1
|
Chen S, Yang J, Liu F. ROS-responsive nanomicelles encapsulating celastrol ameliorate pressure overload-induced cardiac hypertrophy by regulating the NF-κB signaling pathway. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:545-563. [PMID: 39400040 DOI: 10.1080/09205063.2024.2411095] [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: 06/11/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
Celastrol (CEL) belongs to the group of non-steroidal immunosuppressants with the potential to improve cardiac hypertrophy (CH). However, the poor biocompatibility and low bioavailability of CEL limit its in vivo application. This study was aimed to develop a targeted drug delivery system that can efficiently and safely deliver CEL to target tissues, providing a research basis for the application of CEL in CH therapy. A novel ROS-sensitive drug-loaded nanomicelle, dodecanoic acid (DA)-phenylboronic acid pinacol ester-dextran polymer encapsulating CEL (DBD@CEL), was synthesized using chemical synthesis. Then, the morphology, particle size, drug-loaded content, and ROS-responsive release behavior of DBD@CEL were studied. Pharmacokinetics and biocompatibility were evaluated using healthy mice. Finally, the ability and mechanism of DBD@CEL in improving CH in vivo were investigated using a mouse CH model. DBD@CEL was successfully prepared with a drug loading of 18.9%. It exhibited excellent stability with an average particle size of 110.0 ± 1.7 nm. Within 48 h, DBD@CEL released only 19.4% in the absence of H2O2, while in the presence of 1 mM H2O2, the release rate increased to 71.5%. Biocompatibility studies indicated that DBD@CEL did not cause blood cell hemolysis, had no impact on normal organs, and did not result in abnormal blood biochemical indicators, demonstrating excellent biocompatibility. In vivo studies revealed that DBD@CEL regulated the activation of NF-κB signaling, inhibits pyroptosis and oxidative stress, and thereby ameliorates CH. The ROS-responsive DBD@CEL nanodrug delivery system enhances the therapeutic activity of CEL for CH, providing a promising drug delivery system for the clinical treatment of CH.
Collapse
Affiliation(s)
- Shanjiang Chen
- Department of Cardiology, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Jianjian Yang
- Department of Cardiology, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Fuli Liu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Zhao L, Zhang J, Song Q, Dai C, Qin Y, Li A. Comprehensive analysis of disulfidptosis-related genes and the immune microenvironment in heart failure. Front Cell Dev Biol 2025; 12:1516898. [PMID: 39897078 PMCID: PMC11782221 DOI: 10.3389/fcell.2024.1516898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/31/2024] [Indexed: 02/04/2025] Open
Abstract
Background Heart failure (HF) is a prevalent cardiovascular disease that currently lacks effective treatment options due to its intricate pathogenesis. A recent study has linked disulfidoptosis, a novel form of cell demise, with the development of a range of diseases. Nonetheless, the effect of disulfidoptosis on the immune microenvironment of HF is not well comprehended. In this paper, bioinformatics analysis was performed to investigate how disulfidptosis-related genes (DRGs) affect the immune microenvironment of HF. Methods The expression of four DRGs was initially examined using bulk RNA-Seq and single-cell RNA sequencing data. A predictive model was subsequently developed. Consensus clustering was used to distinguish between the two clusters of DRGs. The effect of these DRGs on the characteristics of the immune microenvironment was further explored, such as infiltrating immune cells, immune response gene sets, and HLAs genes. Results All four DRGs were dysregulated in HF samples. The predictive model based on these four DRGs effectively differentiated between HF patients and healthy individuals, which was validated in the experiment. These four DRGs were strongly associated with the abundance of infiltrating monocytes. Moreover, our analysis identified two distinct clusters of DRGs and these clusters exhibited differences in terms of immune cell abundance, immune response, and HLA gene expression. The biological functions associated with these differences were also revealed. Conclusion Our discovery underscores the pivotal role of DRGs in shaping the diversity and intricacy of the immune microenvironment in HF.
Collapse
Affiliation(s)
- Linna Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Juanjuan Zhang
- Faculty of Nursing, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| | - Qiuhang Song
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| | - Cheng Dai
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yiping Qin
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Aiying Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
Zhang T, Li L, Mo X, Xie S, Liu S, Zhao N, Zhang H, Chen S, Zeng X, Wang S, Deng W, Tang Q. Matairesinol blunts adverse cardiac remodeling and heart failure induced by pressure overload by regulating Prdx1 and PI3K/AKT/FOXO1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156054. [PMID: 39306883 DOI: 10.1016/j.phymed.2024.156054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Pathological cardiac remodeling is a critical process leading to heart failure, characterized primarily by inflammation and apoptosis. Matairesinol (Mat), a key chemical component of Podocarpus macrophyllus resin, exhibits a wide range of pharmacological activities, including anti-hydatid, antioxidant, antitumor, and anti-inflammatory effects. PURPOSE This study aims to investigate whether Matairesinol alleviate cardiac hypertrophy and remodeling caused by pressure overload and to elucidate its mechanism of action. METHODS An in vitro pressure loading model was established using neonatal rat cardiomyocytes treated with angiotensin Ⅱ, while an in vivo model was created using C57 mice subjected to transverse aortic constriction (TAC). To activate the PI3K/Akt/FoxO1 pathway, Ys-49 was employed. Moreover, small interfering RNA (siRNA) and short hairpin RNA (shRNA) were utilized to silence Prdx1 expression both in vitro and in vivo. Various techniques, including echocardiography, wheat germ agglutinin (WGA) staining, HE staining, PSR staining, and Masson trichrome staining, were used to assess cardiac function, cardiomyocyte cross-sectional area, and fibrosis levels in rats. Apoptosis in myocardial tissue and in vitro was detected by TUNEL assay, while reactive oxygen species (ROS) content in tissues and cells was measured using DHE staining. Furthermore, the affinity of Prdx1 with Mat and PI3K was analyzed using computer-simulated molecular docking. Western blotting and RT-PCR were utilized to evaluate Prdx1 levels and proteins related to apoptosis and oxidative stress, as well as the mRNA levels of cardiac hypertrophy and fibrosis-related indicators. RESULTS Mat significantly alleviated cardiac hypertrophy and fibrosis induced by TAC, preserved cardiac function, and markedly reduced cardiomyocyte apoptosis and oxidative damage. In vitro, mat attenuated ang Ⅱ - induced hypertrophy of nrvms and activation of neonatal rat fibroblasts. Notably, activation of the PI3K/Akt/FoxO1 pathway and downregulation of Prdx1 expression were observed in TAC mice; however, these effects were reversed by Mat treatment. Furthermore, Prdx1 knockdown activated the PI3K/Akt/FoxO1 pathway, leading to exacerbation of the disease. Molecular docking indicated that Molecular docking indicated that Mat upregulated Prdx1 expression by binding to it, thereby inhibiting the PI3K/Akt/FoxO1 pathway and protecting the heart by restoring Prdx1 expression levels. CONCLUSION Matairesinol alleviates pressure overload-induced cardiac remodeling both in vivo and in vitro by upregulating Prdx1 expression and inhibiting the PI3K/Akt/FoxO1 pathway. This study highlights the therapeutic potential of Matairesinol in the treatment of cardiac hypertrophy and remodeling, providing a promising avenue for future research and clinical application.
Collapse
Key Words
- ANP, atrial natriuretic peptide
- Abbreviations: MAT, matairesinol
- BNP, B-type natriuretic peptide
- Cardiac fibrosis
- Cardiac hypertrophy
- Cardiac remodeling
- LV, left ventricular
- LVEDd, left ventricular end-diastolic dimension
- LVEF, left ventricular ejection fraction
- Matairesinol
- NRCFS, neonatal rat cardiac fibroblasts
- PRDX 1
- PRDX1, peroxiredoxin 1
- ROS, reactive oxygen species
- Sh-RNA, short-hairpin RNA
- Si-RNA, small interfering RNA
- TAC, transverse aortic contraction
- β-MHC, Β-myosin heavy chain
Collapse
Affiliation(s)
- Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Xiaotong Mo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Shiqiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Xiaofeng Zeng
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Shasha Wang
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
4
|
Saedi S, Tan Y, Watson SE, Wintergerst KA, Cai L. Potential pathogenic roles of ferroptosis and cuproptosis in cadmium-induced or exacerbated cardiovascular complications in individuals with diabetes. Front Endocrinol (Lausanne) 2024; 15:1461171. [PMID: 39415790 PMCID: PMC11479913 DOI: 10.3389/fendo.2024.1461171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetes and its complications are major diseases that affect human health. Diabetic cardiovascular complications such as cardiovascular diseases (CVDs) are the major complications of diabetes, which are associated with the loss of cardiovascular cells. Pathogenically the role of ferroptosis, an iron-dependent cell death, and cuproptosis, a copper-dependent cell death has recently been receiving attention for the pathogenesis of diabetes and its cardiovascular complications. How exposure to environmental metals affects these two metal-dependent cell deaths in cardiovascular pathogenesis under diabetic and nondiabetic conditions remains largely unknown. As an omnipresent environmental metal, cadmium exposure can cause oxidative stress in the diabetic cardiomyocytes, leading to iron accumulation, glutathione depletion, lipid peroxidation, and finally exacerbate ferroptosis and disrupt the cardiac. Moreover, cadmium-induced hyperglycemia can enhance the circulation of advanced glycation end products (AGEs). Excessive AGEs in diabetes promote the upregulation of copper importer solute carrier family 31 member 1 through activating transcription factor 3/transcription factor PU.1, thereby increasing intracellular Cu+ accumulation in cardiomyocytes and disturbing Cu+ homeostasis, leading to a decline of Fe-S cluster protein and reactive oxygen species accumulation in cardiomyocytes mitochondria. In this review, we summarize the available evidence and the most recent advances exploring the underlying mechanisms of ferroptosis and cuproptosis in CVDs and diabetic cardiovascular complications, to provide critical perspectives on the potential pathogenic roles of ferroptosis and cuproptosis in cadmium-induced or exacerbated cardiovascular complications in diabetic individuals.
Collapse
Affiliation(s)
- Saman Saedi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Sara E. Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children’s Hospital, Louisville, KY, United States
| | - Kupper A. Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children’s Hospital, Louisville, KY, United States
- The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, United States
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Wendy Novak Diabetes Institute, Norton Children’s Hospital, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
- The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
5
|
Zhao L, Qin Y, Liu Y, An L, Liu W, Zhang C, Song Q, Dai C, Zhang J, Li A. The total xanthones extracted from Gentianella acuta alleviates HFpEF by activating the IRE1α/Xbp1s pathway. J Cell Mol Med 2024; 28:e18466. [PMID: 38847482 PMCID: PMC11157675 DOI: 10.1111/jcmm.18466] [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: 04/09/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/10/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome characterized by pulmonary and systemic congestion resulting from left ventricular diastolic dysfunction and increased filling pressure. Currently, however, there is no evidence on effective pharmacotherapy for HFpEF. In this study, we aimed to investigate the therapeutic effect of total xanthones extracted from Gentianella acuta (TXG) on HFpEF by establishing an high-fat diet (HFD) + L-NAME-induced mouse model. Echocardiography was employed to assess the impact of TXG on the cardiac function in HFpEF mice. Haematoxylin and eosin staining, wheat germ agglutinin staining, and Masson's trichrome staining were utilized to observe the histopathological changes following TXG treatment. The results demonstrated that TXG alleviated HFpEF by reducing the expressions of genes associated with myocardial hypertrophy, fibrosis and apoptosis. Furthermore, TXG improved cardiomyocyte apoptosis by inhibiting the expression of apoptosis-related proteins. Mechanistic investigations revealed that TXG could activate the inositol-requiring enzyme 1α (IRE1α)/X-box-binding protein 1 (Xbp1s) signalling pathway, but the knockdown of IRE1α using the IRE1α inhibitor STF083010 or siRNA-IRE1α impaired the ability of TXG to ameliorate cardiac remodelling in HFpEF models. In conclusion, TXG alleviates myocardial hypertrophy, fibrosis and apoptosis through the activation of the IRE1α/Xbp1s signalling pathway, suggesting its potential beneficial effects on HFpEF patients.
Collapse
Affiliation(s)
- Linna Zhao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Yiping Qin
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Yangong Liu
- The First Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Liping An
- College of Basic MedicineHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Weizhe Liu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
- College of Basic MedicineHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Chuang Zhang
- Department of TechnologyHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Qiuhang Song
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Cheng Dai
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Juanjuan Zhang
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
- Faculty of NursingHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Aiying Li
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
- College of Basic MedicineHebei University of Chinese MedicineShijiazhuangHebeiChina
| |
Collapse
|
6
|
Chen J, Yang X, Li W, Lin Y, Lin R, Cai X, Yan B, Xie B, Li J. Potential molecular and cellular mechanisms of the effects of cuproptosis-related genes in the cardiomyocytes of patients with diabetic heart failure: a bioinformatics analysis. Front Endocrinol (Lausanne) 2024; 15:1370387. [PMID: 38883603 PMCID: PMC11176466 DOI: 10.3389/fendo.2024.1370387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
Background Diabetes mellitus is an independent risk factor for heart failure, and diabetes-induced heart failure severely affects patients' health and quality of life. Cuproptosis is a newly defined type of programmed cell death that is thought to be involved in the pathogenesis and progression of cardiovascular disease, but the molecular mechanisms involved are not well understood. Therefore, we aimed to identify biomarkers associated with cuproptosis in diabetes mellitus-associated heart failure and the potential pathological mechanisms in cardiomyocytes. Materials Cuproptosis-associated genes were identified from the previous publication. The GSE26887 dataset was downloaded from the GEO database. Methods The consistency clustering was performed according to the cuproptosis gene expression. Differentially expressed genes were identified using the limma package, key genes were identified using the weighted gene co-expression network analysis(WGCNA) method, and these were subjected to immune infiltration analysis, enrichment analysis, and prediction of the key associated transcription factors. Consistency clustering identified three cuproptosis clusters. The differentially expressed genes for each were identified using limma and the most critical MEantiquewhite4 module was obtained using WGCNA. We then evaluated the intersection of the MEantiquewhite4 output with the three clusters, and obtained the key genes. Results There were four key genes: HSDL2, BCO2, CORIN, and SNORA80E. HSDL2, BCO2, and CORIN were negatively associated with multiple immune factors, while SNORA80E was positively associated, and T-cells accounted for a major proportion of this relationship with the immune system. Four enriched pathways were found to be associated: arachidonic acid metabolism, peroxisomes, fatty acid metabolism, and dorsoventral axis formation, which may be regulated by the transcription factor MECOM, through a change in protein structure. Conclusion HSDL2, BCO2, CORIN, and SNORA80E may regulate cardiomyocyte cuproptosis in patients with diabetes mellitus-associated heart failure through effects on the immune system. The product of the cuproptosis-associated gene LOXL2 is probably involved in myocardial fibrosis in patients with diabetes, which leads to the development of cardiac insufficiency.
Collapse
Affiliation(s)
- Jinhao Chen
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Xu Yang
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Weiwen Li
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Ying Lin
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Run Lin
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Xianzhen Cai
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Baoxin Yan
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Bin Xie
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jilin Li
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
7
|
Zhou Y, Yan Z, Pang Y, Jiang Y, Zhuang R, Zhang S, Nurmamat A, Xiu M, Li D, Zhao L, Liu X, Li Q, Han Y. Exploring the Multiple Roles of Notch1 in Biological Development: An Analysis and Study Based on Phylogenetics and Transcriptomics. Int J Mol Sci 2024; 25:611. [PMID: 38203782 PMCID: PMC10778765 DOI: 10.3390/ijms25010611] [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: 11/09/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
At present, there is a research gap concerning the specific functions and mechanisms of the Notch gene family and its signaling pathway in jawless vertebrates. In this study, we identified a Notch1 homologue (Lr. Notch1) in the Lethenteron reissneri database. Through bioinformatics analysis, we identified Lr. Notch1 as the likely common ancestor gene of the Notch gene family in higher vertebrates, indicating a high degree of conservation in the Notch gene family and its signaling pathways. To validate the biological function of Lr. Notch1, we conducted targeted silencing of Lr. Notch1 in L. reissneri and analyzed the resultant gene expression profile before and after silencing using transcriptome analysis. Our findings revealed that the silencing of Lr. Notch1 resulted in differential expression of pathways and genes associated with signal transduction, immune regulation, and metabolic regulation, mirroring the biological function of the Notch signaling pathway in higher vertebrates. This article systematically elucidated the origin and evolution of the Notch gene family while also validating the biological function of Lr. Notch1. These insights offer valuable clues for understanding the evolution of the Notch signaling pathway and establish a foundation for future research on the origin of the Notch signaling pathway, as well as its implications in human diseases and immunomodulation.
Collapse
Affiliation(s)
- Yuesi Zhou
- Key Research Base of Humanities and Social Sciences of Ministry of Education, Institute of Marine Sustainable Development, Liaoning Normal University, Dalian 116029, China;
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Zihao Yan
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ya Pang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Yao Jiang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ruyu Zhuang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Shuyuan Zhang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ayqeqan Nurmamat
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Min Xiu
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ding Li
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Liang Zhao
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Xin Liu
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Qingwei Li
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yinglun Han
- Key Research Base of Humanities and Social Sciences of Ministry of Education, Institute of Marine Sustainable Development, Liaoning Normal University, Dalian 116029, China;
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
8
|
Wei M, Lu Z, Zhang H, Fan X, Zhang X, Jiang B, Li J, Xue M. Aspirin and Celecoxib Regulate Notch1/Hes1 Pathway to Prevent Pressure Overload-Induced Myocardial Hypertrophy. Int Heart J 2024; 65:475-486. [PMID: 38825493 DOI: 10.1536/ihj.23-614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study aimed to investigate the molecular mechanisms underlying the protective effects of cyclooxygenase (cox) inhibitors against myocardial hypertrophy.Rat H9c2 cardiomyocytes were induced by mechanical stretching. SD rats underwent transverse aortic constriction to induce pressure overload myocardial hypertrophy. Rats were subjected to echocardiography and tail arterial pressure in 12W. qPCR and western blot were used to detect the expression of Notch-related signaling. The inflammatory factors were tested by ELISA in serum, heart tissue, and cell culture supernatant.Compared with control, levels of pro-inflammatory cytokines IL-6, TNF-α, and IL-1β were increased and anti-inflammatory cytokine IL-10 was reduced in myocardial tissues and serum of rat models. Levels of Notch1 and Hes1 were reduced in myocardial tissues. However, cox inhibitor treatment (aspirin and celecoxib), the improvement of exacerbated myocardial hypertrophy, fibrosis, dysfunction, and inflammation was parallel to the activation of Notch1/Hes1 pathway. Moreover, in vitro experiments showed that, in cardiomyocyte H9c2 cells, application of ~20% mechanical stretching activated inflammatory mediators (IL-6, TNF-α, and IL-1β) and hypertrophic markers (ANP and BNP). Moreover, expression levels of Notch1 and Hes1 were decreased. These changes were effectively alleviated by aspirin and celecoxib.Cox inhibitors may protect heart from hypertrophy and inflammation possibly via the Notch1/Hes1 signaling pathway.
Collapse
Affiliation(s)
- Minghui Wei
- School of Basic Medicine, Inner Mongolia Medical University
| | - Ziyu Lu
- School of Basic Medicine, Inner Mongolia Medical University
| | - Haifeng Zhang
- Office of Academic Affairs, Inner Mongolia Medical University
| | - Xiaomei Fan
- Department of Physiology, Inner Mongolia Medical University
| | - Xin Zhang
- Department of Physiology, Inner Mongolia Medical University
| | - Bihui Jiang
- School of Basic Medicine, Inner Mongolia Medical University
| | - Jianying Li
- School of Basic Medicine, Inner Mongolia Medical University
| | - Mingming Xue
- Office of Academic Affairs, Inner Mongolia Medical University
| |
Collapse
|
9
|
Nie X, Xie R, Fan J, Wang DW. LncRNA MIR217HG aggravates pressure-overload induced cardiac remodeling by activating miR-138/THBS1 pathway. Life Sci 2024; 336:122290. [PMID: 38013141 DOI: 10.1016/j.lfs.2023.122290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023]
Abstract
AIM Cardiac hypertrophy and fibrosis are associated with cardiac remodeling and heart failure. We have previously shown that miRNA-217, embedded within the third intron of MIR217HG, aggravates pressure overload-induced cardiac hypertrophy by targeting phosphatase and tensin homolog. However, whether the MIR217HG transcript itself plays a role in cardiac remodeling remains unknown. METHODS Real-time PCR assays and RNA in situ hybridization were performed to detect MIR217HG expression. Lentiviruses and adeno-associated viruses with a cardiac-specific promoter (cTnT) were used to control MIR217HG expression in vitro and in vivo. Transverse aortic constriction (TAC) surgery was performed to develop cardiac remodeling models. Cardiac structure and function were analyzed using echocardiography and invasive pressure-volume analysis. KEY FINDINGS MIR217HG expression was increased in patients with heart failure. MIR217HG overexpression aggravated pressure-overload-induced myocyte hypertrophy and fibrosis both in vivo and in vitro, whereas MIR217HG knockdown reversed these phenotypes. Mechanistically, MIR217HG increased THBS1 expression by sponging miR-138. MiR-138 recognized the 3'UTR of THBS1 and repressed THBS1 expression in the absence of MIR217HG. Silencing THBS1 expression reversed MIR217HG-induced cardiac hypertrophy and remodeling. CONCLUSION MIR217HG acts as a potent inducer of cardiac remodeling that may contribute to heart failure by activating the miR-138/THBS1 pathway.
Collapse
Affiliation(s)
- Xiang Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Rong Xie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Jiahui Fan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
10
|
Han YC, Wang L, Zhang YD, Zhou AJ, Wang ZP, Dong WH, Wang JP, Wang T, Zou J. Mechanisms Underlying the Therapeutic Effects of Nicotinamide Mononucleotide in Treating High-fat Diet-induced Hypertrophic Cardiomyopathy based on GEO Datasets, Network Pharmacology, and Molecular Docking. Curr Pharm Des 2024; 30:3054-3070. [PMID: 39171590 DOI: 10.2174/0113816128311226240730080713] [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: 03/20/2024] [Revised: 06/16/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND The beneficial effects of nicotinamide mononucleotide (NMN) on heart disease have been reported, but the effects of NMN on high-fat diet-induced hypertrophic cardiomyopathy (HCM) and its mechanisms of action are unclear. In this study, we systematically explored the effects and mechanism of action of NMN in HCM using network pharmacology and molecular docking. METHODS Active targets of NMN were obtained from SWISS, CNKI, PubMed, DrugBank, BingingDB, and ZINC databases. HCM-related targets were retrieved from GEO datasets combined with GeneCards, OMIM, PharmGKB, and DisGeNET databases. A Protein-protein Interaction (PPI) network was built to screen the core targets. DAVID was used for GO and KEGG pathway enrichment analyses. The tissue and organ distribution of targets was evaluated. Interactions between potential targets and active compounds were assessed by molecular docking. A molecular dynamics simulation was conducted for the optimal core protein-compound complexes obtained by molecular docking. RESULTS In total, 265 active targets of NMN and 3918 potential targets of HCM were identified. A topological analysis of the PPI network revealed 10 core targets. GO and KEGG pathway enrichment analyses indicated that the effects of NMN were mediated by genes related to inflammation, apoptosis, and oxidative stress, as well as the FOXO and PI3K-Akt signaling pathways. Molecular docking and molecular dynamics simulations revealed good binding ability between the active compounds and screened targets. CONCLUSION The possible targets and pathways of NMN in the treatment of HCM have been successfully predicted by this investigation. It provides a novel approach for further investigation into the molecular processes of NMN in HCM treatment.
Collapse
Affiliation(s)
- Yuan-Chun Han
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Li Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yi-Dan Zhang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ao-Jia Zhou
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zi-Ping Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Wen-Huan Dong
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jian-Peng Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ting Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Academy of Nutrition and Health, Institute of Advanced Pharmaceutical Technology, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
- Wuhan Asia General Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430056, China
| | - Jun Zou
- Department of Pharmacy, Hainan Women and Children's Medical Center, Haikou, Hainan Province 570100, China
| |
Collapse
|
11
|
Hu L, Gao D, Lv H, Lian L, Wang M, Wang Y, Xie Y, Zhang J. Finding New Targets for the Treatment of Heart Failure: Endoplasmic Reticulum Stress and Autophagy. J Cardiovasc Transl Res 2023; 16:1349-1356. [PMID: 37432587 DOI: 10.1007/s12265-023-10410-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/28/2023] [Indexed: 07/12/2023]
Abstract
Heart failure is a progressive disease with an annual mortality rate of about 10% and is the end-stage stage of various heart diseases, which places a huge socioeconomic burden on the healthcare system. The development of heart failure has received increasing attention as a potential way to improve the treatment of this disease. Many studies have shown that endoplasmic reticulum stress and autophagy play an important role in the occurrence and development of heart failure. With the in-depth study of endoplasmic reticulum stress and autophagy, both are considered promising targets for pharmacological interventions to treat heart failure, but the mechanism of heart failure between the two is not clear. This review will highlight the effects of endoplasmic reticulum stress, autophagy, and their interactions in the development and development of heart failure, thereby helping to provide direction for the future development of targeted therapies for patients with heart failure. CLINICAL RELEVANCE: This study explored the new targets for the treatment of heart failure: endoplasmic reticulum stress and autophagy. Targeted drug therapy for endoplasmic reticulum stress and autophagy is expected to provide a new intervention target for the treatment of heart failure.
Collapse
Affiliation(s)
- Leilei Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Dongjie Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Hao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lu Lian
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Mingyang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yingyu Xie
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China.
| |
Collapse
|
12
|
Fang Z, Yushanjiang F, Wang G, Zheng X, Jiang X. Germacrone mitigates cardiac remodeling by regulating PI3K/AKT-mediated oxidative stress, inflammation, and apoptosis. Int Immunopharmacol 2023; 124:110876. [PMID: 37683399 DOI: 10.1016/j.intimp.2023.110876] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023]
Abstract
Cardiac remodeling is a common consequence of cardiovascular diseases and is closely associated with oxidative stress, inflammation, and apoptosis. Germacrone, a bioactive compound present in Rhizoma curcuma, has been shown to possess anti-oxidative, anti-inflammatory, and anti-apoptotic properties. The aim of this study was to investigate the protective effect of germacrone against cardiac remodeling. Here, C57BL/6 mice were subcutaneous injection with isoproterenol (ISO) once daily for two weeks and were concurrent intragastric injection of germacrone. In vitro, neonatal rat cardiomyocytes (NRCMs) were used to verify the protective effect of germacrone on ISO-induced cardiac injury. Our findings indicated that ISO induce oxidative stress, inflammation, and apoptosis in vivo and in vitro, while germacrone treatment significantly attenuates these effects, thereby attenuating myocardium remodeling and cardiac dysfunction. Mechanistically, germacrone reduced cardiac remodeling-induced activation of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, and the cardioprotective effects of germacrone were abrogated by a PI3K agonist. In conclusion, our results suggest that germacrone attenuates oxidative stress, inflammation, and apoptosis in cardiac remodeling by inhibiting the PI3K/AKT pathway, and may therefore represent a promising therapeutic approach for the treatment of cardiac remodeling.
Collapse
Affiliation(s)
- Zhao Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Feierkaiti Yushanjiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Guangji Wang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxin Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
13
|
Zhou D, Liu W, Zhang J, Dong Y, Wu J, Zhang Y, Dai C, Zhang T, Yang G, Zhang Y, Li A. Bellidifolin ameliorates isoprenaline-induced cardiac hypertrophy by the Nox4/ROS signalling pathway through inhibiting BRD4. Cell Death Discov 2023; 9:279. [PMID: 37528096 PMCID: PMC10394041 DOI: 10.1038/s41420-023-01563-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 08/03/2023] Open
Abstract
To date, there is no effective therapy for pathological cardiac hypertrophy, which can ultimately lead to heart failure. Bellidifolin (BEL) is an active xanthone component of Gentianella acuta (G. acuta) with a protective function for the heart. However, the role and mechanism of BEL action in cardiac hypertrophy remain unknown. In this study, the mouse model of cardiac hypertrophy was established by isoprenaline (ISO) induction with or without BEL treatment. The results showed that BEL alleviated cardiac dysfunction and pathological changes induced by ISO in the mice. The expression of cardiac hypertrophy marker genes, including ANP, BNP, and β-MHC, were inhibited by BEL both in mice and in H9C2 cells. Furthermore, BEL repressed the epigenetic regulator bromodomain-containing protein 4 (BRD4) to reduce the ISO-induced acetylation of H3K122 and phosphorylation of RNA Pol II. The Nox4/ROS/ADAM17 signalling pathway was also inhibited by BEL in a BRD4 dependent manner. Thus, BEL alleviated cardiac hypertrophy and cardiac dysfunction via the BRD4/Nox4/ROS axes during ISO-induced cardiac hypertrophy. These findings clarify the function and molecular mechanism of BEL action in the therapeutic intervention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dingyan Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Weizhe Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China
| | - Juanjuan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Yucui Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jiangli Wu
- Department of Technology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Cheng Dai
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Tingting Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Gaoshan Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Yue Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
- Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China.
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, China.
| | - Aiying Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
- Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, China.
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, China.
| |
Collapse
|
14
|
Li X, Zhang Y, Jin Q, Song Q, Fan C, Jiao Y, Yang C, Chang J, Dong Z, Que Y. Silicate Ions Derived from Calcium Silicate Extract Decelerate Ang II-Induced Cardiac Remodeling. Tissue Eng Regen Med 2023; 20:671-681. [PMID: 36920676 PMCID: PMC10352221 DOI: 10.1007/s13770-023-00523-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Pathological cardiac hypertrophy is one of the main activators of heart failure. Currently, no drug can completely reverse or inhibit the development of pathological cardiac hypertrophy. To this end, we proposed a silicate ion therapy based on extract derived from calcium silicate (CS) bioceramics for the treatment of angiotensin II (Ang II) induced cardiac hypertrophy. METHODS In this study, the Ang II induced cardiac hypertrophy mouse model was established, and the silicate ion extract was injected to mice intravenously. The cardiac function was evaluated by using a high-resolution Vevo 3100 small animal ultrasound imaging system. Wheat germ Agglutinin, Fluo4-AM staining and immunofluorescent staining was conducted to assess the cardiac hypertrophy, intracellular calcium and angiogenesis of heart tissue, respectively. RESULTS The in vitro results showed that silicate ions could inhibit the cell size of cardiomyocytes, reduce cardiac hypertrophic gene expression, including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and β-myosin heavy chain (β-MHC), decrease the content of intracellular calcium induced by Ang II. In vivo experiments in mice confirmed that intravenous injection of silicate ions could remarkably inhibit the cardiac hypertrophy and promote the formation of capillaries, further alleviating Ang II-induced cardiac function disorder. CONCLUSION This study demonstrated that the released silicate ions from CS possessed potential value as a novel therapeutic strategy of pathological cardiac hypertrophy, which provided a new insight for clinical trials.
Collapse
Affiliation(s)
- Xin Li
- School of Mechanical Engineering, Chengdu University, Chengdu, 610106, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Yanxin Zhang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Qishu Jin
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Qiaoyu Song
- School of Mechanical Engineering, Chengdu University, Chengdu, 610106, China
| | - Chen Fan
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Yiren Jiao
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Chen Yang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Jiang Chang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, China
| | - Zhihong Dong
- School of Mechanical Engineering, Chengdu University, Chengdu, 610106, China.
| | - Yumei Que
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| |
Collapse
|
15
|
Zhang F, Wang S, Zhao C, Jiang D, Wang Y, Qi J, Li Y. D-beta-hydroxybutyrate reduced the enhanced cardiac microvascular endothelial FoxO1 to play protective roles in diabetic rats and high glucose-stimulated human cardiac microvascular endothelial cells. Tissue Cell 2023; 81:102031. [PMID: 36701897 DOI: 10.1016/j.tice.2023.102031] [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: 10/05/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
The O subfamily of forkhead (FoxO) 1 may participate in the pathogenesis of diabetic microvascular endothelial injury. However, it is unknown whether D-beta-hydroxybutyrate (BHB) regulates cardiac microvascular endothelial FoxO1 to play protective roles in diabetes. In the study, limb microvascular morphological changes, endothelial distribution of the tight junction protein Claudin-5 and FoxO1, and FoxO1 content in limb tissue from clinical patients were evaluated. Then the effects of BHB on cardiac microvascular morphological changes, cardiac FoxO1 generation and its microvascular distribution in diabetic rats were measured. And the effects of BHB on FoxO1 generation in high glucose (HG)-stimulated human cardiac microvascular endothelial cells (HCMECs) were further analyzed. The results firstly confirmed the enhanced limb microvascular FoxO1 distribution, with reduced Claudin-5 and endothelial injury in clinical patients. Then the elevated FoxO1 generation and its enhanced cardiac microvascular distribution were verified in diabetic rats and HG-stimulated HCMECs. However, BHB inhibited the enhanced cardiac FoxO1 generation and its microvascular distribution with attenuation of endothelial injury in diabetic rats. Furthermore, BHB reduced the HG-stimulated mRNA expression and protein content of FoxO1 in HCMECs. In conclusion, BHB reduced the enhanced cardiac microvascular endothelial FoxO1 to play protective roles in diabetic rats and HG-stimulated HCMECs.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Biochemistry, College of Integrated Chinese and Western Medicine, Hebei Medical University, Hebei, People's Republic of China; Department of Surgery, First Hospital of Hebei Medical University, Hebei, People's Republic of China
| | - Shuai Wang
- Department of Biochemistry, College of Integrated Chinese and Western Medicine, Hebei Medical University, Hebei, People's Republic of China
| | - Chao Zhao
- Department of Biochemistry, College of Integrated Chinese and Western Medicine, Hebei Medical University, Hebei, People's Republic of China
| | - Di Jiang
- Department of Biochemistry, College of Integrated Chinese and Western Medicine, Hebei Medical University, Hebei, People's Republic of China
| | - Yu Wang
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China
| | - Jinsheng Qi
- Department of Biochemistry, College of Integrated Chinese and Western Medicine, Hebei Medical University, Hebei, People's Republic of China.
| | - Yanning Li
- Department of Biochemistry, College of Integrated Chinese and Western Medicine, Hebei Medical University, Hebei, People's Republic of China; Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China.
| |
Collapse
|
16
|
Li T, Wang L, Wu L, Xie Y, Chang M, Wang D, Yi L, Zhu X, Mi M. Integrated Metabolomics and Network Pharmacology Investigation of Cardioprotective Effects of Myricetin after 1-Week High-Intensity Exercise. Nutrients 2023; 15:nu15061336. [PMID: 36986067 PMCID: PMC10054643 DOI: 10.3390/nu15061336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Cardiovascular adverse effects caused by high-intensity exercise (HIE) have become a public health problem of widespread concern. The therapeutic effect and metabolic regulation mechanism of myricetin, a phytochemical with potential therapeutic effects, have rarely been studied. In this study, we established mice models of different doses of myricetin intervention with 1 week of HIE after intervention. Cardiac function tests, serology, and pathological examinations were used to evaluate the protective effect of myricetin on the myocardium. The possible therapeutic targets of myricetin were obtained using an integrated analysis of metabolomics and network pharmacology and verified using molecular docking and RT-qPCR experiments. Different concentrations of myricetin improved cardiac function, significantly reduced the levels of myocardial injury markers, alleviated myocardial ultrastructural damage, reduced the area of ischemia/hypoxia, and increased the content of CX43. We obtained the potential targets and regulated metabolic network of myricetin by combined network pharmacology and metabolomics analysis and validated them by molecular docking and RT-PCR. In conclusion, our findings suggest that myricetin exerts anti-cardiac injury effects of HIE through the downregulation of PTGS2 and MAOB and the upregulation of MAP2K1 and EGFR while regulating the complicated myocardial metabolic network.
Collapse
Affiliation(s)
- Tianyou Li
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Le Wang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Luting Wu
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Yingquan Xie
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Mengyun Chang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Dawei Wang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Long Yi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Xiaohui Zhu
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
- Chongqing Medical Nutrition Research Center, Chongqing 400038, China
- Correspondence: (X.Z.); (M.M.)
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
- Correspondence: (X.Z.); (M.M.)
| |
Collapse
|
17
|
Shi S, Jiang P. Therapeutic potentials of modulating autophagy in pathological cardiac hypertrophy. Biomed Pharmacother 2022; 156:113967. [DOI: 10.1016/j.biopha.2022.113967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
|