, circTHAP4 and circTMC7) were chosen, the subnetwork contained 4 circRNAs, 18 miRNAs and 307 mRNAs. Afterwards, 8 hub genes (SIRT1, GNG7, KDR, FOS, SIN3B, STAT1, SP1, and MAPK3) were extracted from the PPI network. GO and KEGG pathway analyses revealed that the network might be involved in oxidative stress responses, regulation of inflammation, neovascularization, endocrine and cancer-related processes, etc. CONCLUSIONS A circRNA-miRNA-hub gene regulatory network was constructed, and the potential functions of the hub genes were analyzed. Four important circRNAs (circMYO9B, circGRAMD1B, circTHAP4 and circTMC7) might be involved in the occurrence and development of T2DM, and this finding provides new insight into the molecular mechanism of and therapeutic targets for T2DM and its complications. Future studies are needed to validate the sponge effects and mechanisms of these 4 circRNAs.
Collapse
Affiliation(s)
- Zijing Li
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Provincial Clinical Research Center of Diabetes Mellitus and its Chronic Complications, Guangzhou, China
- Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaowen Deng
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Provincial Clinical Research Center of Diabetes Mellitus and its Chronic Complications, Guangzhou, China
- Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuqing Lan
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China -
- Provincial Clinical Research Center of Diabetes Mellitus and its Chronic Complications, Guangzhou, China
- Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Zhao L, Tang P, Lin Y, Du M, Li H, Jiang L, Xu H, Sun H, Han J, Sun Z, Xu R, Lou H, Chen Z, Kopylov P, Liu X, Zhang Y. MiR-203 improves cardiac dysfunction by targeting PARP1-NAD + axis in aging murine. Aging Cell 2024; 23:e14063. [PMID: 38098220 PMCID: PMC10928583 DOI: 10.1111/acel.14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/08/2023] [Accepted: 11/26/2023] [Indexed: 03/13/2024] Open
Abstract
Heart aging is a prevalent cause of cardiovascular diseases among the elderly. NAD+ depletion is a hallmark feature of aging heart, however, the molecular mechanisms that affect NAD+ depletion remain unclear. In this study, we identified microRNA-203 (miR-203) as a senescence-associated microRNA that regulates NAD+ homeostasis. We found that the blood miR-203 level negatively correlated with human age and its expression significantly decreased in the hearts of aged mice and senescent cardiomyocytes. Transgenic mice with overexpressed miR-203 (TgN (miR-203)) showed resistance to aging-induced cardiac diastolic dysfunction, cardiac remodeling, and myocardial senescence. At the cellular level, overexpression of miR-203 significantly prevented D-gal-induced cardiomyocyte senescence and mitochondrial damage, while miR-203 knockdown aggravated these effects. Mechanistically, miR-203 inhibited PARP1 expression by targeting its 3'UTR, which helped to reduce NAD+ depletion and improve mitochondrial function and cell senescence. Overall, our study first identified miR-203 as a genetic tool for anti-heart aging by restoring NAD+ function in cardiomyocytes.
Collapse
Affiliation(s)
- Limin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Pingping Tang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuan Lin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Menghan Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Huimin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lintong Jiang
- Department of Pharmacy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Henghui Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Heyang Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jingjing Han
- Department of Pharmacy, Caoxian People's Hospital, Heze, China
| | - Zeqi Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Run Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Han Lou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhouxiu Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Philipp Kopylov
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Xin Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Yong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, China
| |
Collapse
|
15
|
Entezari M, Soltani BM, Sadeghizadeh M. MicroRNA-203a inhibits breast cancer progression through the PI3K/Akt and Wnt pathways. Sci Rep 2024; 14:4715. [PMID: 38413784 PMCID: PMC10899204 DOI: 10.1038/s41598-024-52940-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/25/2024] [Indexed: 02/29/2024] Open
Abstract
MicroRNA expression in breast cancer (BC) is explored both as a potential biomarker and for therapeutic purposes. Recent studies have revealed that miR-203a-3p is involved in BC, and importantly contributes to BC chemotherapy responses; however, the regulatory pathways of miR-203a in BC remain elusive. Hence, we aimed to investigate the miR-203a regulatory mechanisms and their potential functions in the progress of BC. To this end, the miR-203a potential involving pathways was predicted by databases analyzing its target genes. The relations between miR-203a, the phosphatidylinositol 3'-kinase (PI3K)-Akt, and Wnt signaling pathways were mechanistically investigated. Our results revealed that miR-203a inhibited the activation of the PI3K/Akt and Wnt pathways and reduced its downstream cell cycle signals, including Cyclin D1 and c-Myc. Moreover, the overexpression of miR-203a drastically arrested the cell cycle at subG1 and G1 phases, decreased the viability, proliferation, and migration, and increased apoptosis of BC cells. Therefore, miR-203a-3p may be considered a tumor suppressor factor and a potential biomarker or therapeutic target for BC.
Collapse
Affiliation(s)
- Maryam Entezari
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Bahram M Soltani
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran.
| |
Collapse
|
16
|
Xue B, Kadeerhan G, Sun LB, Chen YQ, Hu XF, Zhang ZK, Wang DW. Circulating exosomal miR-16-5p and let-7e-5p are associated with bladder fibrosis of diabetic cystopathy. Sci Rep 2024; 14:837. [PMID: 38191820 PMCID: PMC10774280 DOI: 10.1038/s41598-024-51451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Diabetic cystopathy (DCP) is a prevalent etiology of bladder dysfunction in individuals with longstanding diabetes, frequently leading to bladder interstitial fibrosis. Research investigating the initial pathological alterations of DCP is notably scarce. To comprehend the development of fibrosis and find effective biomarkers for its diagnosis, we prepared streptozotocin-induced long-term diabetic SD rats exhibiting a type 1 diabetes phenotype and bladder fibrosis in histology detection. After observing myofibroblast differentiation from rats' primary bladder fibroblasts with immunofluorescence, we isolated fibroblasts derived exosomes and performed exosomal miRNA sequencing. The co-differentially expressed miRNAs (DEMis) (miR-16-5p and let-7e-5p) were screened through a joint analysis of diabetic rats and long-term patients' plasma data (GES97123) downloaded from the GEO database. Then two co-DEMis were validated by quantitative PCR on exosomes derived from diabetic rats' plasma. Following with a series of analysis, including target mRNAs and transcription factors (TFs) prediction, hubgenes identification, protein-protein interaction (PPI) network construction and gene enrichment analysis, a miRNA-mediated genetic regulatory network consisting of two miRNAs, nine TFs, and thirty target mRNAs were identified in relation to fibrotic processes. Thus, circulating exosomal miR-16-5p and let-7e-5p are associated with bladder fibrosis of DCP, and the crucial genes in regulatory network might hold immense significance in studying the pathogenesis and molecular mechanisms of fibrosis, which deserves further exploration.
Collapse
Affiliation(s)
- Bo Xue
- Shanxi Medical University, Taiyuan, 030001, China
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Gaohaer Kadeerhan
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Li-Bin Sun
- Shanxi Medical University, Taiyuan, 030001, China
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | | | - Xiao-Feng Hu
- Shanxi Medical University, Taiyuan, 030001, China
| | | | - Dong-Wen Wang
- Shanxi Medical University, Taiyuan, 030001, China.
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China.
| |
Collapse
|
17
|
Zhao S, Liu H, Wang H, He X, Tang J, Qi S, Yang R, Xie J. Inhibition of phosphatidylinositol 3-kinase catalytic subunit alpha by miR-203a-3p reduces hypertrophic scar formation via phosphatidylinositol 3-kinase/AKT/mTOR signaling pathway. BURNS & TRAUMA 2024; 12:tkad048. [PMID: 38179473 PMCID: PMC10762504 DOI: 10.1093/burnst/tkad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 01/06/2024]
Abstract
Background Hypertrophic scar (HS) is a common fibroproliferative skin disease that currently has no truly effective therapy. Given the importance of phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) in hypertrophic scar formation, the development of therapeutic strategies for endogenous inhibitors against PIK3CA is of great interest. Here, we explored the molecular mechanisms underlying the protective effects of miR-203a-3p (PIK3CA inhibitor) against excessive scar. Methods Bioinformatic analysis, immunohistochemistry, immunofluorescence, miRNA screening and fluorescence in situ hybridization assays were used to identify the possible pathways and target molecules mediating HS formation. A series of in vitro and in vivo experiments were used to clarify the role of PIK3CA and miR-203a-3p in HS. Mechanistically, transcriptomic sequencing, immunoblotting, dual-luciferase assay and rescue experiments were executed. Results Herein, we found that PIK3CA and the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway were upregulated in scar tissues and positively correlated with fibrosis. We then identified miR-203a-3p as the most suitable endogenous inhibitor of PIK3CA. miR-203a-3p suppressed the proliferation, migration, collagen synthesis and contractility as well as the transdifferentiation of fibroblasts into myofibroblasts in vitro, and improved the morphology and histology of scars in vivo. Mechanistically, miR-203a-3p attenuated fibrosis by inactivating the PI3K/AKT/mTOR pathway by directly targeting PIK3CA. Conclusions PIK3CA and the PI3K/AKT/mTOR pathway are actively involved in scar fibrosis and miR-203a-3p might serve as a potential strategy for hypertrophic scar therapy through targeting PIK3CA and inactivating the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Shixin Zhao
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Hengdeng Liu
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Hanwen Wang
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Xuefeng He
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Jinming Tang
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Shaohai Qi
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Julin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| |
Collapse
|
18
|
Wu K, Guo S, Zhang J, Wen D, Zhang L, Zhu M, Wang X, Li X, Chen Z, Lin F. Mechanism of Action of NvZhen ErXian HeJi in Ovariectomized Rats with Myocardial Infarction based on Network Pharmacology. Curr Pharm Des 2024; 30:3116-3130. [PMID: 39161145 DOI: 10.2174/0113816128308824240719093114] [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: 02/06/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE NvZhen ErXian HeJi (NZEXHJ) is used to treat perimenopausal syndrome (PS), but its effect on perimenopausal coronary heart disease is unclear. Furthermore, the aim of this research is to study the effect of NZEXHJ on perimenopausal coronary heart disease (PMCHD) in a rat model based on a network pharmacology approach. MATERIALS AND METHODS Based on network pharmacological analysis combined with molecular docking, we predicted the potential therapeutic target and pharmacological mechanism of NZEXHJ in the treatment of PMCHD. We used an ovariectomized rat (OVR) model to understand the effect of NZEXHJ on myocardial injury and further verified the target of NZEXHJ in the intervention of PMCHD. RESULTS We selected 52 active components of NZEXHJ against PMCHD and an intersection of their targets on network pharmacology, to which SCN5A, SER1, AR, and PGR were significantly correlated. The protein- protein interaction network revealed CASP3, CXCL8, IL6, MAPK1, TNF, TP53, and VEGFA in the treatment of PMCHD with NZEXHJ. Kaempferol, luteolin, and mistletoe presented good affinity towards the aforementioned targets by Molecular docking NZEXHJ exerted protecting cardiomyocytes for OVR. The mechanism was related to a reduction in the expression levels of the CXCL8, TNF, and regulating PI3K-Akt signaling pathways. CONCLUSION This study reveals the potential multi-component, multi-target, and multi-pathway pharmacological effects of NZEXHJ and predicts its protection against myocardial infarction in ovariectomized rats through the PI3K Akt pathway, providing a theoretical basis for the treatment of PMCHD.
Collapse
Affiliation(s)
- Kai Wu
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shuxun Guo
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jie Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Desong Wen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Linli Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Mingyang Zhu
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Engineering Research Center for Clinical Treatment of Coronary Heart Disease, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiulong Wang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Engineering Research Center for Clinical Treatment of Coronary Heart Disease, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xuefang Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Engineering Research Center for Clinical Treatment of Coronary Heart Disease, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhigang Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Engineering Research Center for Clinical Treatment of Coronary Heart Disease, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Fei Lin
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Heart Center of Xinxiang Medical University, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Engineering Research Center for Clinical Treatment of Coronary Heart Disease, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
19
|
Beylerli O, Ju J, Beilerli A, Gareev I, Shumadalova A, Ilyasova T, Bai Y, Yang B. The roles of long noncoding RNAs in atrial fibrillation. Noncoding RNA Res 2023; 8:542-549. [PMID: 37602317 PMCID: PMC10432912 DOI: 10.1016/j.ncrna.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 08/22/2023] Open
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia that often occurs in patients with structural heart disease and is a significant cause of morbidity and mortality in clinical settings. AF is typically associated with significant changes of both the structure of the atria and the cardiac conduction system. AF can result in reduced heart function, heart failure, and various other complications. Current drug therapy for AF patients is often ineffective and may have adverse effects. Radiofrequency ablation is more effective than traditional drug therapy, but this invasive procedure carries potential risks and may lead to postoperative recurrence, limiting the clinical benefits to some extent. Therefore, in-depth research into the molecular mechanisms of AF and exploration of new treatment strategies based on research findings are prerequisites for improving the treatment of AF and the associated cardiac conditions. Long noncoding RNAs (lncRNAs) are a new class of noncoding RNA (ncRNAs) with a length exceeding 200 nt, which regulate gene expression at multiple levels. Increasing evidence suggests that lncRNAs participate in many pathological processes of AF initiation, development, and maintenance, such as structural remodeling, electrical remodeling, renin-angiotensin system anomalies, and intracellular calcium deregulation s. LncRNAs that play key roles in structural and electrical remodeling may become molecular markers and targets for AF diagnosis and treatment, respectively, while lncRNAs critical to autonomic nervous system remodeling may bring new insights into the prognosis and recurrence of AF. This review article provides a synopsis on the up-to-date research findings relevant to the roles of lncRNAs in AF.
Collapse
Affiliation(s)
- Ozal Beylerli
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jiaming Ju
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | - Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, 450008, Russia
| | - Alina Shumadalova
- Department of General Chemistry, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, 450008, Russia
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Yunlong Bai
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
20
|
Wang T, Yuan L, Chen Y, Wang J, Li N, Zhou H. Expression profiles and bioinformatic analysis of microRNAs in myocardium of diabetic cardiomyopathy mice. Genes Genomics 2023; 45:1003-1011. [PMID: 37253907 DOI: 10.1007/s13258-023-01403-8] [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: 09/20/2022] [Accepted: 05/15/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) can regulate expression of target genes at post transcriptional level, and mediate the pathophysiological process of many diseases. OBJECTIVE The study will illuminate the miRNA expression profiles of diabetic cardiomyopathy (DCM), seeking probable biomarkers of DCM at early stage and determining a target for the treatment of DCM. METHODS Db/db mice were used as an animal model of type 2 diabetes mellitus. At 22 weeks of age, cardiac function was evaluated by echocardiography, and the structural changes in myocardium were evaluated by HE staining and TEM. The miRNA expression profiles were detected using miRNA sequencing and differentially expressed miRNAs were validated by real-time PCR. Bioinformatic analysis was used to analyze target genes of these miRNAs and relevant pathways in DCM. RESULTS The results showed that 40 miRNAs were differentially expressed, including 28 upregulated miRNAs and 12 downregulated miRNAs. GO and KEGG pathway analysis showed that the target genes of up-regulated miRNAs were involved in 66 pathways, including Wnt, p53 and calcium signaling pathways, as well as FOXO and apoptosis signaling pathways, etc. The target genes of down-regulated miRNAs were involved in 68 pathways, including mitophagy, Ras and MAPK signaling pathways, etc. Moreover, some differentially expressed miRNAs were found in myocardium of DCM for the first time, such as miR-7225-5p, miR-696, miR-3470a, miR-3470b, miR-6240, miR-6538, miR-5128, miR-1195, miR-203-3p and miR-330-5p. CONCLUSIONS It is hoped that a few novel molecular pathways or targets of treatment for DCM would be found through understanding the expression features of miRNAs in diabetic myocardium.
Collapse
Affiliation(s)
- Ting Wang
- Department of Endocrinology, The second Hospital of Hebei Medical University, NO.215 Heping West Road, Xinhua District, 050051, Shijiazhuang, Hebei, PR China
| | - Lingling Yuan
- Department of Endocrinology, The second Hospital of Hebei Medical University, NO.215 Heping West Road, Xinhua District, 050051, Shijiazhuang, Hebei, PR China
| | - Yanxia Chen
- Department of Endocrinology, The second Hospital of Hebei Medical University, NO.215 Heping West Road, Xinhua District, 050051, Shijiazhuang, Hebei, PR China
| | - Jing Wang
- Department of Endocrinology, The second Hospital of Hebei Medical University, NO.215 Heping West Road, Xinhua District, 050051, Shijiazhuang, Hebei, PR China
| | - Na Li
- Department of Endocrinology, The second Hospital of Hebei Medical University, NO.215 Heping West Road, Xinhua District, 050051, Shijiazhuang, Hebei, PR China
| | - Hong Zhou
- Department of Endocrinology, The second Hospital of Hebei Medical University, NO.215 Heping West Road, Xinhua District, 050051, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
21
|
Li Y, Du X, Li W, Jiang Q, Ye Y, Yang Y, Liu X, Zhao Y, Che X. Two genes related to apoptosis in the hepatopancreas of juvenile prawn, Macrobrachium nipponense: Molecular characterization and transcriptional response to nanoplastic exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 877:162863. [PMID: 36931509 DOI: 10.1016/j.scitotenv.2023.162863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 05/06/2023]
Abstract
Nanoplastics have been widely found in the global water environment, causing plastic pollution and affecting human beings and numerous organisms. Studies involving freshwater crustacean exposure to nanoplastics, however, are limited. In this study, juvenile prawns (Macrobrachium nipponense) were exposed to 75 nm polystyrene nanoplastics at different concentrations (0, 5, 10, 20, or 40 mg/L) for a 28-d chronic exposure experiment. To study the effects of exposure to nanoplastics on hepatopancreas cell apoptosis, C-Jun N-terminal kinase (JNK) and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) genes were selected, and hepatotoxic enzyme activities and Toll pathway- and apoptosis-related gene expression were determined. For the first time, full-length Mn-JNK and Mn-PIK3CA cDNAs were cloned from M. nipponense. Homologous comparisons showed that JNK and PIK3CA had conserved functional sequences. The apoptosis rate in the high-concentration nanoplastic group (40 mg/L) was significantly higher than in the low-concentration nanoplastic (5 mg/L) and control groups (0 mg/L). The alanine aminotransferase (ALT), aspartate aminotransferase (AST), glutamyl transpeptidase (GGT) and xanthine oxidase (XOD) enzyme activities in the hepatopancreas increased with exposure to higher concentrations of nanoplastics. In addition, the levels of apoptosis- and Toll pathway-related gene expression and JNK and PIK3CA gene expression were initially increased, then decreased with exposure to higher concentrations of nanoplastics. This study showed that polystyrene nanoplastics activate toll-related pathways leading to apoptosis and hepatopancreas damage, which provides theoretical support for future aquatic toxicological research.
