1
|
Jiang J, Peng W, Sun N, Zhao D, Cui W, Lai Y, Zhang C, Duan C, Zeng W. Unraveling the anoikis-cancer nexus: a bibliometric analysis of research trends and mechanisms. Future Sci OA 2025; 11:2484159. [PMID: 40160087 PMCID: PMC11959893 DOI: 10.1080/20565623.2025.2484159] [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: 09/24/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Cancer, influenced by genetics and the environment, involves anoikis, a cell death mechanism upon extracellular matrix detachment crucial for metastasis. Understanding this relationship is key for therapy. We analyze cancer and anoikis trends using bibliometrics. METHODS A search was conducted from Web of Science Core, PubMed, Scopus and non-English databases such as the CNKI (inception- 21 December 2024). Data analysis employed Microsoft Excel, VOSviewer, CiteSpace, R software, and the online platform (https://bibliometric.com/). RESULTS 2510 publications were retrieved, with a significant increase in the last decade. China led, the University of Texas system was productive, and the Oncogene Journal was popular. Breast, and colorectal cancers were frequently studied. Among them, representative tumor-related mechanisms were identified, commonalities such as (EMT, ECM, autophagy) and respective specific mechanisms were summarized. CONCLUSION This bibliometric analysis highlights rapid advances in anoikis research in cancer, emphasizing EMT and FAK pathways' translational potential, guiding targeted therapies, and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Junjie Jiang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Wei Peng
- Department of Oncology, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Nianzhe Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Weifang Cui
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Yuwei Lai
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
- Institute of Medical Sciences, Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Wei Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
2
|
Liu Y, Zhang Z, Song H, Li R, Mi K. An improved stacking model for predicting myocardial infarction risk in imbalanced data. Health Inf Sci Syst 2025; 13:16. [PMID: 39830450 PMCID: PMC11739442 DOI: 10.1007/s13755-024-00329-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Early diagnosis and treatment of myocardial infarction (MI) can significantly reduce the severity of the disease. Disease data are often imbalanced, which can lead to poor prediction outcomes when using conventional models. Therefore, developing a risk prediction model for MI with imbalanced datasets has become challenging. This paper presents a novel model called 2GDNN-FL-Stacked, which aims to address the issue of predicting the risk of MI in imbalanced data. Our group mitigates the impact of data imbalance on the model by employing random under-sampling and cost-sensitive techniques. We improve the model's identification capabilities by stacking and combining 2GDNN-FL, CatBoost, RandomForest, and LightGBM. Our model's Matthews Correlation Coefficient(MCC), F1-score, and Area Under the ROC Curve(AUC) scores increased by 0.87% - 15.70%, 0.55% - 9.81%, and 0.75% - 8.11% respectively, compared to some baseline models, which represent a significant improvement over the performance of a single model on imbalanced datasets. This paper demonstrates the effectiveness of each component through ablation experiments, showing that removing either component affects model performance and proves the efficacy of all components. The method offers new insights into predicting heart attack risks and has the potential to offer potent assistance in making clinical decisions.
Collapse
Affiliation(s)
- Yan Liu
- Wuhan University of Technology, Wuhan, 100190 Hubei China
| | - Zhiyu Zhang
- Wuhan University of Technology, Wuhan, 100190 Hubei China
| | - Huazhu Song
- Wuhan University of Technology, Wuhan, 100190 Hubei China
| | - Renjie Li
- Wuhan University of Technology, Wuhan, 100190 Hubei China
| | - Kaituo Mi
- Beijing Anngeen Technology Co., ltd, Beijing, 100176 Beijing China
| |
Collapse
|
3
|
Zhao K, Yan L, Sun X, Hu X. O-GlcNAc transferase-mediated O-GlcNAcylation of CD36 against myocardial ischemia-reperfusion injury. Tissue Cell 2025; 95:102878. [PMID: 40154105 DOI: 10.1016/j.tice.2025.102878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
CD36 affects lipid metabolism and is involved in the development of myocardial infarction (MI). O-GlcNAcylation is a promising therapeutic target for myocardial ischemia-reperfusion (I/R) injury. This study aimed to investigate the effects of CD36 on myocardial I/R injury and its O-GlcNAcylation. H9C2 cardiomyocytes were induced by hypoxia/reoxygenation (H/R), and phenotypes were evaluated using cell counting kit-8, EdU assay, flow cytometry, and TUNEL assay. The O-GlcNAcylation was evaluated by immunoprecipitation, immunoblotting, and cycloheximide chase assay. The role of CD36 in vivo was analyzed by TTC staining and TUNEL assay. The results showed that CD36 protein levels were downregulated in I/R rats and H/R-induced H9C2 cells. OGT and O-GlcNAcylation levels were decreased by H/R. Overexpression of CD36 or OGT promoted cell proliferation and inhibited apoptosis of H/R-treated cells. Moreover, OGT facilitated the O-GlcNAcylation of CD36 at S195 site and enhanced CD36 protein stability. Knockdown of CD36 abrogated the effects of cellular behaviors caused by OGT, and CD36 mutation at S195 site reversed the promotion of proliferation and lipid uptake and the inhibition of apoptosis induced by wild-type CD36. Additionally, overexpression of CD36 attenuated infarction and apoptosis in the myocardium of rats. In conclusion, OGT-mediated O-GlcNAcylation of CD36 attenuates myocardial I/R injury through promoting the proliferation and inhibiting apoptosis of cardiomyocytes. The findings suggest that targeting CD36 O-GlcNAcylation may be a promising therapy for MI.
Collapse
Affiliation(s)
- Kechao Zhao
- Department of Cardiosurgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Laisha Yan
- Department of Cardiosurgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Xinyi Sun
- Department of Cardiosurgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Xiaoyan Hu
- Department of Cardiosurgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China.
| |
Collapse
|
4
|
Zhu Y, Cao B, Huang K, Liu J. Systematic identification of SNCA as a key gene in ischemic cardiomyopathy via integrated weighted gene co-expression network analysis and experimental validation. Biochem Biophys Res Commun 2025; 772:152063. [PMID: 40414013 DOI: 10.1016/j.bbrc.2025.152063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/09/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Ischemic cardiomyopathy (ICM) is associated with high mortality and hospitalization rates, and current treatments are suboptimal. The aim of this research was to identify novel therapeutic targets for ICM. The datasets GSE57338 and GSE5406 were obtained from the Gene Expression Omnibus (GEO) database. Gene modules were constructed using weighted gene co-expression network analysis (WGCNA), and the three relevant modules associated with ICM were identified. The biological functions and signaling pathways of the genes in these modules were further explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Protein-protein interaction (PPI) networks were employed to identify hub genes within these modules. Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression was performed to establish gene models, and 16 genes associated with left ventricular ejection fraction (LVEF) in ICM were identified. The genes were validated in heart tissues from human using quantitative real-time polymerase chain reaction (qRT-PCR). Among them, growth factor receptor-bound protein 14 (GRB14), ubiquitin C-terminal hydrolase L1 (UCHL1), and synuclein alpha (SNCA) were identified as key genes significantly associated with ICM. Additionally, key genes correlated with immune and stromal cell-related types were screened using xCell. In vivo and in vitro experiments demonstrated that inhibiting SNCA could improve cardiac dysfunction, inflammatory infiltration, and fibrosis in ICM. In conclusion, this study identified 16 genes closely related to LVEF and revealed that SNCA was a key gene in ICM, providing potential biomarkers and therapeutic targets for ICM.
Collapse
Affiliation(s)
- Yaoxi Zhu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Bingxin Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
5
|
Yuan J, Deng T, Yang Q, Lv D, Zhou Z, You L, Feng Q, Meng X, Pang Q, Li H, Zhu B. Loss of LSD1 ameliorates myocardial infarction by regulating angiogenesis via transcriptional activation of Vegfa. Life Sci 2025; 372:123613. [PMID: 40210117 DOI: 10.1016/j.lfs.2025.123613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
AIMS Our study aims to explore the regulatory role and underlying mechanisms of Lysine-specific demethylase 1 (LSD1) in angiogenesis following myocardial infarction (MI). MATERIALS AND METHODS We generated inducible cardiomyocyte-specific Lsd1 knockout (Lsd1-cKO) mice and established a MI model. The function of LSD1 in cardiac angiogenesis in MI mice was assessed through echocardiography, histopathological staining, and immunofluorescence analysis. In vitro, Lsd1 silencing in cardiomyocytes was achieved by transfecting small interfering RNA (siRNA), followed by hypoxic treatment to simulate the in vivo MI model. The above cardiomyocyte-conditioned medium was collected and used to treat endothelial cells to observe changes in endothelial function. Additionally, we employed Cleavage Under Targets and Tagmentation sequencing (CUT&Tag-seq) to investigate the potential mechanisms by which LSD1 exerts its effects. KEY FINDINGS We found that the absence of LSD1 protected against cardiac dysfunction and promoted angiogenesis in mice with MI. Lsd1-silenced cardiomyocytes enhance the migration and tube formation function of endothelial cells by releasing vascular endothelial growth factor A (VEGF-A) under hypoxic conditions. The combined analysis of CUT&Tag-seq data revealed that silencing of Lsd1 promoted the monomethylation of H3K4 at the Vegfa promoter and region, leading to the transcriptional activation of Vegfa mRNA in cardiomyocytes. SIGNIFICANCE Our research indicates that lowered level of LSD1 in cardiomyocytes enhances VEGF-A paracrine secretion and improves endothelial cell function through cross-talk, ultimately promoting angiogenesis. These findings suggest that targeting LSD1 might be an effective therapeutic approach to protect against MI.
Collapse
Affiliation(s)
- Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingshan Yang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danyi Lv
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenfang Zhou
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qipu Feng
- Animal Experiment Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Xiang G, Shi T, Nwaele CO, Xiao H, Liu Y, Wang Q, Zhang J, Zheng Y. Inhibition of the Sp1/PI3K/AKT signaling pathway exacerbates doxorubicin-induced cardiomyopathy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119960. [PMID: 40246177 DOI: 10.1016/j.bbamcr.2025.119960] [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: 12/26/2024] [Revised: 04/06/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVE This study aimed to investigate the interaction and underlying mechanisms between specificity protein 1 (Sp1) and the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signaling pathway in the context of doxorubicin-induced cardiomyopathy (DIC). METHODS A rat model of DIC was established by intraperitoneal injection of doxorubicin (1 mg/kg) twice a week for eight weeks. Cardiac function was evaluated using echocardiography, and myocardial histopathology was assessed by hematoxylin-eosin (HE) staining. In vitro, H9c2 cardiomyocytes were treated with doxorubicin (2 μmol/L) to induce cardiotoxicity, followed by co-treatment with the Sp1 inhibitor plicamycin or the PI3K/AKT inhibitor LY294002. Cell viability was measured by the CCK-8 assay. Oxidative stress markers, including reactive oxygen species (ROS) and lactate dehydrogenase (LDH), were quantified using flow cytometry and colorimetric assays. Apoptosis was detected via TUNEL staining, and protein expression of Sp1, PI3K, AKT, and Caspase-3 was analyzed by Western blotting. RESULTS Doxorubicin treatment significantly impaired cardiac function in rats, as evidenced by an increase in both left ventricular internal diameters during diastole (LVIDd) and systole (LVIDs), along with decreased ejection fraction (EF) and fractional shortening (FS) (p < 0.01). Myocardial HE staining in doxorubicin-treated rats revealed disorganized cardiomyocyte structures, edema, and cellular necrosis. In vitro, doxorubicin exposure led to reduced H9c2 cell viability, elevated ROS and LDH levels, and increased apoptosis rates (p < 0.01). Western blotting demonstrated that doxorubicin significantly downregulated the expression of Sp1, PI3K, and AKT while upregulating Caspase-3. Inhibition of Sp1 or PI3K/AKT exacerbated these effects, resulting in further cardiac dysfunction, oxidative stress, and apoptosis. Moreover, Sp1 inhibition led to decreased PI3K/AKT pathway activation, while PI3K/AKT inhibition reciprocally suppressed Sp1 expression, indicating a bidirectional regulatory relationship. CONCLUSION Doxorubicin induces cardiotoxicity by promoting oxidative stress and apoptosis through the downregulation of the Sp1/PI3K/AKT signaling pathway. Inhibition of this pathway exacerbates cardiac injury, suggesting that targeting Sp1 and PI3K/AKT may offer novel therapeutic strategies for the prevention and treatment of DIC.
Collapse
Affiliation(s)
- Guojian Xiang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Tingting Shi
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China; Fuzhou Jian Jia Rehabilitation Hospital, Fuzhou 350007, Fujian, China
| | | | - Huazhen Xiao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Yucheng Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Qingfeng Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Jiancheng Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology in South Branch, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China.
| | - Yonghong Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China.
| |
Collapse
|
7
|
Wang J, Ma B, Jiang X, Li C, Lin Z, Wang Y, Shi J, Wang G, Cui C. H 2 protects H9c2 cells from hypoxia/reoxygenation injury by inhibiting the Wnt/CX3CR1 signaling pathway. Med Gas Res 2025; 15:339-347. [PMID: 39511756 PMCID: PMC11918467 DOI: 10.4103/mgr.medgasres-d-24-00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 11/15/2024] Open
Abstract
Myocardial ischemia‒reperfusion injury is a severe cardiovascular disease, and its treatment and prevention are crucial for improving patient prognosis and reducing the economic burden. This study aimed to explore the impact of hydrogen (H 2 ) on hypoxia/reoxygenation (H/R) injury in H9c2 cells (derived from rat embryonic heart tissue) induced by hydrogen peroxide (H 2 O 2 ) and to elucidate its underlying mechanism. An H/R injury model was established in H9c2 cells via exposure to 15 μM H 2 O 2 for 3 hours, followed by incubation in a 5% CO 2 atmosphere at 37°C for 24 hours. Then, the cells were treated with H 2 (50%) for 6, 12 or 24 hours. The results demonstrated that H9c2 cells exposed to H 2 O 2 and subjected to H/R injury presented a marked decrease in the cell survival rate, accompanied by severe morphological alterations, such as curling and wrinkling, and elevated lactate dehydrogenase levels. Notably, H 2 mitigated H/R injury induced by H 2 O 2 in a time-dependent manner, improving the morphological damage observed in H9c2 cells and decreasing lactate dehydrogenase levels. Compared with the model group, treatment with H 2 increased the activities of antioxidant enzymes, including catalase, superoxide dismutase, and glutathione peroxidase, while concurrently reducing the level of malondialdehyde, an indicator of cellular damage. Furthermore, H 2 treatment downregulated the expression of inflammatory cytokines and inflammatory-related factors, specifically interleukin-6, high-mobility group box 1, tumor necrosis factor-alpha, and Toll-like receptor 4, in H9c2 cells post-H/R injury. Furthermore, H 2 treatment resulted in a marked decrease in the expression levels of proteins associated with the Wnt/C-X3-C-motif receptor 1 signaling pathway, such as β-catenin, glycogen synthase kinase-3 beta, adenomatous polyposis coli, and Wnt and C-X3-C-motif receptor 1. This observation suggests a potential mechanism for its protective effects against H/R injury. Therefore, H 2 exerts a protective effect against H/R injury in H9c2 cells induced by H 2 O 2 , potentially by inhibiting the activated Wnt/C-X3-C-motif receptor 1 signaling pathway. This inhibition, in turn, prevents the generation of oxidative stress, inflammatory cytokines, and inflammation-associated factors.
Collapse
Affiliation(s)
- Jingsheng Wang
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Bin Ma
- Department of Cardiovascular Medicine, Taian City Taishan District People’s Hospital, Taian, Shandong Province, China
| | - Xue Jiang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Chao Li
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Yumei Wang
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Jingfei Shi
- Shandong First Medical University, Jinan, Shandong Province, China
| | - Gang Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Chao Cui
- Department of Pharmacy, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong Province, China
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| |
Collapse
|
8
|
Abdullah-Al-Mamun M, Islam D, Roy DC, Ashraf A, Saifullah, Lyzu C, Kabir Talukder ME, Akhter S, Lipy EP, Mohanta LC. Cardioprotective function of mixed spices against myocardial infarction injury: In-vivo and In-silico study. J Genet Eng Biotechnol 2025; 23:100492. [PMID: 40390502 PMCID: PMC12032904 DOI: 10.1016/j.jgeb.2025.100492] [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: 12/10/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/21/2025]
Abstract
Myocardial infarction is the permanent necrosis of heart tissue caused by an artery obstruction. It causes an inadequate delivery of oxygen and nutrients, resulting in muscle injury in the afflicted areas. Here, water extract of mixed spices-onion, garlic, ginger, red chili, turmeric, cumin seed, coriander, cardamom, black pepper, cloves, fenugreek, nigella, cinnamon, and carom seed-was prepared to evaluate cardioprotective function in albino rats. To systematically investigate cardioprotective efficacy, isoproterenol was injected into albino rats to induce myocardial injury. The prepared extract was administrated orally to rats daily for 28 days (200 mg/kg body weight) before infusion of isoproterenol (100 mg/kg body weight) on 29th and 30th days. The induced cardiac injury was significantly ameliorated in rats based on cardiac hypertrophy, histopathology, and Caspase-3 mRNA expression analysis by qRT-PCR. The Indian Medicinal Plants, Phytochemistry And Therapeutics (IMPPAT) chemical database of 820 natural compounds from the mixed spices was then screened against CASP-3 protein using cheminformatics tools, where thymohydroquinone, 4-isopropylbenzoic acid, and 1-naphthylacetic acid were found to be the best interacting ligands, with binding energy scores of -6.112 kcal/mol, -6.206 kcal/mol, and -6.112 kcal/mol, respectively. Notably, thymohydroquinone exhibited the lowest predicted cytotoxicity. Furthermore, molecular dynamic simulation was used to validate the binding stability of the thymohydroquinone with CASP-3 protein compared to CID-6167 (control). Thus, this study explored that mixed spices have cardioprotective effects in rats and identified thymohydroquinone as a natural lead compound against CASP-3, which may pave the way for the development of pharmacotherapy for myocardial damage.
Collapse
Affiliation(s)
- Md Abdullah-Al-Mamun
- Bangladesh Jute Research Institute, Dhaka-1207, Bangladesh; Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh.
| | - Dipa Islam
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research, Dhaka-1205, Bangladesh.
| | - Dipankar Chandra Roy
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research, Dhaka-1205, Bangladesh
| | - Ayesha Ashraf
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh
| | - Saifullah
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Chadni Lyzu
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research, Dhaka-1205, Bangladesh
| | - Md Enamul Kabir Talukder
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Samina Akhter
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research, Dhaka-1205, Bangladesh
| | - Evana Parvin Lipy
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research, Dhaka-1205, Bangladesh
| | - Liton Chandra Mohanta
- Biomedical and Toxicological Research Institute, