1
|
Liu F, Lv LF, Bi FF, Pan QM, Jing ZH, Cui C, Cao M, Yu T, Li J, He YJ, Xiao HW, Tian H, Wu Y, Shan HL, Zhou YH. ITFG2 as a NEDD4-2 inhibitor: Preserving calcium homeostasis to prevent myocardial ischemic injury. Biochem Pharmacol 2024; 230:116597. [PMID: 39477020 DOI: 10.1016/j.bcp.2024.116597] [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: 06/30/2024] [Revised: 09/15/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
This study aimed to investigate the role of ITFG2, a protein highly expressed in cardiac tissues, in myocardial ischemic injury and its potential interactions with NEDD4-2. An in vivo myocardial infarction (MI) model was induced in mice via left anterior descending artery ligation, and ITFG2 expression was modulated using adeno-associated virus AAV9 vectors. Echocardiography was used to assess cardiac function, and primary mouse cardiomyocytes were cultured and subjected to hypoxia. ITFG2 expression was found to be significantly reduced following MI and in hypoxia-treated neonatal cardiomyocytes. Overexpression of ITFG2 improved cardiac contractility, reduced apoptosis, and stabilized calcium levels by inhibiting NEDD4-2-mediated ubiquitination of SERCA2a. Conversely, ITFG2 knockdown exacerbated calcium overload and cardiac dysfunction. Mechanistically, ITFG2 binds to NEDD4-2, decreasing its interaction with SERCA2a and preventing SERCA2a degradation. These findings suggest that ITFG2 acts as a critical inhibitor of NEDD4-2, preserving SERCA2a function and maintaining calcium homeostasis in cardiomyocytes under ischemic conditions. Therefore, ITFG2 may represent a potential therapeutic target for preventing myocardial ischemic injury and improving outcomes in MI patients.
Collapse
Affiliation(s)
- Fu Liu
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Li-Fang Lv
- 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, Heilongjiang 150081, PR China; Department of Physiology, Department of Basic Medicine, Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Fang-Fang Bi
- 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, Heilongjiang 150081, PR China
| | - Qing-Ming Pan
- 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, Heilongjiang 150081, PR China
| | - Ze-Hong Jing
- 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, Heilongjiang 150081, PR China
| | - Chen Cui
- 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, Heilongjiang 150081, PR China
| | - Miao Cao
- 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, Heilongjiang 150081, PR China
| | - Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China
| | - Jin Li
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China; Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Yi-Jie He
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China; Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Hong-Wen Xiao
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Hua Tian
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Yun Wu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, PR China
| | - Hong-Li Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China.
| | - Yu-Hong Zhou
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College of Respiratory Diseases, Xiamen Medical College, Xiamen, Fujian 361023, PR China.
| |
Collapse
|
2
|
Zheng H, Mao X, Fu Z, Chen C, Lv J, Wang Y, Wang Y, Wu H, Li Y, Tan Y, Gao X, Zhao L, Xu X, Zhang B, Song Q. The role of circulating cytokines in heart failure: a bidirectional, two-sample Mendelian randomization study. Front Cardiovasc Med 2024; 11:1332015. [PMID: 39502198 PMCID: PMC11534875 DOI: 10.3389/fcvm.2024.1332015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 07/19/2024] [Indexed: 11/08/2024] Open
Abstract
Background Cytokines play a pivotal role in the progression of heart failure (HF) by modulating inflammatory responses, promoting vasoconstriction, and facilitating endothelial injury. However, it is now difficult to distinguish the causal relationship between HF and cytokines in observational studies. Mendelian randomization (MR) analyses of cytokines probably could enhance our comprehension to the underlying biological processes of HF. Methods This study was to explore the correlation between 41 cytokines with HF at the genetic level by MR analysis. We selected a HF dataset from the Heart Failure Molecular Epidemiology for Therapeutic Targets (HERMES) 2018 and a cytokine dataset from a meta-analysis of cytokine levels in Finns. Two-sample, bidirectional MR analyses were performed using Inverse Variance Weighted (IVW), Weighted Median and MR- egger, and the results were tested for heterogeneity and pleiotropy, followed by sensitivity analysis. Results Genetic prediction of high levels of circulating Macrophage inflammatory pro-tein-1β(MIP-1β) (P = 0.0389), Interferon gamma induced protein 10(IP-10) (P = 0.0029), and Regu-lated on activation, normal T cell expressed and secreted(RANTES) (P = 0.0120) expression was associated with an elevated risk of HF. HF was associated with the increased levels of circulating Interleukin-2 receptor, alpha subunit(IL-2ra) (P = 0.0296), Beta nerve growth fac-tor(β-NGF) (P = 0.0446), Interleukin-17(IL-17) (P = 0.0360), Basic fibroblast growth factor(FGF-basic) (P = 0.0220), Platelet derived growth factor BB(PDGF-BB) (P = 0.0466), and Interferon-gamma(IFN-γ) (P = 0.0222); and with decreased levels of Eotaxin (P = 0.0133). The heterogeneity and pleiotropy of the cytokines were acceptable, except for minor heterogeneity of FGF-basic and IL-17. Conclusion These findings provide compelling evidence for a genetically predictive relationship between cytokines and HF, emphasizing a great potential of targeted modulation of cytokines in slowing the progression of HF. This study draws further conclusions at the genetic level, providing a basis for future large-scale clinical trials.
Collapse
Affiliation(s)
- Haoran Zheng
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinxin Mao
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunmei Chen
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayu Lv
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajiao Wang
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxin Wang
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huaqin Wu
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yvmeng Li
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiya Gao
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Zhao
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xia Xu
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqiao Song
- General Internal Medicine Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Shen J, Shentu J, Zhong C, Huang Q, Duan S. RNA splicing factor RBFOX2 is a key factor in the progression of cancer and cardiomyopathy. Clin Transl Med 2024; 14:e1788. [PMID: 39243148 PMCID: PMC11380049 DOI: 10.1002/ctm2.1788] [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: 04/03/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Alternative splicing of pre-mRNA is a fundamental regulatory process in multicellular eukaryotes, significantly contributing to the diversification of the human proteome. RNA-binding fox-1 homologue 2 (RBFOX2), a member of the evolutionarily conserved RBFOX family, has emerged as a critical splicing regulator, playing a pivotal role in the alternative splicing of pre-mRNA. This review provides a comprehensive analysis of RBFOX2, elucidating its splicing activity through direct and indirect binding mechanisms. RBFOX2 exerts substantial influence over the alternative splicing of numerous transcripts, thereby shaping essential cellular processes such as differentiation and development. MAIN BODY OF THE ABSTRACT Dysregulation of RBFOX2-mediated alternative splicing has been closely linked to a spectrum of cardiovascular diseases and malignant tumours, underscoring its potential as a therapeutic target. Despite significant progress, current research faces notable challenges. The complete structural characterisation of RBFOX2 remains elusive, limiting in-depth exploration beyond its RNA-recognition motif. Furthermore, the scarcity of studies focusing on RBFOX2-targeting drugs poses a hindrance to translating research findings into clinical applications. CONCLUSION This review critically assesses the existing body of knowledge on RBFOX2, highlighting research gaps and limitations. By delineating these areas, this analysis not only serves as a foundational reference for future studies but also provides strategic insights for bridging these gaps. Addressing these challenges will be instrumental in unlocking the full therapeutic potential of RBFOX2, paving the way for innovative and effective treatments in various diseases.
Collapse
Affiliation(s)
- Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| | - Jianqiao Shentu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| | - Chenming Zhong
- Medical Genetics Center, School of MedicineNingbo UniversityNingboChina
| | - Qiankai Huang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of MedicineHangzhou City UniversityHangzhouChina
| |
Collapse
|
4
|
Shang AQ, Yu CJ, Bi X, Jiang WW, Zhao ML, Sun Y, Guan H, Zhang ZR. Blocking CTLA-4 promotes pressure overload-induced heart failure via activating Th17 cells. FASEB J 2024; 38:e23851. [PMID: 39108204 DOI: 10.1096/fj.202400384r] [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: 02/19/2024] [Revised: 06/22/2024] [Accepted: 07/21/2024] [Indexed: 09/17/2024]
Abstract
Targeting cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) with specific antibody offers long-term benefits for cancer immunotherapy but can cause severe adverse effects in the heart. This study aimed to investigate the role of anti-CTLA-4 antibody in pressure overload-induced cardiac remodeling and dysfunction. Transverse aortic constriction (TAC) was used to induce cardiac hypertrophy and heart failure in mice. Two weeks after the TAC treatment, mice received anti-CTLA-4 antibody injection twice a week at a dose of 10 mg/kg body weight. The administration of anti-CTLA-4 antibody exacerbated TAC-induced decline in cardiac function, intensifying myocardial hypertrophy and fibrosis. Further investigation revealed that anti-CTLA-4 antibody significantly elevated systemic inflammatory factors levels and facilitated the differentiation of T helper 17 (Th17) cells in the peripheral blood of TAC-treated mice. Importantly, anti-CTLA-4 mediated differentiation of Th17 cells and hypertrophic phenotype in TAC mice were dramatically alleviated by the inhibition of interleukin-17A (IL-17A) by an anti-IL-17A antibody. Furthermore, the C-X-C motif chemokine receptor 4 (CXCR4) antagonist AMD3100, also reversed anti-CTLA-4-mediated cardiotoxicity in TAC mice. Overall, these results suggest that the administration of anti-CTLA-4 antibody exacerbates pressure overload-induced heart failure by activating and promoting the differentiation of Th17 cells. Targeting the CXCR4/Th17/IL-17A axis could be a potential therapeutic strategy for mitigating immune checkpoint inhibitors-induced cardiotoxicity.
Collapse
Affiliation(s)
- An-Qi Shang
- Departments of Cardiology and Critical Care Medicine, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang-Jiang Yu
- Departments of Pharmacy and Cardiology, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Bi
- Departments of Cardiology and Critical Care Medicine, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei-Wei Jiang
- Departments of Cardiology and Critical Care Medicine, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ming-Luan Zhao
- Departments of Pharmacy and Cardiology, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Sun
- Departments of Cardiology and Critical Care Medicine, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong Guan
- Departments of Cardiology and Critical Care Medicine, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhi-Ren Zhang
- Departments of Cardiology and Critical Care Medicine, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Departments of Pharmacy and Cardiology, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin Medical University Cancer Hospital, Harbin, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Zhou J, Wu C, Zhao M. TAOK1-mediated regulation of the YAP/TEAD pathway as a potential therapeutic target in heart failure. PLoS One 2024; 19:e0308619. [PMID: 39121041 PMCID: PMC11315341 DOI: 10.1371/journal.pone.0308619] [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: 02/29/2024] [Accepted: 07/26/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND This study aimed to determine the roles of interleukin (IL)-17, TAO kinase 1 (TAOK1), and NOD-like receptor protein 3 (NLRP3) in cardiomyocyte pyroptosis and proliferation. METHODS The IL-17-treated H9C2 cells were used as in vitro heart failure (HF) models. These cells were subjected to TAOK1 overexpression or knockdown and treated with BMS-986299 (NLRP3 inflammasome agonist), MCC950 (NLRP3 inflammasome inhibitor), or verteporfin (Yes-associated protein [YAP] inhibitor). Thereafter, their pyroptosis, proliferative capacity, and gene and protein expression levels were detected. Doxorubicin-induced HF rats were used as in vivo models and subjected to TAOK1 overexpression. Thereafter, their myocardial pathology, NLRP3 inflammasome-mediated pyroptosis, and YAP/TEAD pathway function were evaluated. RESULTS IL-17 treatment increased the pyroptosis and decreased the proliferative capacity of H9C2 cells. Additionally, IL-17 treatment inducedto the activation of the NLRP3 inflammasomes and inhibition of the YAP/TEAD pathway in the H9C2 cells. Moreover, the IL-17-mediated effects on the H9C2 cells were alleviated by TAOK1 overexpression and augmented by TAOK1 knockdown. Furthermore, treatment with BMS-986299 or verteporfin affected the pyroptosis, proliferative capacity, and NLRP3 inflammasome activation of the H9C2 cells independently of TAOK1 expression. In the doxorubicin-induced HF rat model, TAOK1 overexpression mitigated myocardial injury, suppressed NLRP3 inflammasome pathway activation, and restored the YAP/TEAD pathway activity. CONCLUSION TAOK1 played a crucial role in regulating IL-17-mediated increase in the pyroptosis and decrease in the proliferation of cardiomyocytes by regulating the activities of the NLRP3 inflammasomes and the YAP/TEAD pathway.
Collapse
Affiliation(s)
- Jiani Zhou
- Department of General Practice, Ningbo Medical Treatment Center Li Huili Hospital, Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Chaoqun Wu
- Department of General Practice, Ningbo Medical Treatment Center Li Huili Hospital, Affiliated to Ningbo University, Ningbo, Zhejiang, China
| | - Miaohui Zhao
- Department of General Practice, Ningbo Medical Treatment Center Li Huili Hospital, Affiliated to Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
6
|
Yang H, Xuan L, Wang S, Luo H, Duan X, Guo J, Cui S, Xin J, Hao J, Li X, Chen J, Sun F, Hu X, Li S, Zhang Y, Jiao L, Yang B, Sun L. LncRNA CCRR maintains Ca 2+ homeostasis against myocardial infarction through the FTO-SERCA2a pathway. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1601-1619. [PMID: 38761356 DOI: 10.1007/s11427-023-2527-5] [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: 12/08/2023] [Accepted: 01/11/2024] [Indexed: 05/20/2024]
Abstract
Cardiac conduction regulatory RNA (CCRR) has been documented as an antiarrhythmic lncRNA in our earlier investigation. This study aimed to evaluate the effects of CCRR on SERCA2a and the associated Ca2+ homeostasis in myocardial infarction (MI). Overexpression of CCRR via AAV9-mediated delivery not only partially reversed ischemia-induced contractile dysfunction but also alleviated abnormal Ca2+ homeostasis and reduced the heightened methylation level of SERCA2a following MI. These effects were also observed in CCRR over-expressing transgenic mice. A conserved sequence domain of CCRR mimicked the protective function observed with the full length. Furthermore, silencing CCRR in healthy mice led to intracellular Ca2+ overloading of cardiomyocytes. CCRR increased SERCA2a protein stability by upregulating FTO expression. The direct interaction between CCRR and FTO protein was characterized by RNA-binding protein immunoprecipitation (RIP) analysis and RNA pulldown experiments. Activation of NFATc3 was identified as an upstream mechanism responsible for CCRR downregulation in MI. This study demonstrates that CCRR is a protective lncRNA that acts by maintaining the function of FTO, thereby reducing the m6A RNA methylation level of SERCA2a, ultimately preserving calcium homeostasis for myocardial contractile function in MI. Therefore, CCRR may be considered a promising therapeutic strategy with a beneficial role in cardiac pathology.
Collapse
Affiliation(s)
- Hua Yang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Lina Xuan
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Shengjie Wang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Huishan Luo
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaomeng Duan
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jianjun Guo
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Shijia Cui
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jieru Xin
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Junwei Hao
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiufang Li
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jun Chen
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Feihan Sun
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaolin Hu
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Siyun Li
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ying Zhang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Lei Jiao
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Lihua Sun
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
7
|
Zhang C, Song Y, Cen L, Huang C, Zhou J, Lian J. Screening of Secretory Proteins Linking Major Depressive Disorder with Heart Failure Based on Comprehensive Bioinformatics Analysis and Machine Learning. Biomolecules 2024; 14:793. [PMID: 39062507 PMCID: PMC11275063 DOI: 10.3390/biom14070793] [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: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) plays a crucial role in the occurrence of heart failure (HF). This investigation was undertaken to explore the possible mechanism of MDD's involvement in HF pathogenesis and identify candidate biomarkers for the diagnosis of MDD with HF. METHODS GWAS data for MDD and HF were collected, and Mendelian randomization (MR) analysis was performed to investigate the causal relationship between MDD and HF. Differential expression analysis (DEA) and WGCNA were used to detect HF key genes and MDD-associated secretory proteins. Protein-protein interaction (PPI), functional enrichment, and cMAP analysis were used to reveal potential mechanisms and drugs for MDD-related HF. Then, four machine learning (ML) algorithms (including GLM, RF, SVM, and XGB) were used to screen candidate biomarkers, construct diagnostic nomograms, and predict MDD-related HF. Furthermore, the MCPcounter algorithm was used to explore immune cell infiltration in HF, and MR analysis was performed to explore the causal effect of immunophenotypes on HF. Finally, the validation of the association of MDD with reduced left ventricular ejection fraction (LVEF) and the performance assessment of diagnostic biomarkers was accomplished based on animal models mimicking MDD. RESULTS The MR analysis showed that the MDD was linked to an increased risk of HF (OR = 1.129, p < 0.001). DEA combined with WGCNA and secretory protein gene set identified 315 HF key genes and 332 MDD-associated secretory proteins, respectively. Through PPI and MCODE analysis, 78 genes were pinpointed as MDD-related pathogenic genes for HF. The enrichment analysis revealed that these genes were predominantly enriched in immune and inflammatory regulation. Through four ML algorithms, two hub genes (ISLR/SFRP4) were identified as candidate HF biomarkers, and a nomogram was developed. ROC analysis showed that the AUC of the nomogram was higher than 0.90 in both the HF combined dataset and two external cohorts. In addition, an immune cell infiltration analysis revealed the immune dysregulation in HF, with ISLR/SFRP4 displaying notable associations with the infiltration of B cells, CD8 T cells, and fibroblasts. More importantly, animal experiments showed that the expression levels of ISLR (r = -0.653, p < 0.001) and SFRP4 (r = -0.476, p = 0.008) were significantly negatively correlated with LVEF. CONCLUSIONS The MR analysis indicated that MDD is a risk factor for HF at the genetic level. Bioinformatics analysis and the ML results suggest that ISLR and SFRP4 have the potential to serve as diagnostic biomarkers for HF. Animal experiments showed a negative correlation between the serum levels of ISLR/SFRP4 and LVEF, emphasizing the need for additional clinical studies to elucidate their diagnostic value.
Collapse
Affiliation(s)
- Chuanjing Zhang
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Yongfei Song
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Lichao Cen
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Chen Huang
- Department of Genetics, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Jianqing Zhou
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Jiangfang Lian
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| |
Collapse
|
8
|
Zhao R, Yan Y, Dong Y, Wang X, Li X, Qiao R, Zhang H, Cui N, Han Y, Wang C, Han J, Ma Q, Liu D, Yang J, Gu G, Wang C. FGF13 deficiency ameliorates calcium signaling abnormality in heart failure by regulating microtubule stability. Biochem Pharmacol 2024; 225:116329. [PMID: 38821375 DOI: 10.1016/j.bcp.2024.116329] [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: 02/09/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Calcium signaling abnormality in cardiomyocytes, as a key mechanism, is closely associated with developing heart failure. Fibroblast growth factor 13 (FGF13) demonstrates important regulatory roles in the heart, but its association with cardiac calcium signaling in heart failure remains unknown. This study aimed to investigate the role and mechanism of FGF13 on calcium mishandling in heart failure. Mice underwent transaortic constriction to establish a heart failure model, which showed decreased ejection fraction, fractional shortening, and contractility. FGF13 deficiency alleviated cardiac dysfunction. Heart failure reduces calcium transients in cardiomyocytes, which were alleviated by FGF13 deficiency. Meanwhile, FGF13 deficiency restored decreased Cav1.2 and Serca2α expression and activity in heart failure. Furthermore, FGF13 interacted with microtubules in the heart, and FGF13 deficiency inhibited the increase of microtubule stability during heart failure. Finally, in isoproterenol-stimulated FGF13 knockdown neonatal rat ventricular myocytes (NRVMs), wildtype FGF13 overexpression, but not FGF13 mutant, which lost the binding site of microtubules, promoted calcium transient abnormality aggravation and Cav1.2 downregulation compared with FGF13 knockdown group. Generally, FGF13 deficiency improves abnormal calcium signaling by inhibiting the increased microtubule stability in heart failure, indicating the important role of FGF13 in cardiac calcium homeostasis and providing new avenues for heart failure prevention and treatment.
Collapse
Affiliation(s)
- Ran Zhao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China
| | - Yingke Yan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China
| | - Yiming Dong
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiangchong Wang
- Department of Pharmacology, Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Xuyan Li
- College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Ruoyang Qiao
- College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, China
| | - Nanqi Cui
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yanxue Han
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China
| | - Cong Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China
| | - Jiabing Han
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China
| | - Qianli Ma
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Demin Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Jing Yang
- Department of Pathology and Pathophysiology, Hangzhou Normal University, Hangzhou 311121, China.
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| | - Chuan Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
9
|
An X, Xu W, Zhao X, Chen H, Yang J, Wu Y, Wang D, Cheng W, Li H, Zeng L, Ma J, Wang Q, Wang X, Hou Y, Ai J. Bazi Bushen capsule attenuates cardiac systolic injury via SIRT3/SOD2 pathway in high-fat diet-fed ovariectomized mice. Heliyon 2024; 10:e32159. [PMID: 38912487 PMCID: PMC11190601 DOI: 10.1016/j.heliyon.2024.e32159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024] Open
Abstract
Background Bazi Bushen capsule (BZBS) is a Chinese herbal compound that is clinically used to treat fatigue and forgetfulness. However, it is still unclear whether and how BZBS affects heart function decline in menopausal women. This study aimed to examine the effect of BZBS on cardiac function in a high-fat diet-fed ovariectomy (HFD-fed OVX) mouse model and elucidate the underlying mechanism of this effect. Methods The experimental animals were divided into five groups: sham group, HFD-fed OVX group, and BZBS (0.7, 1.4, 2.8 g/kg) intervention groups. Senescence β-galactosidase staining and echocardiography were used to evaluate cardiac function. SwissTargetPrediction, KEGG and GO enrichment analyses were used to screen the underlying mechanism of BZBS. The morphological and functional changes in cardiac mitochondria and the underlying molecular mechanism were assessed by transmission electron microscopy, western blotting and biochemical assays. STRING database was used to analysis protein-protein interaction (PPI) network. Molecular docking studies were employed to predict the interactions of specific BZBS compounds with their protein targets. Results BZBS treatment ameliorated cardiac senescence and cardiac systole injury in HFD-fed OVX mice. GO and KEGG analyses revealed that the 530 targets of the 14 main components of BZBS were enriched mainly in the oxidative stress-associated pathway, which was confirmed by the finding that BZBS treatment prevented abnormal morphological changes and oxidative stress damage to cardiac mitochondria in HFD-fed OVX mice. Furthermore, the STRING database showed that the targets of BZBS were broadly related to the Sirtuins family. And BZBS upregulated the SIRT3 and elevated the activity of SOD2 in the hearts of HFD-fed OVX mice, which was also verified in vitro. Additionally, we revealed that imperatorin and osthole from the BZBS upregulated the expression of SIRT3 by directly docking with the transcription factors HDAC1, HDAC2, and BRD4, which regulate the expression of SIRT3. Conclusion This research shows that the antioxidative effect and cardioprotective role of BZBS on HFD-fed OVX mice involves an increase in the activity of the SIRT3/SOD2 pathway, and the imperatorin and osthole of BZBS may play central roles in this process.
Collapse
Affiliation(s)
- Xiaobin An
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Wentao Xu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Xinyue Zhao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Haihui Chen
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Jinan Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Yan Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Dongyang Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Wei Cheng
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Hongrong Li
- New Drug Evaluation Center, Shijiazhuang Yiling Pharmaceutical Co., Ltd., Shijiazhuang, Hebei Province, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei Province, 050035, China
| | - Lu Zeng
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Jing Ma
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Qin Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Xuqiao Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Yunlong Hou
- New Drug Evaluation Center, Shijiazhuang Yiling Pharmaceutical Co., Ltd., Shijiazhuang, Hebei Province, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei Province, 050035, China
| | - Jing Ai
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), National Key Laboratory of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| |
Collapse
|
10
|
Li C, Zhang Y, Shen J, Bao H, Zhao Y, Li D, Li S, Liu Y, Yang J, Zhou Z, Gao K, Zhao L, Pei Y, Lu Y, Pan Z, Cai B. Cfp1 Controls Cardiomyocyte Maturation by Modifying Histone H3K4me3 of Structural, Metabolic, and Contractile Related Genes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305992. [PMID: 38196272 PMCID: PMC10953565 DOI: 10.1002/advs.202305992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/26/2023] [Indexed: 01/11/2024]
Abstract
Cardiomyocyte maturation is the final stage of heart development, and abnormal cardiomyocyte maturation will lead to serious heart diseases. CXXC zinc finger protein 1 (Cfp1), a key epigenetic factor in multi-lineage cell development, remains underexplored in its influence on cardiomyocyte maturation. This study investigates the role and mechanisms of Cfp1 in this context. Cardiomyocyte-specific Cfp1 knockout (Cfp1-cKO) mice died within 4 weeks of birth. Cardiomyocytes derived from Cfp1-cKO mice showed an inhibited maturation phenotype, characterized by structural, metabolic, contractile, and cell cycle abnormalities. In contrast, cardiomyocyte-specific Cfp1 transgenic (Cfp1-TG) mice and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) overexpressing Cfp1 displayed a more mature phenotype. Mechanistically, deficiency of Cfp1 led to a reduction in trimethylation on lysine 4 of histone H3 (H3K4me3) modification, accompanied by the formation of ectopic H3K4me3. Furthermore, Cfp1 deletion decreased the level of H3K4me3 modification in adult genes and increased the level of H3K4me3 modification in fetal genes. Collectively, Cfp1 modulates the expression of genes crucial to cardiomyocyte maturation by regulating histone H3K4me3 modification, thereby intricately influencing the maturation process. This study implicates Cfp1 as an important molecule regulating cardiomyocyte maturation, with its dysfunction strongly linked to cardiac disease.
Collapse
Affiliation(s)
- Changzhu Li
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Yang Zhang
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Jingling Shen
- Institute of Life SciencesCollege of Life and Environmental SciencesWenzhou UniversityWenzhou325035P. R. China
| | - Hairong Bao
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Yue Zhao
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Desheng Li
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Sijia Li
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Yining Liu
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Jiming Yang
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Zhiwen Zhou
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Kangyi Gao
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Lexin Zhao
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Yao Pei
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Yanjie Lu
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| | - Zhenwei Pan
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
- Research Unit of Noninfectious Chronic Diseases in Frigid ZoneChinese Academy of Medical Sciences2019 Research Unit 070HarbinHeilongjiang150086P. R. China
- Key Laboratory of Cell TransplantationThe First Affiliated HospitalHarbin Medical UniversityP. R. China
| | - Benzhi Cai
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular DiseaseKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinHeilongjiang150086P. R. China
| |
Collapse
|
11
|
Markousis-Mavrogenis G, Baumhove L, Al-Mubarak AA, Aboumsallem JP, Bomer N, Voors AA, van der Meer P. Immunomodulation and immunopharmacology in heart failure. Nat Rev Cardiol 2024; 21:119-149. [PMID: 37709934 DOI: 10.1038/s41569-023-00919-6] [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] [Accepted: 08/03/2023] [Indexed: 09/16/2023]
Abstract
The immune system is intimately involved in the pathophysiology of heart failure. However, it is currently underused as a therapeutic target in the clinical setting. Moreover, the development of novel immunomodulatory therapies and their investigation for the treatment of patients with heart failure are hampered by the fact that currently used, evidence-based treatments for heart failure exert multiple immunomodulatory effects. In this Review, we discuss current knowledge on how evidence-based treatments for heart failure affect the immune system in addition to their primary mechanism of action, both to inform practising physicians about these pleiotropic actions and to create a framework for the development and application of future immunomodulatory therapies. We also delineate which subpopulations of patients with heart failure might benefit from immunomodulatory treatments. Furthermore, we summarize completed and ongoing clinical trials that assess immunomodulatory treatments in heart failure and present several therapeutic targets that could be investigated in the future. Lastly, we provide future directions to leverage the immunomodulatory potential of existing treatments and to foster the investigation of novel immunomodulatory therapeutics.
Collapse
Affiliation(s)
- George Markousis-Mavrogenis
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lukas Baumhove
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ali A Al-Mubarak
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
12
|
Baumhove L, Bomer N, Tromp J, van Essen BJ, Dickstein K, Cleland JG, Lang CC, Ng LL, Samani NJ, Anker SD, Metra M, van Veldhuisen DJ, van der Meer P, Voors AA. Clinical characteristics and prognosis of patients with heart failure and high concentrations of interleukin-17D. Int J Cardiol 2024; 396:131384. [PMID: 37739044 DOI: 10.1016/j.ijcard.2023.131384] [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: 07/12/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
AIMS Heart failure (HF) is associated with cytokine activation and inflammation. Experimental evidence suggests that plasma interleukin-17 (IL-17) is associated with myocardial fibrosis and cardiac dysfunction in HF. IL-17D, a subtype of IL-17 originates from particular tissues such as the heart. However, there is very limited data on the IL-17 cytokine family in patients with HF. Therefore, we investigated the association between circulating IL-17D levels, clinical characteristics and outcome in a large cohort of patients with heart failure. METHODS AND RESULTS Plasma IL-17D was measured in 2032 patients with HF from 11 European countries using a proximity extension assay. The primary outcome was a composite of HF hospitalization or all-cause mortality. Patients with higher plasma IL-17D concentrations were more likely to have atrial fibrillation (AF), renal dysfunction and heart failure with preserved ejection fraction (HFpEF) and had higher plasma N-terminal pro-brain natriuretic peptide (NT-proBNP) concentrations (all p < 0.001). IL-17D was not associated with interleukin-6 (IL-6) or C-reactive protein (CRP) concentrations. After adjustment for confounders in a multivariable Cox regression analysis, patients in the highest quartile of plasma IL-17D had a significantly increased risk of the composite outcome of HF hospitalization or all-cause mortality compared to patients in the lowest quartile [Hazard ratio (HR) 1.28, 95% confidence interval (CI) 1.05-1.57]. CONCLUSION In patients with HF, elevated plasma IL-17D concentrations are associated with higher plasma NT-proBNP concentrations and a higher prevalence of AF and renal dysfunction. High IL-17D concentrations are independently associated with worse outcome.
Collapse
Affiliation(s)
- Lukas Baumhove
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jasper Tromp
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands; Duke-NUS Medical School, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Bart J van Essen
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | | | - John G Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow. UK
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Stefan D Anker
- Department of Cardiology (CVK) and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Metra
- Institute of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
13
|
Deng X, Yang Z, Li T, Wang Y, Yang Q, An R, Xu J. Identification of 4 autophagy-related genes in heart failure by bioinformatics analysis and machine learning. Front Cardiovasc Med 2024; 11:1247079. [PMID: 38347953 PMCID: PMC10859477 DOI: 10.3389/fcvm.2024.1247079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/18/2024] [Indexed: 02/15/2024] Open
Abstract
Introduction Autophagy refers to the process of breaking down and recycling damaged or unnecessary components within a cell to maintain cellular homeostasis. Heart failure (HF) is a severe medical condition that poses a serious threat to the patient's life. Autophagy is known to play a pivotal role in the pathogenesis of HF. However, our understanding of the specific mechanisms involved remains incomplete. Here, we identify autophagy-related genes (ARGs) associated with HF, which we believe will contribute to further comprehending the pathogenesis of HF. Methods By searching the GEO (Gene Expression Omnibus) database, we found the GSE57338 dataset, which was related to HF. ARGs were obtained from the HADb and HAMdb databases. Annotation of GO and enrichment analysis of KEGG pathway were carried out on the differentially expressed ARGs (AR-DEGs). We employed machine learning algorithms to conduct a thorough screening of significant genes and validated these genes by analyzing external dataset GSE76701 and conducting mouse models experimentation. At last, immune infiltration analysis was conducted, target drugs were screened and a TF regulatory network was constructed. Results Through processing the dataset with R language, we obtained a total of 442 DEGs. Additionally, we retrieved 803 ARGs from the database. The intersection of these two sets resulted in 15 AR-DEGs. Upon performing functional enrichment analysis, it was discovered that these genes exhibited significant enrichment in domains related to "regulation of cell growth", "icosatetraenoic acid binding", and "IL-17 signaling pathway". After screening and verification, we ultimately identified 4 key genes. Finally, an analysis of immune infiltration illustrated significant discrepancies in 16 distinct types of immune cells between the HF and control group and up to 194 potential drugs and 16 TFs were identified based on the key genes. Discussion In this study, TPCN1, MAP2K1, S100A9, and CD38 were considered as key autophagy-related genes in HF. With these relevant data, further exploration of the molecular mechanisms of autophagy in HF can be carried out.
Collapse
Affiliation(s)
- Xiwei Deng
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Oncology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Ziqi Yang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Tongzheng Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yang Wang
- Department of Oncology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Qinchuan Yang
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Rui An
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jian Xu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Interventional Surgery Center, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
14
|
Hu Y, Chen X, Mei X, Luo Z, Wu H, Zhang H, Zeng Q, Ren H, Xu D. Identification of diagnostic immune-related gene biomarkers for predicting heart failure after acute myocardial infarction. Open Med (Wars) 2023; 18:20230878. [PMID: 38152337 PMCID: PMC10751901 DOI: 10.1515/med-2023-0878] [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: 09/05/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Post-myocardial infarction heart failure (HF) is a major public health concern. Previous studies have reported the critical role of immune response in HF pathogenesis. However, limited studies have reported predictive immune-associated biomarkers for HF. So we attempted to identify potential immune-related indicators for HF early diagnosis and therapy guidance. This study identified two potential immune-related hub genes (IRHGs), namely CXCR5 and FOS, using bioinformatic approaches. The expression levels of CXCR5 and FOS and their ability to predict long-term HF were analyzed. Functional enrichment analysis revealed that the hub genes were enriched in immune system processes, including the interleukin-17 and nuclear factor-kappa B signaling pathways, which are involved in the pathogenesis of HF. Quantitative real-time polymerase chain reaction revealed that the Fos mRNA levels, but not the Cxcr5 mRNA levels, were downregulated in the mice of the HF group. This study successfully identified two IRHGs that were significantly and differentially expressed in the HF group and could predict long-term HF, providing novel insights for future studies on HF and developing novel therapeutic targets for HF.
Collapse
Affiliation(s)
- Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaoyu Chen
- Department of Nephrology, Rheumatism and Immunology, Chongqing Jiulongpo People’s Hospital, Chongqing, 400050, China
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hongguang Wu
- Department of Arrhythmic, Cardiovascular Medical Center, Shenzhen Hospital of University of Hong Kong, Shenzhen, 518040, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qingchun Zeng
- Department of Cardiology, Nanfang Hospital, Southern Medical University,
Guangzhou, 510515, Guangdong, China
| | - Hao Ren
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University,
Guangzhou, 510515, Guangdong, China
| |
Collapse
|
15
|
Luo X, Wang R, Zhang X, Wen X, Deng S, Xie W. Identification CCL2,CXCR2,S100A9 of the immune-related gene markers and immune infiltration characteristics of inflammatory bowel disease and heart failure via bioinformatics analysis and machine learning. Front Cardiovasc Med 2023; 10:1268675. [PMID: 38034382 PMCID: PMC10687362 DOI: 10.3389/fcvm.2023.1268675] [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: 08/04/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Background Recently, heart failure (HF) and inflammatory bowel disease (IBD) have been considered to be related diseases with increasing incidence rates; both diseases are related to immunity. This study aims to analyze and identify immune-related gene (IRG) markers of HF and IBD through bioinformatics and machine learning (ML) methods and to explore their immune infiltration characteristics. Methods This study used gene expressiondata (GSE120895, GSE21610, GSE4183) from the Gene Expression Omnibus (GEO) database to screen differentially expressed genes (DEGs) and compare them with IRGs from the ImmPort database to obtain differentially expressed immune-related genes (DIRGs). Functional enrichment analysis of IRGs was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Subsequently, three machine models and protein-protein interactions (PPIs) were established to identify diagnostic biomarkers. The receiver operating characteristic (ROC) curves were applied to evaluate the diagnostic value of the candidate biomarkersin the validation set (GSE1145, GSE36807) and obtain their correlations with immune cells through the Spearman algorithm. Finally, the CIBERSORT algorithm was used to evaluate the immune cell infiltration of the two diseases. Results Thirty-four DIRGs were screened and GO and KEGG analysis results showed that these genes are mainly related to inflammatory and immune responses. CCL2, CXCR2 and S100A9 were identified as biomarkers.The immune correlation results indicated in both diseases that CCL2 is positively correlated with mast cell activation, CXCR2 is positively correlated with neutrophils and S100A9 is positively correlated with neutrophils and mast cell activation. Analysis of immune characteristics showed that macrophages M2, macrophages M0 and neutrophils were present in both diseases. Conclusions CCL2, CXCR2 and S100A9 are promising biomarkers that will become potential immunogenetic biomarkers for diagnosing comorbidities of HF and IBD. macrophages M2, macrophages M0, neutrophil-mediated inflammation and immune regulation play important roles in the development of HF and IBD and may become diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Xu Luo
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Wang
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zhang
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Wen
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siwei Deng
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Xie
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Cardiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Li X, Sun T, Liu J, Wei S, Yang Y, Liu J, Zhang B, Li W. Phloretin alleviates doxorubicin-induced cardiotoxicity through regulating Hif3a transcription via targeting transcription factor Fos. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155046. [PMID: 37659297 DOI: 10.1016/j.phymed.2023.155046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Doxorubicin (Dox), a chemotherapeutic agent known for its efficacy, has been associated with the development of severe cardiotoxicity, commonly referred to as doxorubicin-induced cardiotoxicity (DIC). The role and mechanism of action of phloretin (Phl) in cardiovascular diseases are well-established; however, its specific function and underlying mechanism in the context of DIC have yet to be fully elucidated. OBJECTIVE This research aimed to uncover the protective effect of Phl against DIC in vivo and in vitro, while also providing a comprehensive understanding of the underlying mechanisms involved. METHODS DIC cell and murine models were established. The action targets and mechanism of Phl against DIC were comprehensively examined by systematic network pharmacology, molecular docking, transcriptomics technologies, transcription factor (TF) prediction, and experimental validation. RESULTS Phl relieved Dox-induced cell apoptosis in vitro and in vivo. Through network pharmacology analysis, a total of 554 co-targeted genes of Phl and Dox were identified. Enrichment analysis revealed several key pathways including the PI3K-Akt signaling pathway, Apoptosis, and the IL-17 signaling pathway. Protein-protein interaction (PPI) analysis identified 24 core co-targeted genes, such as Fos, Jun, Hif1a, which were predicted to bind well to Phl based on molecular docking. Transcriptomics analysis was performed to identify the top 20 differentially expressed genes (DEGs), and 202 transcription factors (TFs) were predicted for these DEGs. Among these TFs, 10 TFs (Fos, Jun, Hif1a, etc.) are also the co-targeted genes, and 3 TFs (Fos, Jun, Hif1a) are also the core co-targeted genes. Further experiments validated the finding that Phl reduced the elevated levels of Hif3a (one of the top 20 DEGs) and Fos (one of Hif3a's predicted TFs) induced by Dox. Moreover, the interaction between Fos protein and the Hif3a promoter was confirmed through luciferase reporter assays. CONCLUSION Phl actively targeted and down-regulated the Fos protein to inhibit its binding to the promoter region of Hif3a, thereby providing protection against DIC.
Collapse
Affiliation(s)
- Xiangyun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China; School of Pharmacy, Central South University, Changsha, Hunan 410078, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiaqin Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Yuanying Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China; School of Pharmacy, Central South University, Changsha, Hunan 410078, China.
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
17
|
Dridi H, Santulli G, Bahlouli L, Miotto MC, Weninger G, Marks AR. Mitochondrial Calcium Overload Plays a Causal Role in Oxidative Stress in the Failing Heart. Biomolecules 2023; 13:1409. [PMID: 37759809 PMCID: PMC10527470 DOI: 10.3390/biom13091409] [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: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Heart failure is a serious global health challenge, affecting more than 6.2 million people in the United States and is projected to reach over 8 million by 2030. Independent of etiology, failing hearts share common features, including defective calcium (Ca2+) handling, mitochondrial Ca2+ overload, and oxidative stress. In cardiomyocytes, Ca2+ not only regulates excitation-contraction coupling, but also mitochondrial metabolism and oxidative stress signaling, thereby controlling the function and actual destiny of the cell. Understanding the mechanisms of mitochondrial Ca2+ uptake and the molecular pathways involved in the regulation of increased mitochondrial Ca2+ influx is an ongoing challenge in order to identify novel therapeutic targets to alleviate the burden of heart failure. In this review, we discuss the mechanisms underlying altered mitochondrial Ca2+ handling in heart failure and the potential therapeutic strategies.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Laith Bahlouli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Marco C. Miotto
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| |
Collapse
|
18
|
Luo Z, Lu G, Yang Q, Ding J, Wang T, Hu P. Identification of Shared Immune Cells and Immune-Related Co-Disease Genes in Chronic Heart Failure and Systemic Lupus Erythematosus Based on Transcriptome Sequencing. J Inflamm Res 2023; 16:2689-2705. [PMID: 37408607 PMCID: PMC10319289 DOI: 10.2147/jir.s418598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023] Open
Abstract
Purpose The purpose was to identify shared immune cells and co-disease genes in chronic heart failure (HF) and systemic lupus erythematosus (SLE), as well as explore the potential mechanisms of action between HF and SLE. Methods A collection of peripheral blood mononuclear cells (PBMCs) from ten patients with HF and SLE and ten normal controls (NC) was used for transcriptome sequencing. Differentially expressed genes (DEGs) analysis, enrichment analysis, immune infiltration analysis, weighted gene co-expression network analysis (WGCNA), protein-protein interaction (PPI) analysis, and machine learning were applied for the screening of shared immune cells and co-disease genes in HF and SLE. Gene expression analysis and correlation analysis were used to explore the potential mechanisms of co-disease genes and immune cells in HF and SLE. Results In this study, it was found that two immune cells, T cells CD4 naïve and Monocytes, displayed similar expression patterns in HF and SLE at the same time. By taking intersection of the above immune cell-associated genes with the DEGs common to both HF and SLE, four immune-associated co-disease genes, CCR7, RNASE2, RNASE3 and CXCL10, were finally identified. CCR7, as one of the four key genes, was significantly down-regulated in HF and SLE, while the rest three key genes were all significantly up-regulated in both diseases. Conclusion T cells CD4 naïve and Monocytes were first revealed as possible shared immune cells of HF and SLE, and CCR7, RNASE2, RNASE3 and CXCL10 were identified as possible key genes common to HF and SLE as well as potential biomarkers or therapeutic targets for HF and SLE.
Collapse
Affiliation(s)
- Ziyue Luo
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Guifang Lu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310005, People's Republic of China
| | - Qiang Yang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Juncan Ding
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Tianyu Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310005, People's Republic of China
| |
Collapse
|
19
|
Liu Y, Zhang D, Yin D. Pathophysiological Effects of Various Interleukins on Primary Cell Types in Common Heart Disease. Int J Mol Sci 2023; 24:ijms24076497. [PMID: 37047468 PMCID: PMC10095356 DOI: 10.3390/ijms24076497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Myocardial infarction (MI), heart failure, cardiomyopathy, myocarditis, and myocardial ischemia-reperfusion injury (I/R) are the most common heart diseases, yet there is currently no effective therapy due to their complex pathogenesis. Cardiomyocytes (CMs), fibroblasts (FBs), endothelial cells (ECs), and immune cells are the primary cell types involved in heart disorders, and, thus, targeting a specific cell type for the treatment of heart disease may be more effective. The same interleukin may have various effects on different kinds of cell types in heart disease, yet the exact role of interleukins and their pathophysiological pathways on primary cell types remain largely unexplored. This review will focus on the pathophysiological effects of various interleukins including the IL-1 family (IL-1, IL-18, IL-33, IL-37), IL-2, IL-4, the IL-6 family (IL-6 and IL-11), IL-8, IL-10, IL-17 on primary cell types in common heart disease, which may contribute to the more precise and effective treatment of heart disease.
Collapse
Affiliation(s)
- Yong Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
- Correspondence: (D.Z.); (D.Y.)
| | - Dan Yin
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan 430062, China
- Correspondence: (D.Z.); (D.Y.)
| |
Collapse
|
20
|
Combined Toxicity of the Most Common Indoor Aspergilli. Pathogens 2023; 12:pathogens12030459. [PMID: 36986381 PMCID: PMC10058518 DOI: 10.3390/pathogens12030459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
The most common Aspergilli isolated from indoor air samples from occupied buildings and a grain mill were extracted and analyzed for their combined (Flavi + Nigri, Versicolores + Nigri) cytotoxic, genotoxic and pro-inflammatory properties on human adenocarcinoma cells (A549) and monocytic leukemia cells induced in macrophages (THP-1 macrophages). Metabolite mixtures from the Aspergilli series Nigri increase the cytotoxic and genotoxic potency of Flavi extracts in A549 cells suggesting additive and/or synergistic effects, while antagonizing the cytotoxic potency of Versicolores extracts in THP-1 macrophages and genotoxicity in A549 cells. All tested combinations significantly decreased IL-5 and IL-17, while IL-1β, TNF-α and IL-6 relative concentrations were increased. Exploring the toxicity of extracted Aspergilli deepens the understanding of intersections and interspecies differences in events of chronic exposure to their inhalable mycoparticles.
Collapse
|
21
|
Gergely TG, Kucsera D, Tóth VE, Kovács T, Sayour NV, Drobni ZD, Ruppert M, Petrovich B, Ágg B, Onódi Z, Fekete N, Pállinger É, Buzás EI, Yousif LI, Meijers WC, Radovits T, Merkely B, Ferdinandy P, Varga ZV. Characterization of immune checkpoint inhibitor-induced cardiotoxicity reveals interleukin-17A as a driver of cardiac dysfunction after anti-PD-1 treatment. Br J Pharmacol 2023; 180:740-761. [PMID: 36356191 DOI: 10.1111/bph.15984] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/06/2022] [Accepted: 10/29/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Immune checkpoint inhibitors (ICI), such as anti-PD-1 monoclonal antibodies, have revolutionized cancer therapy by enhancing the cytotoxic effects of T-cells against tumours. However, enhanced T-cell activity also may cause myocarditis and cardiotoxicity. Our understanding of the mechanisms of ICI-induced cardiotoxicity is limited. Here, we aimed to investigate the effect of PD-1 inhibition on cardiac function and explore the molecular mechanisms of ICI-induced cardiotoxicity. EXPERIMENTAL APPROACH C57BL6/J and BALB/c mice were treated with isotype control or anti-PD-1 antibody. Echocardiography was used to assess cardiac function. Cardiac transcriptomic changes were investigated by bulk RNA sequencing. Inflammatory changes were assessed by qRT-PCR and immunohistochemistry in heart, thymus, and spleen of the animals. In follow-up experiments, anti-CD4 and anti-IL-17A antibodies were used along with PD-1 blockade in C57BL/6J mice. KEY RESULTS Anti-PD-1 treatment led to cardiac dysfunction and left ventricular dilation in C57BL/6J mice, with increased nitrosative stress. Only mild inflammation was observed in the heart. However, PD-1 inhibition resulted in enhanced thymic inflammatory signalling, where Il17a increased most prominently. In BALB/c mice, cardiac dysfunction was not evident, and thymic inflammatory activation was more balanced. Inhibition of IL-17A prevented anti-PD-1-induced cardiac dysfunction in C57BL6/J mice. Comparing myocardial transcriptomic changes in C57BL/6J and BALB/c mice, differentially regulated genes (Dmd, Ass1, Chrm2, Nfkbia, Stat3, Gsk3b, Cxcl9, Fxyd2, and Ldb3) were revealed, related to cardiac structure, signalling, and inflammation. CONCLUSIONS PD-1 blockade induces cardiac dysfunction in mice with increased IL-17 signalling in the thymus. Pharmacological inhibition of IL-17A treatment prevents ICI-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Viktória E Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Tamás Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Zsófia D Drobni
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Balázs Petrovich
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary.,MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Nóra Fekete
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Laura I Yousif
- Department of Cardiology, Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Division of Experimental Cardiology, Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wouter C Meijers
- Department of Cardiology, Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Division of Experimental Cardiology, Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary.,MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Wang A, Li Z, Sun Z, Wang Y, Fu S, Zhang D, Ma X. Heart failure with preserved ejection fraction and non-alcoholic fatty liver disease: new insights from bioinformatics. ESC Heart Fail 2023; 10:416-431. [PMID: 36266995 PMCID: PMC9871724 DOI: 10.1002/ehf2.14211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/17/2022] [Accepted: 10/02/2022] [Indexed: 01/29/2023] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) and non-alcoholic fatty liver disease (NAFLD) are related conditions with an increasing incidence. The mechanism of their relationship remains undefined. Here, we aimed to explore the potential mechanisms, diagnostic markers, and therapeutic options for HFpEF and NAFLD. METHODS AND RESULTS HFpEF and NAFLD datasets were downloaded from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (DEGs) were screened for functional annotation. A protein-protein interaction network was constructed based on the STRING database, and hub genes were analysed using GeneMANIA annotation. ImmuCellAI (Immune Cell Abundance Identifier) was employed for analysis of immune infiltration. We also used validation datasets to validate the expression levels of hub genes and the correlation of immune cells. To screen for diagnostic biomarkers, we employed the least absolute shrinkage and selection operator and support vector machine-recursive feature elimination. Drug signature database was used to predict potential therapeutic drugs. Our analyses identified a total of 33 DEGs. Inflammation and immune infiltration played important roles in the development of both diseases. The data showed a close relationship between chemokine signalling pathway, cytokine-cytokine receptor interaction, calcium signalling pathway, neuroactive ligand-receptor interaction, osteoclast differentiation, and cyclic guanosine monophosphate-protein kinase G signalling pathway. We demonstrated that PRF1 (perforin 1) and IL2RB (interleukin-2 receptor subunit beta) proteins were perturbed by the diseases and may be the hub genes. The analysis showed that miR-375 may be a potential diagnostic marker for both diseases. Our drug prediction analysis showed that bosentan, eldecalcitol, ramipril, and probucol could be potential therapeutic options for the diseases. CONCLUSIONS Our findings revealed common pathogenesis, diagnostic markers, and therapeutic agents for HFpEF and NAFLD. There is need for further experimental studies to validate our findings.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
- Graduate SchoolChina Academy of Chinese Medical SciencesBeijingChina
| | - Zhendong Li
- Qingdao West Coast New Area People's HospitalQingdaoChina
| | - Zhuo Sun
- Qingdao West Coast New Area People's HospitalQingdaoChina
| | - Yifei Wang
- Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
- Beijing University of Chinese MedicineBeijingChina
| | - Shuangqing Fu
- Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
- Beijing University of Chinese MedicineBeijingChina
| | - Dawu Zhang
- Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
- National Clinical Research Center for Chinese Medicine CardiologyBeijingChina
| | - Xiaochang Ma
- Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
- National Clinical Research Center for Chinese Medicine CardiologyBeijingChina
| |
Collapse
|
23
|
Wang Y, Li Y, Zhang W, Yuan Z, Lv S, Zhang J. NLRP3 Inflammasome: a Novel Insight into Heart Failure. J Cardiovasc Transl Res 2023; 16:166-176. [PMID: 35697978 DOI: 10.1007/s12265-022-10286-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
Among numerous cardiovascular diseases, heart failure is a final and fatal stage, and its morbidity, mortality, and rehospitalization rate remain high, which reduces the exercise tolerance of patients and brings great medical burden and economic pressure to the society. Inflammation takes on a major influence in the occurrence, development, and prognosis of heart failure (HF). The NLRP3 inflammasome is a key node in a chronic inflammatory response, which can accelerate the production of pro-inflammatory cytokines IL-1β and IL-18, leading to the inflammatory response. Therefore, whether it is possible to suppress the downstream factors of NLRP3 inflammasome and its signaling path is expected to provide a new intervention mediator for the therapy of heart failure. This article synopsizes the research progress of NLRP3 inflammasome in heart failure, to provide a reference for clinical treatment. CLINICAL RELEVANCE: This study explored the downstream factors of NLRP3 inflammasome and its signal pathway. Targeted drug therapy for NLRP3 inflammasome is expected to provide a new intervention target for the treatment of heart failure.
Collapse
Affiliation(s)
- Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wanqin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Zhuo Yuan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Shichao Lv
- 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
|
24
|
Fir(e)ing the Rhythm. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
25
|
Mohamad HE, Asker ME, Shaheen MA, Baraka NM, Fantoukh OI, Alqahtani A, Salama AE, Mahmoud YK. Secukinumab and Black Garlic Downregulate OPG/RANK/RANKL Axis and Devitalize Myocardial Interstitial Fibrosis Induced by Sunitinib in Experimental Rats. Life (Basel) 2023; 13:life13020308. [PMID: 36836664 PMCID: PMC9962443 DOI: 10.3390/life13020308] [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: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Sunitinib has been associated with several cardiotoxic effects such as cardiac fibrosis. The present study was designed to explore the role of interleukin (IL)-17 in sunitinib-induced myocardial fibrosis (MF) in rats and whether its neutralization and/or administration of black garlic (BG), a form of fermented raw garlic (Allium sativum L.), could extenuate this adverse effect. Male Wistar albino rats received sunitinib (25 mg/kg three times a week, orally) and were co-treated with secukinumab (3 mg/kg, subcutaneously, three times total) and/or BG (300 mg/kg/day, orally) for four weeks. Administration of sunitinib induced significant increase in cardiac index, cardiac inflammatory markers, and cardiac dysfunction that were ameliorated by both secukinumab and BG, and to a preferable extent, with the combined treatment. Histological examination revealed disruption in the myocardial architecture and interstitial fibrosis in cardiac sections of the sunitinib group, which were reversed by both secukinumab and BG treatments. Both drugs and their co-administration restored normal cardiac functions, downregulated cardiac inflammatory cytokines, mainly IL-17 and NF-κB, along with increasing the MMP1/TIMP1 ratio. Additionally, they attenuated sunitinib-induced upregulation of the OPG/RANK/RANKL axis. These findings highlight another new mechanism through which sunitinib can induce interstitial MF. The current results propose that neutralizing IL-17 by secukinumab and/or supplementation with BG can be a promising therapeutic approach for ameliorating sunitinib-induced MF.
Collapse
Affiliation(s)
- Hoda E. Mohamad
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Correspondence: ; Tel.: +20-10-2799-4483
| | - Mervat E. Asker
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Shaheen
- Department of Histology & Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Nourhan M. Baraka
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Omer I. Fantoukh
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdulaziz Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alaa E. Salama
- Department of Cardiology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Yasmin K. Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
26
|
Zhang Y, Wang Y, Liu Y, Gong T, Hou M. The Anti-inflammatory Effect of Chitosan Oligosaccharide on Heart Failure in Mice. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8746530. [PMID: 36246979 PMCID: PMC9556196 DOI: 10.1155/2022/8746530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/19/2022]
Abstract
Heart failure is currently one of the leading causes of death worldwide, and the inflammatory factors play an important role in its development. Chitosan oligosaccharide (COS), a low-molecular-weight form of chitosan, has many specific biological activities. In this study, COS effects on heart failure were studied for the first time by performing transverse arch constriction (TAC) surgery in mice, as an animal model of heart failure. Our findings revealed that COS administration (in both 40 mg/kg and 80 mg/kg doses) significantly ameliorated TCA-induced left ventricular (LV) hypertrophy as well as the increase in lung and heart weight in mice, while improving TAC-induced LV dysfunction. Both doses effectively attenuated LV cardiomyocyte hypertrophy, while decreasing heart inflammation after heart failure in mice. In conclusion, our results indicated that the supplementation of COS in normal diet might be an effective way to prevent further myocardial tissue damage in patients suffering from heart failure.
Collapse
Affiliation(s)
- Yubiao Zhang
- Department of Food Science, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning, China 110866
| | - Yu Wang
- Department of Food Science, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning, China 110866
| | - Yunen Liu
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, China 110016
| | - Tianxing Gong
- Sino-Dutch Biomedical and Information Engineering School, Northeastern University, No. 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning, China 110169
| | - Mingxiao Hou
- Department of Food Science, Shenyang Agricultural University, No. 120 Dongling Road, Shenhe District, Shenyang, Liaoning, China 110866
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, China 110016
| |
Collapse
|
27
|
Abstract
Heart regenerative medicine has been gradually evolving from a view of the heart as a nonregenerative organ with terminally differentiated cardiac muscle cells. Understanding the biology of the heart during homeostasis and in response to injuries has led to the realization that cellular communication between all cardiac cell types holds great promise for treatments. Indeed, recent studies highlight new disease-reversion concepts in addition to cardiomyocyte renewal, such as matrix- and vascular-targeted therapies, and immunotherapy with a focus on inflammation and fibrosis. In this review, we will discuss the cross-talk within the cardiac microenvironment and how specific therapies aim to target the hostile cardiac milieu under pathological conditions.
Collapse
Affiliation(s)
- Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, 60594 Frankfurt, Germany.,Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research, RheinMain, Frankfurt, Germany
| |
Collapse
|
28
|
Jiang X, Yang F, Ou D, Huang L, Li H, Lang M. MCC950 ameliorates ventricular arrhythmia vulnerability induced by heart failure. Bioengineered 2022; 13:8593-8604. [PMID: 35287557 PMCID: PMC9162026 DOI: 10.1080/21655979.2022.2053813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
MCC950, a specific NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inhibitor, has been reported to play a role in various cardiovascular diseases. However, its role in heart failure (HF)-induced ventricular arrhythmias (VAs) remains unclear. Hence, the present study aimed to clarify the role and underlying mechanisms of MCC950 in HF-induced VAs. Male C57BL/6 mice were induced with HF via transverse aortic constriction (TAC). Histological analysis, echocardiography, electrophysiological investigation, and western blot analysis were conducted to evaluate VA vulnerability induced by TAC and the potential mechanisms underlying the effects. MCC950 markedly improved cardiac function and decreased pulmonary edema induced by HF. Moreover, MCC950 also decreased VA vulnerability, as shown by the shortened QTc duration and action potential duration 90 (APD90), reduced APD alternans threshold, and decreased VA induction rate. Furthermore, MCC950 treatment significantly reversed TAC-induced cardiac hypertrophy and fibrosis. In addition, MCC950 administration increased the protein levels of ion channels (Kv4.2, KChIP2, and Cav1.2). Mechanistically, the above changes induced by MCC950 were due to the inhibition of the NLRP3 inflammasome. As a specific NLRP3 inhibitor, MCC950 significantly decreased HF-induced VA vulnerability by reversing cardiac structural remodeling and electrical remodeling, and the mechanism through which MCC950 exhibited this effect was inhibition of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xiaobo Jiang
- Department of Cardiology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, PR China.,Chengdu University of Traditional Chinese Medicine Affiliated Fifth People's Hospital, Chengdu 611130, Sichuan, China
| | - Fan Yang
- Department of Cardiology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, PR China.,Department of endocrinology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, China
| | - Dengke Ou
- Department of Cardiology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, PR China.,Chengdu University of Traditional Chinese Medicine Affiliated Fifth People's Hospital, Chengdu 611130, Sichuan, China
| | - Luyong Huang
- Department of Cardiology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, PR China.,Chengdu University of Traditional Chinese Medicine Affiliated Fifth People's Hospital, Chengdu 611130, Sichuan, China
| | - Hongfei Li
- Department of Cardiology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, PR China.,Chengdu University of Traditional Chinese Medicine Affiliated Fifth People's Hospital, Chengdu 611130, Sichuan, China
| | - Mingjian Lang
- Department of Cardiology, Fifth People's Hospital of Chengdu, Chengdu 611130, Sichuan, PR China.,Chengdu University of Traditional Chinese Medicine Affiliated Fifth People's Hospital, Chengdu 611130, Sichuan, China
| |
Collapse
|
29
|
Szabo TM, Frigy A, Nagy EE. Targeting Mediators of Inflammation in Heart Failure: A Short Synthesis of Experimental and Clinical Results. Int J Mol Sci 2021; 22:13053. [PMID: 34884857 PMCID: PMC8657742 DOI: 10.3390/ijms222313053] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 01/22/2023] Open
Abstract
Inflammation has emerged as an important contributor to heart failure (HF) development and progression. Current research data highlight the diversity of immune cells, proteins, and signaling pathways involved in the pathogenesis and perpetuation of heart failure. Chronic inflammation is a major cardiovascular risk factor. Proinflammatory signaling molecules in HF initiate vicious cycles altering mitochondrial function and perturbing calcium homeostasis, therefore affecting myocardial contractility. Specific anti-inflammatory treatment represents a novel approach to prevent and slow HF progression. This review provides an update on the putative roles of inflammatory mediators involved in heart failure (tumor necrosis factor-alpha; interleukin 1, 6, 17, 18, 33) and currently available biological and non-biological therapy options targeting the aforementioned mediators and signaling pathways. We also highlight new treatment approaches based on the latest clinical and experimental research.
Collapse
Affiliation(s)
- Timea Magdolna Szabo
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania;
| | - Attila Frigy
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania;
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
| |
Collapse
|