Collapse
Affiliation(s)
- Yiming Li
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fisheries Sciences, Shanghai 200092, China
| | - Xinglin Du
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Wen Li
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Qichen Jiang
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing 210017, China
| | - Yucong Ye
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Ying Yang
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Xingguo Liu
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fisheries Sciences, Shanghai 200092, China
| | - Yunlong Zhao
- School of Life Science, East China Normal University, Shanghai 200241, China.
| | - Xuan Che
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fisheries Sciences, Shanghai 200092, China.
| |
Collapse
|
22
|
Macvanin MT, Gluvic Z, Radovanovic J, Essack M, Gao X, Isenovic ER. Diabetic cardiomyopathy: The role of microRNAs and long non-coding RNAs. Front Endocrinol (Lausanne) 2023; 14:1124613. [PMID: 36950696 PMCID: PMC10025540 DOI: 10.3389/fendo.2023.1124613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Diabetes mellitus (DM) is on the rise, necessitating the development of novel therapeutic and preventive strategies to mitigate the disease's debilitating effects. Diabetic cardiomyopathy (DCMP) is among the leading causes of morbidity and mortality in diabetic patients globally. DCMP manifests as cardiomyocyte hypertrophy, apoptosis, and myocardial interstitial fibrosis before progressing to heart failure. Evidence suggests that non-coding RNAs, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), regulate diabetic cardiomyopathy-related processes such as insulin resistance, cardiomyocyte apoptosis and inflammation, emphasizing their heart-protective effects. This paper reviewed the literature data from animal and human studies on the non-trivial roles of miRNAs and lncRNAs in the context of DCMP in diabetes and demonstrated their future potential in DCMP treatment in diabetic patients.
Collapse
Affiliation(s)
- Mirjana T. Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran Gluvic
- University Clinical-Hospital Centre Zemun-Belgrade, Clinic of Internal Medicine, Department of Endocrinology and Diabetes, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Magbubah Essack
- King Abdullah University of Science and Technology (KAUST), Computer, Electrical, and Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), Thuwal, Saudi Arabia
| | - Xin Gao
- King Abdullah University of Science and Technology (KAUST), Computer, Electrical, and Mathematical Sciences and Engineering (CEMSE) Division, Computational Bioscience Research Center (CBRC), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
23
|
Abdel Mageed SS, Doghish AS, Ismail A, El-Husseiny AA, Fawzi SF, Mahmoud AMA, El-Mahdy HA. The role of miRNAs in insulin resistance and diabetic macrovascular complications - A review. Int J Biol Macromol 2023; 230:123189. [PMID: 36623613 DOI: 10.1016/j.ijbiomac.2023.123189] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023]
Abstract
Diabetes is the most prevalent metabolic disturbance disease and has been regarded globally as one of the principal causes of mortality. Diabetes is accompanied by several macrovascular complications, including stroke, coronary artery disease (CAD), and cardiomyopathy as a consequence of atherosclerosis. The onset of type 2 diabetes is closely related to insulin resistance (IR). miRNAs have been linked to various metabolic processes, including glucose homeostasis, regulation of lipid metabolism, gluconeogenesis, adipogenesis, glucose transporter type 4 expression, insulin sensitivity, and signaling. Consequently, miRNA dysregulation mediates IR in some target organs, comprising liver, muscle, and adipose tissue. Moreover, miRNAs are crucial in developing diabetes and its associated macrovascular complications through their roles in several signaling pathways implicated in inflammation, apoptosis, cellular survival and migration, the proliferation of vascular smooth muscle cells, neurogenesis, angiogenesis, autophagy, oxidative stress, cardiac remodeling, and fibrosis. Therefore, the purpose of this review is to clarify the role of miRNAs in hepatic, muscle, and adipose tissue IR and explain their roles in the pathogenesis of macrovascular diabetic complications, including stroke, CAD, and cardiomyopathy. Also, explain their roles in gestational diabetes mellitus (GDM). Besides, this review discusses the latest updates on the alteration of miRNA expression in diabetic macrovascular complications.
Collapse
Affiliation(s)
- Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt
| | - Sylvia F Fawzi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Abdulla M A Mahmoud
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt
| |
Collapse
|
24
|
Zhou Y, Suo W, Zhang X, Yang Y, Zhao W, Li H, Ni Q. Targeting epigenetics in diabetic cardiomyopathy: Therapeutic potential of flavonoids. Biomed Pharmacother 2023; 157:114025. [PMID: 36399824 DOI: 10.1016/j.biopha.2022.114025] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/05/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
The pathophysiological mechanisms of diabetic cardiomyopathy have been extensively studied, but there is still a lack of effective prevention and treatment methods. The ability of flavonoids to protect the heart from diabetic cardiomyopathy has been extensively described. In recent years, epigenetics has received increasing attention from scholars in exploring the etiology and treatment of diabetes and its complications. DNA methylation, histone modifications and non-coding RNAs play key functions in the development, maintenance and progression of diabetic cardiomyopathy. Hence, prevention or reversal of the epigenetic alterations that have occurred during the development of diabetic cardiomyopathy may alleviate the personal and social burden of the disease. Flavonoids can be used as natural epigenetic modulators in alternative therapies for diabetic cardiomyopathy. In this review, we discuss the epigenetic effects of different flavonoid subtypes in diabetic cardiomyopathy and summarize the evidence from preclinical and clinical studies that already exist. However, limited research is available on the potential beneficial effects of flavonoids on the epigenetics of diabetic cardiomyopathy. In the future, clinical trials in which different flavonoids exert their antidiabetic and cardioprotective effects through various epigenetic mechanisms should be further explored.
Collapse
Affiliation(s)
- Yutong Zhou
- Guang'an Men Hospital, China Academy of Chinese Medicine, Beijing 100053, China
| | - Wendong Suo
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xinai Zhang
- Guang'an Men Hospital, China Academy of Chinese Medicine, Beijing 100053, China
| | - Yanan Yang
- Guang'an Men Hospital, China Academy of Chinese Medicine, Beijing 100053, China
| | - Weizhe Zhao
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing 100105, China
| | - Hong Li
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Qing Ni
- Guang'an Men Hospital, China Academy of Chinese Medicine, Beijing 100053, China.
| |
Collapse
|
25
|
Chen H, Lin Y, Zeng L, Liu S. Elucidating the mechanism of Hongjinshen decoction in the treatment of pulmonary fibrosis based on network pharmacology and molecular docking. Medicine (Baltimore) 2022; 101:e32323. [PMID: 36595795 PMCID: PMC9794259 DOI: 10.1097/md.0000000000032323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND To explore the mechanism of compound Hongginshen decoction in improving pulmonary fibrosis based on network pharmacology. METHODS The active components and targets of ginseng and Salvia miltiorrhiza were screened from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database. The chemical components of Rhodiola, Ophiopogon japonicus, and Dendrobium were screened using the Traditional Chinese Medicine Integrated Database (TCMID), and the target compounds were predicted by the Swisstargets method. The related target genes of pulmonary fiber (PF) were screened by the Genecards database and the National Center of Biotechnology Information (NCBI) database. The protein-protein interaction network was drawn using the string database and Cytoscape software, and the network topology was analyzed. Then, using R3.6.3 software, biological processes, molecular function, cell component enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were carried out on the common targets of drugs and diseases. The network diagram of the "traditional Chinese medicine composition disease target" of Compound Hongginshen Decoction was constructed and analyzed with the software of Cytoscape 3.6.1. RESULTS We identified 159 active components and 2820 targets in Compound Hongginshen Decoction, and 2680 targets in pulmonary fibrosis. A total of 343 common targets were obtained by the intersection of drug targets and disease targets. protein-protein interaction protein interaction network analysis showed that PIK3CA, PIK3R1, MAPK1, SRC, AKT1, and so on may be the core targets of the compound Hongjingshen recipe in the treatment of pulmonary fibrosis. Gene Ontology (GO) enrichment analysis identified 3463 items, and KEGG pathway enrichment analysis identified 181 related signaling pathways, including the PI3K-Akt signaling pathway, HCMV pathway, Hb pathway, PGs pathway, and KSHV signaling pathway. CONCLUSION Compound Hongginshen Decoction has the characteristics of a multichannel and multitargeted effect in the treatment of pulmonary fibrosis. Radix Ophiopogonis and Dendrobium officinale play a key role in the treatment of pulmonary fibrosis. The whole compound prescription may play a therapeutic role by affecting cell metabolism, being anti-inflammatory, regulating the immune system, promoting angiogenesis, and improving anaerobic metabolism.
Collapse
Affiliation(s)
- Haixu Chen
- Department of Basic Medicine, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Yu Lin
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Department of Clinical and Medical Technology, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China
| | - Lianlin Zeng
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- * Correspondence: Shiwei Liu, Department of Rehabilitation Medicine, Zigong No.4 People’s Hospital, Sichuan 643000, China (e-mail: ) and Lianlin Zeng, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China (e-mail: )
| | - Shiwei Liu
- Department of Rehabilitation Medicine, Zigong No.4 People’s Hospital, Sichuan, China
- * Correspondence: Shiwei Liu, Department of Rehabilitation Medicine, Zigong No.4 People’s Hospital, Sichuan 643000, China (e-mail: ) and Lianlin Zeng, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China (e-mail: )
| |
Collapse
|
26
|
Zeng F, Xu Y, Tang C, Yan Z, Wei C. Integrated bioinformatics and in silico approaches reveal the biological targets and molecular mechanisms of 1,25-dihydroxyvitamin D against COVID-19 and diabetes mellitus. Front Nutr 2022; 9:1060095. [DOI: 10.3389/fnut.2022.1060095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) and diabetes mellitus (DM) are two major diseases threatening human health. The susceptibility of DM patients to COVID-19 and their worse outcomes have forced us to explore efficient routes to combat COVID-19/DM. As the most active form of Vitamin D, 1,25-dihydroxyvitamin D (1,25(OH)2D) has been shown a beneficial effect in the treatment of COVID-19/DM. However, the anti-COVID-19/DM mechanisms of 1,25(OH)2D remain unclear. In this study, an approach combining network pharmacology and molecular docking was performed to reveal the potential hub target genes and underlying mechanisms of 1,25(OH)2D in the treatment of COVID-19/DM. The hub targets and interaction pathways related to 1,25(OH)2D were identified by integrating the key 1,25(OH)2D-target-signaling pathway-COVID-19/DM networks. Fifteen hub targets of 1,25(OH)2D against COVID-19DM were determined, including EGFR, PIK3R1, PIK3CA, STAT3, MAPK1, ESR1, HSP90AA1, LCK, MTOR, IGF1, AR, NFKB1, PIK3CB, PTPN1, and MAPK14. An enrichment analysis of the hub targets further revealed that the effect of 1,25(OH)2D against COVID-19/DM involved multiple biological processes, cellular components, molecular functions and biological signaling pathways. Molecular docking disclosed that 1,25(OH)2D docked nicely with the hub target proteins, including EGFR, PIK3R1, and PIK3CA. These findings suggested that the potential mechanisms of 1,25(OH)2D against COVID-19/DM may be related to multiple biological targets and biological signaling pathways.
Collapse
|
27
|
Li J, Wang N, Wen X, Huang LY, Cui RQ, Zhang J. Serum miRNA-203 as a Novel Biomarker for the Early Prediction of Acute ST-elevation Myocardial Infarction. J Cardiovasc Transl Res 2022; 15:1406-1413. [PMID: 35507256 DOI: 10.1007/s12265-022-10269-2] [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: 02/26/2022] [Accepted: 04/27/2022] [Indexed: 12/16/2022]
Abstract
Existing markers of myocardial infarction have limited diagnostic value for infarction, so it is necessary to identify new markers of infarction. To study the predictive value of serum miRNA-203 for acute ST-elevation myocardial infarction. Seventy patients with STEMI who were diagnosed in Hefei Second People's Hospital from December 2020 to December 2021 were selected, and 35 patients with transient chest pain who were hospitalized for other diseases in the Cardiology Department of our hospital during the same period were selected as the control group. The sera of the two groups of patients were collected, and a miRNA-203 semiquantitative experiment was performed. The miRNA-203 level in the STEMI group was higher than that in the control group. The AUC area of miRNA-203 in predicting STEMI was 0.912. Logistic regression analysis showed that miRNA-203 and white blood cell counts were independent risk factors for STEMI (P<0.05), and their ORs (95% CI) were 3.913 (1.574-9.728) and 2.13 (1.247-3.641), respectively. The present study reveals that miRNA-203 could be a possible candidate for a novel biomarker in the early prediction of STEMI.
Collapse
Affiliation(s)
- Jun Li
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
- Anhui Medical University, Hefei, 230000, Anhui, China
| | - Na Wang
- Department of Obstetrics and Gynecology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
- Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Xiang Wen
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Lu-Yao Huang
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Rui-Qing Cui
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Jing Zhang
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China.
| |
Collapse
|
28
|
Zhao J, Wu Q, Yang T, Nie L, Liu S, Zhou J, Chen J, Jiang Z, Xiao T, Yang J, Chu C. Gaseous signal molecule SO 2 regulates autophagy through PI3K/AKT pathway inhibits cardiomyocyte apoptosis and improves myocardial fibrosis in rats with type II diabetes. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:541-556. [PMID: 36302628 PMCID: PMC9614393 DOI: 10.4196/kjpp.2022.26.6.541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022]
Abstract
Myocardial fibrosis is a key link in the occurrence and development of diabetic cardiomyopathy. Its etiology is complex, and the effect of drugs is not good. Cardiomyocyte apoptosis is an important cause of myocardial fibrosis. The purpose of this study was to investigate the effect of gaseous signal molecule sulfur dioxide (SO2) on diabetic myocardial fibrosis and its internal regulatory mechanism. Masson and TUNEL staining, Western-blot, transmission electron microscopy, RT-qPCR, immunofluorescence staining, and flow cytometry were used in the study, and the interstitial collagen deposition, autophagy, apoptosis, and changes in phosphatidylinositol 3-kinase (PI3K)/AKT pathways were evaluated from in vivo and in vitro experiments. The results showed that diabetic myocardial fibrosis was accompanied by cardiomyocyte apoptosis and down-regulation of endogenous SO2-producing enzyme aspartate aminotransferase (AAT)1/2. However, exogenous SO2 donors could up-regulate AAT1/2, reduce apoptosis of cardiomyocytes induced by diabetic rats or high glucose, inhibit phosphorylation of PI3K/AKT protein, up-regulate autophagy, and reduce interstitial collagen deposition. In conclusion, the results of this study suggest that the gaseous signal molecule SO2 can inhibit the PI3K/AKT pathway to promote cytoprotective autophagy and inhibit cardiomyocyte apoptosis to improve myocardial fibrosis in diabetic rats. The results of this study are expected to provide new targets and intervention strategies for the prevention and treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Junxiong Zhao
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Qian Wu
- Department of General Practice, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Yang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,School of Pharmaceutical Science of University of South China, Hengyang 421000, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Shengquan Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jia Zhou
- Department of Ultrasound Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jian Chen
- Department of Critical Care Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Zhentao Jiang
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Xiao
- Department of Cardiology, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guang-dong Medical University, Shenzhen 518000, China,Ting Xiao, E-mail:
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,Jun Yang, E-mail:
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,Correspondence Chun Chu, E-mail:
| |
Collapse
|
29
|
Wang S, Tian C, Gao Z, Zhang B, Zhao L. Research status and trends of the diabetic cardiomyopathy in the past 10 years (2012–2021): A bibliometric analysis. Front Cardiovasc Med 2022; 9:1018841. [PMID: 36337893 PMCID: PMC9630656 DOI: 10.3389/fcvm.2022.1018841] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022] Open
Abstract
Background Diabetic cardiomyopathy is one of the most life-threatening diabetic complications. However, the previous studies only discuss a particular aspect or characteristic of DCM, the current state and trends were explored by limited research. We aimed to perform a systemically bibliometric study of DCM research progress status in the past decade, visualize the internal conceptual structure and potential associations, and further explore the prospective study trends. Methods Articles related to DCM published from January 2012 to December 2021 were collected in the Web of Science core collection (WoSCC) database on June 24, 2022. We exported all bibliographic records, including titles, abstracts, keywords, authorship, institutions, addresses, publishing sources, references, citation times, and year of publication. In addition, the journal Impact Factor and Hirsch index were obtained from the Journal Citation Report. We conducted the data screening, statistical analysis, and visualization via the Bibliometrix R package. VOS viewer software was employed to generate the collaboration network map among countries and institutions for better performance in visualization. Results In total, 1,887 original research articles from 2012 to 2021 were identified. The number of annual publications rapidly increased from 107 to 278, and a drastic increase in citation times was observed in 2017–2019. As for global contributions, the United States was the most influential country with the highest international collaboration, while China was the most productive country. Professor Cai Lu was the most prolific author. Shandong University published the most articles. Cardiovascular Diabetology journal released the most DCM-related articles. “Metabolic Stress-induced Activation of FoxO1 Triggers Diabetic Cardiomyopathy in Mice” Battiprolu PK et al., J Clin Invest, 2012. was the most top-cited article regarding local citations. The top three keywords in terms of frequency were apoptosis, oxidative stress, and fibrosis. The analysis of future topic trends indicated that “Forkhead box protein O1,” “Heart failure with preserved ejection fraction,” “Dapagliflozin,” “Thioredoxin,” “Mitochondria dysfunction,” “Glucose,” “Pyroptosis,” “Cardiac fibroblast” and “Long non-coding RNA” could be promising hotspots. Conclusion This study provides meaningful insights into DCM, which is expected to assist cardiologists and endocrinologists in exploring frontiers and future research directions in the domain through a refined and concise summary.
Collapse
Affiliation(s)
- Sicheng Wang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanxi Tian
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zezheng Gao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Boxun Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Boxun Zhang,
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Linhua Zhao,
| |
Collapse
|
30
|
Zhu L, Wang Y, Zhao S, Lu M. Detection of myocardial fibrosis: Where we stand. Front Cardiovasc Med 2022; 9:926378. [PMID: 36247487 PMCID: PMC9557071 DOI: 10.3389/fcvm.2022.926378] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Myocardial fibrosis, resulting from the disturbance of extracellular matrix homeostasis in response to different insults, is a common and important pathological remodeling process that is associated with adverse clinical outcomes, including arrhythmia, heart failure, or even sudden cardiac death. Over the past decades, multiple non-invasive detection methods have been developed. Laboratory biomarkers can aid in both detection and risk stratification by reflecting cellular and even molecular changes in fibrotic processes, yet more evidence that validates their detection accuracy is still warranted. Different non-invasive imaging techniques have been demonstrated to not only detect myocardial fibrosis but also provide information on prognosis and management. Cardiovascular magnetic resonance (CMR) is considered as the gold standard imaging technique to non-invasively identify and quantify myocardial fibrosis with its natural ability for tissue characterization. This review summarizes the current understanding of the non-invasive detection methods of myocardial fibrosis, with the focus on different techniques and clinical applications of CMR.
Collapse
Affiliation(s)
- Leyi Zhu
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yining Wang
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shihua Zhao
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjie Lu
- State Key Laboratory of Cardiovascular Disease, Department of Magnetic Resonance Imaging, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Cardiovascular Imaging (Cultivation), Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Minjie Lu
| |
Collapse
|
31
|
Ye Y, Duan B, Zhou Z, Han L, Huang F, Li J, Wang Q, Zeng X, Yu X. Integrated metabolomics and network pharmacology to reveal the mechanisms of Guizhi-Fuling treatment for myocardial ischemia. Chem Biodivers 2022; 19:e202200386. [PMID: 36073658 DOI: 10.1002/cbdv.202200386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/08/2022] [Indexed: 11/11/2022]
Abstract
Myocardial ischemia is a cardio-physiological condition caused by a decrease in blood perfusion to the heart, resulting in reduced oxygen supply and abnormal myocardial energy metabolism. Guizhi-Fuling (GZFL) is effective in treating Myocardial ischemia. However, its mechanism of action remains unclear and requires further exploration. we hope to reveal the mechanisms of GZFL treating Myocardial ischemia by integrating metabolomics and network pharmacology. In this study, myocardial metabolomic analysis was first performed using GC-MS to discover the potential mechanism of action of GZFL on myocardial ischemia. Then, network pharmacology was used to analyze key pathways and construct a pathway-core target network. Molecular docking was used to validate core targets in network pharmacological signaling pathways. Finally, western blots were used to verify core targets of metabolomics and network pharmacology integrated pathways as well as key targets in signaling pathways. As a result, we identified 22 important biomarkers of GZFL for the treatment of myocardial ischemia. Most of these metabolites were restored by modulation after GZFL treatment. Based on the network pharmacology, 297 targets of GZFL in the treatment of myocardial ischemia were obtained. The further comprehensive analysis focused on 3 key targets, including Tyrosine hydroxylase (TH), myeloperoxidase (MPO), and phosphatidylinositol 3-kinases (PIK3CA), and their associated metabolites and pathways. Compared with the model group, the protein expression levels of TH, MPO and PIK3CA were decreased in GZFL. Therefore, the mechanism of GZFL for treating myocardial ischemia may be to inhibit myocardial inflammatory factors, reduce myocardial inflammation, and restore endothelial function, while regulating norepinephrine release and uric acid concentration.
Collapse
Affiliation(s)
- Yan Ye
- Hubei University of Chinese Medicine, college of pharmacy, , 430065, Wuhan, CHINA
| | - Bailu Duan
- Hubei University of Chinese Medicine, College of Basic Medicine, Qingling Street, Wuhan, 430065, wuhan, CHINA
| | - Zhenxiang Zhou
- Hubei University of Chinese Medicine, College of Basic Medicine, Qingling Street, Wuhan, 430065, Wuhan, CHINA
| | - Lintao Han
- Hubei University of Chinese Medicine, College of Pharmacy, Qingling Street, Wuhan, wuhan, CHINA
| | - Fang Huang
- Hubei University of Chinese Medicine, College of Basic Medicine, Qingling Street, Wuhan, wuhan, CHINA
| | - Jingjing Li
- Hubei University of Chinese Medicine, College of Basic Medicine, Qingling Street, Wuhan, wuhan, CHINA
| | - Qiong Wang
- Hubei University of Chinese Medicine, College of Basic Medicine, Qingling Street, Wuhan, wuhan, CHINA
| | - Xiangfa Zeng
- Hubei University of Chinese Medicine, Qingling Street, Wuhan, wuhan, CHINA
| | - Xiaoming Yu
- Hubei University of Chinese Medicine, College of Basic Medicine, Qingling Street, Wuhan, wuhan, CHINA
| |
Collapse
|
32
|
Pisklova M, Osmak G, Favorova O. Regulation of SMAD Signaling Pathway by miRNAs Associated with Myocardial Fibrosis: In silico Analysis of Target Gene Networks. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:832-838. [PMID: 36171647 DOI: 10.1134/s0006297922080144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 06/16/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a hereditary heart disease caused by mutations in the sarcomere genes, which is accompanied by myocardial fibrosis leading to progressive heart failure and arrhythmias. Recent studies suggest that the HCM development involves dysregulation of gene expression. Among the molecules involved in this process are microRNAs (miRNAs), which are short non-coding RNAs. Typically, one miRNA regulates several target genes post-transcriptionally, hence, it might be difficult to determine the role of a particular miRNA in the disease pathogenesis. In this study, using the PubMed database, we selected 15 miRNAs whose expression is associated with myocardial fibrosis, one of the critical pathological processes in HCM. We then used an earlier developed algorithm to search in silico for the signaling pathways regulated by these miRNAs and found that ten of them participate in the regulation of the TGF-β/SMAD signaling pathway. At the same time, among the SMAD signaling pathway genes, the target of the most identified miRNAs was the MYC gene, which is involved in the development of fibrosis in some tissues. In our earlier work, we found that the TGF-β/SMAD pathway is also regulated by a set of other miRNAs associated with the myocardial hypertrophy in HCM. The fact that two sets of miRNAs identified in two independent bioinformatic studies are involved in the regulation of the same signaling pathway indicates that the SMAD signaling cascade is indeed a key element in the regulation of pathological processes in HCM. The obtained data might contribute to understanding pathological processes underlying HCM development.
Collapse
Affiliation(s)
- Maria Pisklova
- Chazov National Medical Research Center of Cardiology, Moscow, 121552, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - German Osmak
- Chazov National Medical Research Center of Cardiology, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Olga Favorova
- Chazov National Medical Research Center of Cardiology, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
33
|
lncRNA PINK1-AS Aggravates Cerebral Ischemia/Reperfusion Oxidative Stress Injury through Regulating ATF2 by Sponging miR-203. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1296816. [PMID: 35855866 PMCID: PMC9288285 DOI: 10.1155/2022/1296816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/19/2022] [Indexed: 01/07/2023]
Abstract
Ischemic stroke is a common disease that led to high mortality and high disability. NADPH oxidase 2- (NOX2-) mediated oxidative stress and long noncoding RNA have important roles in cerebral ischemia/reperfusion (CI/R) injury, whereas whether there is interplay between them remains to be clarified. This study was performed to observe the role of lncRNA PINK1-antisense RNA (PINK1-AS) in NOX2 expression regulation. An in vivo rat model (MCAO) and an in vitro cell model (H/R: hypoxia/reoxygenation) were utilized for CI/R oxidative stress injury investigation. The expression levels of lncRNA PINK1-AS, activating transcription factor 2 (ATF2), NOX2, and caspase-3 and the production level of ROS and cell apoptosis were significantly increased in CI/R injury model rats or in H/R-induced SH-SY5Y cells, but miR-203 was significantly downregulated. There was positive correlation between PINK1-AS expression level and ROS production level. PINK1-AS and ATF2 were found to be putative targets of miR-203. Knockdown of lncRNA PINK1-AS or ATF2 or the overexpression of miR-203 significantly reduced oxidative stress injury via inhibition of NOX2. Overexpression of lncRNA PINK1 significantly led to oxidative stress injury in SH-SY5Y cells through downregulating miR-203 and upregulating ATF2 and NOX2. lncRNA PINK1-AS and ATF2 were the targets of miR-203, and the lncRNA PINK1-AS/miR-203/ATF2/NOX2 axis plays pivotal roles in CI/R injury. Therefore, lncRNA PINK1-AS is a possible target for CR/I injury therapy by sponging miR-203.
Collapse
|
34
|
Hamidi AA, Taghehchian N, Basirat Z, Zangouei AS, Moghbeli M. MicroRNAs as the critical regulators of cell migration and invasion in thyroid cancer. Biomark Res 2022; 10:40. [PMID: 35659780 PMCID: PMC9167543 DOI: 10.1186/s40364-022-00382-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/07/2022] [Indexed: 12/14/2022] Open
Abstract
Thyroid cancer (TC) is one of the most frequent endocrine malignancies that is more common among females. Tumor recurrence is one of the most important clinical manifestations in differentiated TC which is associated with different factors including age, tumor size, and histological features. Various molecular processes such as genetic or epigenetic modifications and non-coding RNAs are also involved in TC progression and metastasis. The epithelial-to-mesenchymal transition (EMT) is an important biological process during tumor invasion and migration that affects the initiation and transformation of early-stage tumors into invasive malignancies. A combination of transcription factors, growth factors, signaling pathways, and epigenetic regulations affect the thyroid cell migration and EMT process. MicroRNAs (miRNAs) are important molecular factors involved in tumor metastasis by regulation of EMT-activating signaling pathways. Various miRNAs are involved in the signaling pathways associated with TC metastasis which can be used as diagnostic and therapeutic biomarkers. Since, the miRNAs are sensitive, specific, and non-invasive, they can be suggested as efficient and optimal biomarkers of tumor invasion and metastasis. In the present review, we have summarized all of the miRNAs which have been significantly involved in thyroid tumor cells migration and invasion. We also categorized all of the reported miRNAs based on their cellular processes to clarify the molecular role of miRNAs during thyroid tumor cell migration and invasion. This review paves the way of introducing a non-invasive diagnostic and prognostic panel of miRNAs in aggressive and metastatic TC patients.
Collapse
Affiliation(s)
- Amir Abbas Hamidi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Basirat
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Pujar M, Vastrad B, Kavatagimath S, Vastrad C, Kotturshetti S. Identification of candidate biomarkers and pathways associated with type 1 diabetes mellitus using bioinformatics analysis. Sci Rep 2022; 12:9157. [PMID: 35650387 PMCID: PMC9160069 DOI: 10.1038/s41598-022-13291-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 05/16/2022] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a metabolic disorder for which the underlying molecular mechanisms remain largely unclear. This investigation aimed to elucidate essential candidate genes and pathways in T1DM by integrated bioinformatics analysis. In this study, differentially expressed genes (DEGs) were analyzed using DESeq2 of R package from GSE162689 of the Gene Expression Omnibus (GEO). Gene ontology (GO) enrichment analysis, REACTOME pathway enrichment analysis, and construction and analysis of protein–protein interaction (PPI) network, modules, miRNA-hub gene regulatory network and TF-hub gene regulatory network, and validation of hub genes were performed. A total of 952 DEGs (477 up regulated and 475 down regulated genes) were identified in T1DM. GO and REACTOME enrichment result results showed that DEGs mainly enriched in multicellular organism development, detection of stimulus, diseases of signal transduction by growth factor receptors and second messengers, and olfactory signaling pathway. The top hub genes such as MYC, EGFR, LNX1, YBX1, HSP90AA1, ESR1, FN1, TK1, ANLN and SMAD9 were screened out as the critical genes among the DEGs from the PPI network, modules, miRNA-hub gene regulatory network and TF-hub gene regulatory network. Receiver operating characteristic curve (ROC) analysis confirmed that these genes were significantly associated with T1DM. In conclusion, the identified DEGs, particularly the hub genes, strengthen the understanding of the advancement and progression of T1DM, and certain genes might be used as candidate target molecules to diagnose, monitor and treat T1DM.
Collapse
Affiliation(s)
- Madhu Pujar
- Department of Pediatrics, J J M Medical College, Davangere, Karnataka, 577004, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag, Karnataka, 582101, India
| | - Satish Kavatagimath
- Department of Pharmacognosy, K.L.E. College of Pharmacy, Belagavi, Karnataka, 590010, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India.
| | - Shivakumar Kotturshetti
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India
| |
Collapse
|
36
|
Cheng Z, Zhang Y, Wu S, Zhao R, Yu Y, Zhou Y, Zhou Z, Dong Y, Qiu A, Xu H, Liu Y, Zhang W, Tian T, Wu Q, Gu H, Chu M. Peripheral blood circular RNA hsa_circ_0058493 as a potential novel biomarker for silicosis and idiopathic pulmonary fibrosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 236:113451. [PMID: 35378401 DOI: 10.1016/j.ecoenv.2022.113451] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/16/2022] [Accepted: 03/20/2022] [Indexed: 06/14/2023]
Abstract
Existing studies reported that some circular RNAs (circRNAs) play vital roles in the development of pulmonary fibrosis. However, few studies explored the biomarker potential of circRNAs for pulmonary fibrosis based on population data. Therefore, we aimed to identify peripheral blood circRNAs as potential biomarkers for diagnosing silicosis and idiopathic pulmonary fibrosis (IPF). In brief, an RNA-seq screening based on 4 silicosis cases and 4 controls was initially performed. Differentially expressed circRNAs were combined with the human serum circRNA dataset to identify overlapping serum-detectable circRNAs, followed by validation using the GEO dataset (3 IPF cases and 3 controls) and subsequent qRT-PCR, including 84 additional individuals. Following the above steps, 243 differentially expressed circRNAs were identified during the screening stage, with fold changes ≥ 1.5 and P < 0.05. Of note, the human serum circRNA dataset encompassed 28 of 243 circRNAs. GEO (GSE102660) validation revealed two highly expressed circRNAs (P < 0.05) in the IPF case group. Furthermore, at the enlarged sample validation stage, hsa_circ_0058493 was highly expressed in both silicosis and IPF cases (silicosis: P = 1.16 × 10-6; IPF: P = 7.46 × 10-5). Additionally, hsa_circ_0058493 expression was significantly increased in MRC-5 cells upon TGF-β1 treatment, while hsa_circ_0058493 knockdown inhibited the expression of fibrotic molecules by affecting the epithelial-mesenchymal transition process. These shreds of evidence indicated that hsa_circ_0058493 might serve as a novel biomarker for diagnosing silicosis and IPF.
Collapse
Affiliation(s)
- Zhounan Cheng
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Yingyi Zhang
- Department of Occupational Disease, the Occupational Disease Institute of Wuxi, Wuxi, Jiangsu, China
| | - Shuangshuang Wu
- Department of Geriatrics, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Zhao
- Department of Occupational Disease, the Occupational Disease Institute of Wuxi, Wuxi, Jiangsu, China
| | - Yuhui Yu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Yan Zhou
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Zhen Zhou
- Department of Mathematics and Applied Mathematics, University of Science and Technology of China, Hefei, Anhui, China
| | - Yang Dong
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Anni Qiu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Huiwen Xu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Yiran Liu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Wendi Zhang
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Tian Tian
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Qiuyun Wu
- School of Public Health, Xuzhou Medical University, Xuzhou, China.
| | - Hongyan Gu
- Department of Respiratory, the Sixth People's Hospital of Nantong, Nantong, Jiangsu, China.
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
37
|
Mittal A, Garg R, Bahl A, Khullar M. Molecular Mechanisms and Epigenetic Regulation in Diabetic Cardiomyopathy. Front Cardiovasc Med 2022; 8:725532. [PMID: 34977165 PMCID: PMC8716459 DOI: 10.3389/fcvm.2021.725532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus (DM) is an important lifestyle disease. Type 2 diabetes is one of the prime contributors to cardiovascular diseases (CVD) and diabetic cardiomyopathy (DbCM) and leads to increased morbidity and mortality in patients with DM. DbCM is a typical cardiac disease, characterized by cardiac remodeling in the presence of DM and in the absence of other comorbidities such as hypertension, valvular diseases, and coronary artery disease. DbCM is associated with defective cardiac metabolism, altered mitochondrial structure and function, and other physiological and pathophysiological signaling mechanisms such as oxidative stress, inflammation, myocardial apoptosis, and autophagy. Epigenetic modifiers are crucial players in the pathogenesis of DbCM. Thus, it is important to explore the role of epigenetic modifiers or modifications in regulating molecular pathways associated with DbCM. In this review, we have discussed the role of various epigenetic mechanisms such as histone modifications (acetylation and methylation), DNA methylation and non-coding RNAs in modulating molecular pathways involved in the pathophysiology of the DbCM.
Collapse
Affiliation(s)
- Anupam Mittal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajni Garg
- Council of Scientific and Industrial Research - Institute of Microbial Technology, Chandigarh, India
| | - Ajay Bahl
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
38
|
Zhu C, Zhang H, Wei D, Sun Z. Silencing lncRNA GAS5 alleviates apoptosis and fibrosis in diabetic cardiomyopathy by targeting miR-26a/b-5p. Acta Diabetol 2021; 58:1491-1501. [PMID: 34091757 DOI: 10.1007/s00592-021-01745-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND LncRNA GAS5 is associated with high glucose-induced cardiomyocyte injury, but its role in diabetic cardiomyopathy (DCM) remains unclear. METHODS Mice were administered with streptozotocin to construct the diabetic model (DM). Primary mouse cardiomyocytes were isolated and treated with 30 mmol/L high glucose to mimic the diabetic condition in vitro. GAS5 expression was detected by quantitative reverse transcription polymerase chain reaction. The relationship between GAS5 and miR-26a/b-5p was determined by bioinformatic prediction, luciferase reporter assay and RNA immunoprecipitation assay. The cardiac function of diabetic mice was evaluated by two-dimensional echocardiography. RESULTS GAS5 was significantly upregulated in diabetic cardiomyopathy both in vitro and in vivo. GAS5 knockdown and miR-26a/b-5p overexpression not only effectively attenuated myocardial fibrosis of diabetic mice in vivo but also inhibited high glucose-induced cardiomyocyte injury in vitro. miR-26a/b-5p was identified as a target of GAS5. GAS5 knockdown efficiently attenuated myocardial fibrosis and high glucose-induced cardiomyocyte injury through negatively regulating miR-26a/b-p. CONCLUSION Our study showed that GAS5 promotes DCM progression by regulating miR-26a/b-5p, suggesting that GAS5 might be a potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Chunping Zhu
- Department of Cardiac Function, The First Hospital of Qiqihar & Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, People's Republic of China
| | - Haijun Zhang
- The Second Department of Endocrinology, The First Hospital of Qiqihar & Affiliated Qiqihar Hospital, Southern Medical University, No. 30 Park Road, Longsha, Qiqihar, 161005, Heilongjiang, People's Republic of China.
| | - Dongmei Wei
- Department of Traditional Chinese Medicine Geriatrics, The First Hospital of Qiqihar & Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, People's Republic of China
| | - Zhe Sun
- The Second Department of Endocrinology, The First Hospital of Qiqihar & Affiliated Qiqihar Hospital, Southern Medical University, No. 30 Park Road, Longsha, Qiqihar, 161005, Heilongjiang, People's Republic of China
| |
Collapse
|
39
|
The role of microRNAs in diseases and related signaling pathways. Mol Biol Rep 2021; 49:6789-6801. [PMID: 34718938 DOI: 10.1007/s11033-021-06725-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 10/19/2022]
Abstract
MicroRNAs (miRNAs) are epigenetic regulators of the gene expression and act through posttranslational modification. They bind to 3'-UTR of target mRNAs to inhibit translation or increase the degradation mRNA in many tissues. Any alteration in the level of miRNA expression in many human diseases indicates their involvement in the pathogenesis of many diseases. On the other hand, the regulation of the signaling pathways is necessary for the maintenance of natural and physiological characteristics of any cell. It is worth mentioning that dysfunction of the signaling pathways manifests itself as a disorder or disease. The significant evidence report that miRNAs regulate the several signaling pathways in many diseases. Base on previous studies, miRNAs can be used for therapeutic or diagnostic purposes. According to the important role of miRNAs on the cell signaling pathways, this article reviews miRNAs involvement in incidence of diseases by changing signaling pathways.
Collapse
|
40
|
Jin ZQ. MicroRNA targets and biomarker validation for diabetes-associated cardiac fibrosis. Pharmacol Res 2021; 174:105941. [PMID: 34656765 DOI: 10.1016/j.phrs.2021.105941] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Cardiac fibrosis is one of the main characteristics of diabetic cardiomyopathy and manifests excessive accumulation of extracellular matrix proteins in the heart. Several signaling pathways have been proposed for pathogenesis of cardiac fibrosis in the diabetic heart. TGF-β/Smad2/3-dependent or independent pathway is the major signaling molecule core in the pathogenesis of cardiac fibrosis. MicroRNAs (miRNAs, miR) are ~22-nuceotide regulatory RNAs that are involved in gene silencing through the degradation of post-transcriptional mRNA or suppression of the expressed proteins. Hyperglycemia in the diabetic heart regulates expression of some miRNAs. Target molecules of miRNAs can be identified through biocomputational database initial screening and dual luciferase assay validation. miR-21, miR-150-5p, miR-155, miR-216a-3p, miR-221-3p, miR-223, and miR-451 were up-regulated in the diabetic heart and promoted cardiac fibrosis through targeting signaling pathways in cardiac fibroblasts, endothelial cells, and cardiac myocytes. miR-15a/-15b, miR-18a-5p, miR-20a-5p, miR-26b-5p, miR-29, miR-133a, miR-141, miR-146, miR-200b, miR-203, miR-222, and miR-551b-5p were down-regulated in the diabetic heart and exhibited anti-fibrosis when they were overexpressed. miRNAs are stable molecules and may reflect the pathological changes of organs. Some miRNAs have been detected in the plasma or serum in patients with diabetes mellitus or heart failure. Exploration of targets and biomarkers of miRNA may provide additional information on pathogenesis and diagnosis of cardiac fibrosis and novel targets to tackle diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhu-Qiu Jin
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA 95757, USA.
| |
Collapse
|
41
|
Zhang Y, Xu C. Integrative analysis of miRNA-mRNA expression profiles in esophageal fibrosis after ESD. Exp Ther Med 2021; 22:1176. [PMID: 34504621 PMCID: PMC8393932 DOI: 10.3892/etm.2021.10610] [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: 10/18/2020] [Accepted: 03/01/2021] [Indexed: 11/25/2022] Open
Abstract
The incidence of esophageal fibrosis and benign esophageal stricture (BES) has increased in recent years due to the curative therapy for early-stage esophageal carcinoma, including partial esophagectomy and esophageal endoscopic submucosal dissection (ESD). The aim of the present study was to identify key genes and associated pathways of esophageal fibrosis after the ESD procedure. During the esophageal ESD procedure, the esophageal tissue in the remaining submucosal layer, referred to as normal esophageal (NE) tissue, was collected, and 1 week thereafter, post-operative esophageal (PE) tissue was obtained. High-throughput sequencing was used to identify dysregulated microRNAs (miRNAs/miRs) between NE and PE tissues. According to the differentially expressed (DE) miRNAs, putative target genes were predicted. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes enrichment analysis and DEmiRNA interaction network analysis were performed. Reverse transcription-quantitative PCR (RT-qPCR) was performed to validate the RNA-sequencing results. A total of 199 miRNAs were determined to be DE between NE and PE tissues. Compared with the expression in the NE group, 83 miRNAs were significantly upregulated, while 116 miRNAs were significantly downregulated. According to these DE miRNAs, forkhead box O1 (FOXO1), paired box 6 (PAX6), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and adrenoceptor β1 (ADRB1) were DE genes regulated by five DE miRNAs, including miR-223-3p, miR-142-5p, miR-582-5p, miR-21-3p and miR-218-5p. The results suggested that certain pathways were markedly dysregulated, including FOXO, MAPK, AMP-activated protein kinase and signaling pathways regulating the pluripotency of stem cells and proteoglycans in cancer. According to the RT-qPCR results, the expression levels of FOXO1, PAX6, ADRB1, miR-223-3p, miR-582-5p, miR-21-3p and miR-218-5p were consistent with the integrated analysis. In conclusion, FOXO1, PAX6, PIK3CA and ADRB1 may have a role in esophageal fibrosis, regulated by miR-223-3p, miR-142-5p, miR-582-5p, miR-21-3p and miR-218-5p. The present results provided an improved understanding of the changes in the microenvironment during the process of esophageal fibrosis, as well as novel potential targets for the treatment of esophageal fibrosis and BES.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Digestive Diseases, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
42
|
Tuleta I, Frangogiannis NG. Fibrosis of the diabetic heart: Clinical significance, molecular mechanisms, and therapeutic opportunities. Adv Drug Deliv Rev 2021; 176:113904. [PMID: 34331987 PMCID: PMC8444077 DOI: 10.1016/j.addr.2021.113904] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/02/2023]
Abstract
In patients with diabetes, myocardial fibrosis may contribute to the pathogenesis of heart failure and arrhythmogenesis, increasing ventricular stiffness and delaying conduction. Diabetic myocardial fibrosis involves effects of hyperglycemia, lipotoxicity and insulin resistance on cardiac fibroblasts, directly resulting in increased matrix secretion, and activation of paracrine signaling in cardiomyocytes, immune and vascular cells, that release fibroblast-activating mediators. Neurohumoral pathways, cytokines, growth factors, oxidative stress, advanced glycation end-products (AGEs), and matricellular proteins have been implicated in diabetic fibrosis; however, the molecular links between the metabolic perturbations and activation of a fibrogenic program remain poorly understood. Although existing therapies using glucose- and lipid-lowering agents and neurohumoral inhibition may act in part by attenuating myocardial collagen deposition, specific therapies targeting the fibrotic response are lacking. This review manuscript discusses the clinical significance, molecular mechanisms and cell biology of diabetic cardiac fibrosis and proposes therapeutic targets that may attenuate the fibrotic response, preventing heart failure progression.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA.
| |
Collapse
|
43
|
Lin Y, Shen Y, Chen J, Hu C, Zhou Z, Yuan C. The Function of LncRNA FTX in Several Common Cancers. Curr Pharm Des 2021; 27:2381-2386. [PMID: 33121404 DOI: 10.2174/1381612826666201029164036] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND LncRNA is a kind of non-coding RNA and its research is more popular in recent years, which has more than 200 nucleotides. It plays a significant part in various biological functions, including chromosome modification, genome modification, transcriptional activation, transcriptional interference, and other processes. FTX, at the center of the X chromosome inactivation and it has been shown that lncRNA FTX regulates cancer cells' development, migration, and invasion in many studies. METHODS Relevant literature was collected through the PubMed system search and is summarized in this article. RESULTS LncRNA FTX abnormally increased in tumor cells, such as liver cancer, stomach cancer, leukemia, renal cell carcinoma, colorectal cancer, glioma, osteosarcoma, etc. However, the expression level decreased in temporal lobe epilepsy, liver cirrhosis, heart failure, etc. Conclusion: FTX may be an important regulatory factor and a potential therapeutic target in cancers.
Collapse
Affiliation(s)
- Yexiang Lin
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yujie Shen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Jinlan Chen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Chengyu Hu
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Zixian Zhou
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
44
|
Overexpressing microRNA-203 alleviates myocardial infarction via interacting with long non-coding RNA MIAT and mitochondrial coupling factor 6. Arch Pharm Res 2021; 44:525-535. [PMID: 33942232 DOI: 10.1007/s12272-021-01324-8] [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: 10/18/2020] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
Myocardial infarction (MI) is one of the leading causes of high mortality worldwide. Long non-coding RNA myocardial infarction associated transcript (MIAT) and mitochondrial coupling factor 6 (CF6) aggravate MI. This study aimed to elucidate whether miR-203 interacted with MIAT and CF6 in MI. Results revealed that MIAT and CF6 expressions were upregulated and that miR-203 was downregulated in mouse myocardial tissues after MI, as well as in hypoxic mouse cardiomyocytes. The overexpression of MIAT in mouse cardiomyocytes raised CF6 expression, whereas the knockdown of MIAT had the opposite effect. Mechanistically, the luciferase reporter and RNA pull-down assays corroborated the binding between miR-203 and CF6 3'UTR and between miR-203 and MIAT. The simultaneous overexpression of miR-203 and MIAT restored the reduction of CF6 caused by miR-203 overexpression alone, and the overexpression of miR-203 diminished the percentage of infarct area and the apoptosis of cardiomyocytes in vivo. Our findings corroborate that overexpressing miR-203 alleviates MI via interacting with MIAT and CF6.
Collapse
|
45
|
Duan YR, Chen BP, Chen F, Yang SX, Zhu CY, Ma YL, Li Y, Shi J. LncRNA lnc-ISG20 promotes renal fibrosis in diabetic nephropathy by inducing AKT phosphorylation through miR-486-5p/NFAT5. J Cell Mol Med 2021; 25:4922-4937. [PMID: 33939247 PMCID: PMC8178263 DOI: 10.1111/jcmm.16280] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Long non‐coding RNA (lncRNA) lnc‐ISG20 has been found aberrantly up‐regulated in the glomerular in the patients with diabetic nephropathy (DN). We aimed to elucidate the function and regulatory mechanism of lncRNA lnc‐ISG20 on DN‐induced renal fibrosis. Expression patterns of lnc‐ISG20 in kidney tissues of DN patients were determined by RT‐qPCR. Mouse models of DN were constructed, while MCs were cultured under normal glucose (NG)/high glucose (HG) conditions. The expression patterns of fibrosis marker proteins collagen IV, fibronectin and TGF‐β1 were measured with Western blot assay. In addition, the relationship among lnc‐ISG20, miR‐486‐5p, NFAT5 and AKT were analysed using dual‐luciferase reporter assay and RNA immunoprecipitation. The effect of lnc‐ISG20 and miR‐486/NFAT5/p‐AKT axis on DN‐associated renal fibrosis was also verified by means of rescue experiments. The expression levels of lnc‐ISG20 were increased in DN patients, DN mouse kidney tissues and HG‐treated MCs. Lnc‐ISG20 silencing alleviated HG‐induced fibrosis in MCs and delayed renal fibrosis in DN mice. Mechanistically, miR‐486‐5p was found to be a downstream miRNA of lnc‐ISG20, while miR‐486‐5p inhibited the expression of NFAT5 by binding to its 3'UTR. NFAT5 overexpression aggravated HG‐induced fibrosis by stimulating AKT phosphorylation. However, NFAT5 silencing reversed the promotion of in vitro and in vivo fibrosis caused by lnc‐ISG20 overexpression. Our collective findings indicate that lnc‐ISG20 promotes the renal fibrosis process in DN by activating AKT through the miR‐486‐5p/NFAT5 axis. High‐expression levels of lnc‐ISG20 may be a useful indicator for DN.
Collapse
Affiliation(s)
- Yu-Rui Duan
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Bao-Ping Chen
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Fang Chen
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Su-Xia Yang
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Chao-Yang Zhu
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Ya-Li Ma
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Yang Li
- Department of Urology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jun Shi
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
46
|
Zheng W, Li T, Wei J, Zhang Y, Zuo Q, Lin Y. Identification of miR-145 as a regulator of the cardiomyocyte inflammatory response and oxidative stress under hyperglycemia. Exp Ther Med 2021; 21:467. [PMID: 33763154 PMCID: PMC7983182 DOI: 10.3892/etm.2021.9898] [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: 01/16/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
The current study aimed to explore the effects of microRNA (miR)-145 on the inflammatory response and oxidative stress (OS) in high glucose (HG)-induced cardiomyocytes, as well as the specific mechanism underlying this action. H9c2 cells were treated with 33 mmol/l glucose (HG group) or cotreated with 24.5 mmol/l mannitol and 5.5 mmol/l glucose (hypertonic group), and the expression levels of miR-145 and ADP ribosylation factor 6 (ARF6) were detected. The cells were transfected with pcDNA3.1-ARF6, miR-145 mimics or corresponding negative controls prior to the assessment of cell survival rate. Levels of lactate dehydrogenase (LDH), reactive oxygen species (ROS) and malondialdehyde (MDA), as well as the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), and the levels of IL-6, TNF-α and monocyte chemoattractant protein-1 (MCP-1) were subsequently determined. The apoptotic rate of H9c2 cells was examined by flow cytometry. The interaction between miR-145-ARF6 was predicted and confirmed by luciferase reporter assays. In the HG group, miR-145 expression was significantly decreased and ARF6 expression significantly increased compared with controls. Furthermore, the levels of inflammatory factors (IL-6, TNF-α and MCP-1), LDH, ROS and MDA were significantly elevated in the HG group compared with controls. Significantly decreased SOD, CAT and GPx activities and significantly increased numbers of apoptotic cells were observed in the HG group compared with controls. The cells transfected with miR-145 mimics exhibited significantly decreased LDH, ROS and MDA levels, significantly increased antioxidant enzyme activities and significantly decreased apoptotic rates compared with controls, while the opposite results were observed in cells transfected with pcDNA3.1-ARF6. Moreover, co-transfection with miR-145 mimics and pcDNA3.1-ARF6 exacerbated the inflammatory response and OS injury in HG-induced cardiomyocytes compared with cells transfected with miR-145 mimics alone. Furthermore, miR-145 negatively targeted ARF6. miR-145 attenuated the HG-induced inflammatory response and OS injury in cardiomyocytes by negatively regulating ARF6, which may contribute to providing a theoretical basis for the treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Wan Zheng
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Tianfa Li
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Junping Wei
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Yuanyuan Zhang
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Qi Zuo
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Yun Lin
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| |
Collapse
|
47
|
Jakubik D, Fitas A, Eyileten C, Jarosz-Popek J, Nowak A, Czajka P, Wicik Z, Sourij H, Siller-Matula JM, De Rosa S, Postula M. MicroRNAs and long non-coding RNAs in the pathophysiological processes of diabetic cardiomyopathy: emerging biomarkers and potential therapeutics. Cardiovasc Diabetol 2021; 20:55. [PMID: 33639953 PMCID: PMC7916283 DOI: 10.1186/s12933-021-01245-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/13/2021] [Indexed: 02/08/2023] Open
Abstract
The epidemic of diabetes mellitus (DM) necessitates the development of novel therapeutic and preventative strategies to attenuate complications of this debilitating disease. Diabetic cardiomyopathy (DCM) is a frequent disorder affecting individuals diagnosed with DM characterized by left ventricular hypertrophy, diastolic and systolic dysfunction and myocardial fibrosis in the absence of other heart diseases. Progression of DCM is associated with impaired cardiac insulin metabolic signaling, increased oxidative stress, impaired mitochondrial and cardiomyocyte calcium metabolism, and inflammation. Various non-coding RNAs, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), as well as their target genes are implicated in the complex pathophysiology of DCM. It has been demonstrated that miRNAs and lncRNAs play an important role in maintaining homeostasis through regulation of multiple genes, thus they attract substantial scientific interest as biomarkers for diagnosis, prognosis and as a potential therapeutic strategy in DM complications. This article will review the different miRNAs and lncRNA studied in the context of DM, including type 1 and type 2 diabetes and the contribution of pathophysiological mechanisms including inflammatory response, oxidative stress, apoptosis, hypertrophy and fibrosis to the development of DCM .
Collapse
Affiliation(s)
- Daniel Jakubik
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Alex Fitas
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Joanna Jarosz-Popek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.,Doctoral School, Medical University of Warsaw, 02-091, Warsaw, Poland
| | - Anna Nowak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Pamela Czajka
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.,Centro de Matemática, Computação e Cognição, Universidade Federal Do ABC, São Paulo, Brazil
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.,Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Cardiovascular Research Center, "Magna Graecia" University, Catanzaro, Italy
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CEPT, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland.
| |
Collapse
|
48
|
Lv S, Yuan P, Lu C, Dong J, Li M, Qu F, Zhu Y, Zhang J. QiShenYiQi pill activates autophagy to attenuate reactive myocardial fibrosis via the PI3K/AKT/mTOR pathway. Aging (Albany NY) 2021; 13:5525-5538. [PMID: 33582656 PMCID: PMC7950250 DOI: 10.18632/aging.202482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/20/2020] [Indexed: 01/08/2023]
Abstract
QiShenYiQi pill (QSYQ), a traditional Chinese medicine, is used to treat cardiovascular diseases. However, the dose-effect relationship of its intervention in the reactive myocardial fibrosis is elusive. In this work, rat models of reactive myocardial fibrosis induced by partial abdominal aortic coarctation were constructed and randomly classified into the model group, 3-methyladenine group, rapamycin group, QSYQ low-dose group, QSYQ medium-dose group, QSYQ high-dose group, and sham-operated rats (control group). We revealed that QSYQ lowered the heart mass index (HMI), left ventricular mass index (LVMI), and myocardial collagen volume fraction (CVF) levels in a dose-dependent mechanism. Additionally, QSYQ increased the number of autophagosomes, and the expression of myocardial Beclin-1 and LC3B. In contrast, it reduced the expression of myocardial p62 and decreased the ratios of myocardial p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR. In conclusion, our results have revealed that QSYQ impacts anti-reactive myocardial fibrosis in a dose-dependent mechanism which is mediated by the activation of myocardial autophagy via the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Peng Yuan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Chunmiao Lu
- Jiashan Hospital of Traditional Chinese Medicine, Jiaxing 314100, Zhejiang, China
| | - Jianping Dong
- Health Center of Balitai Town, Jinnan, Tianjin 300350, China
| | - Meng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Fan Qu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yaping Zhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| |
Collapse
|
49
|
Ding Y, Wang Y, Zhang W, Jia Q, Wang X, Li Y, Lv S, Zhang J. Roles of Biomarkers in Myocardial Fibrosis. Aging Dis 2020; 11:1157-1174. [PMID: 33014530 PMCID: PMC7505259 DOI: 10.14336/ad.2020.0604] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Myocardial fibrosis is observed in various cardiovascular diseases and plays a key role in the impairment of cardiac function. Endomyocardial biopsy, as the gold standard for the diagnosis of myocardial fibrosis, has limitations in terms of clinical application. Therefore, biomarkers have been recommended for noninvasive assessment of myocardial fibrosis. This review discusses the role of biomarkers in myocardial fibrosis from the perspective of collagen.
Collapse
Affiliation(s)
- Yuejia Ding
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yuan Wang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Wanqin Zhang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qiujin Jia
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoling Wang
- 3Qian'an Hospital of Traditional Chinese Medicine, Qian'an 064400, China
| | - Yanyang Li
- 4Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shichao Lv
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.,2Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300000, China
| | - Junping Zhang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| |
Collapse
|
50
|
Ren BC, Zhang YF, Liu SS, Cheng XJ, Yang X, Cui XG, Zhao XR, Zhao H, Hao MF, Li MD, Tie YY, Qu L, Li XY. Curcumin alleviates oxidative stress and inhibits apoptosis in diabetic cardiomyopathy via Sirt1-Foxo1 and PI3K-Akt signalling pathways. J Cell Mol Med 2020; 24:12355-12367. [PMID: 32961025 PMCID: PMC7687015 DOI: 10.1111/jcmm.15725] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
Diabetes is a disorder of glucose metabolism, and over 90% are type 2 diabetes. Diabetic cardiomyopathy (DCM) is one of the type 2 diabetes complications, usually accompanied by changes in myocardial structure and function, together with cardiomyocyte apoptosis. Our study investigated the effect of curcumin on regulating oxidative stress (OS) and apoptosis in DCM. In vivo, diabetes was induced in an experimental rat model by streptozoticin (STZ) together with high‐glucose and high‐fat (HG/HF) diet feeding. In vitro, H9c2 cardiomyocytes were cultured with high‐glucose and saturated free fatty acid palmitate. Curcumin was orally or directly administered to rats or cells, respectively. Streptozoticin ‐induced diabetic rats showed metabolism abnormalities and elevated markers of OS (superoxide dismutase [SOD], malondialdehyde [MDA], gp91phox, Cyt‐Cyto C), enhanced cell apoptosis (Bax/Bcl‐2, Cleaved caspase‐3, TUNEL‐positive cells), together with reduced Akt phosphorylation and increased Foxo1 acetylation. Curcumin attenuated the myocardial dysfunction, OS and apoptosis in the heart of diabetic rats. Curcumin treatment also enhanced phosphorylation of Akt and inhibited acetylation of Foxo1. These results strongly suggest that apoptosis was increased in the heart of diabetic rats, and curcumin played a role in diabetic cardiomyopathy treatment by modulating the Sirt1‐Foxo1 and PI3K‐Akt pathways.
Collapse
Affiliation(s)
- Bin-Cheng Ren
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu-Fei Zhang
- State Key Laboratory of Crop Stress Biology for Arid Areas and College of Life Sciences, Northwest A&F University, Yangling, China
| | - Shan-Shan Liu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Jing Cheng
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Yang
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Guang Cui
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin-Rui Zhao
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Zhao
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Min-Feng Hao
- Department of Neurology, Xi'an Central Hospital, Xi'an, China
| | - Meng-Dan Li
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuan-Yuan Tie
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Qu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xue-Yi Li
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|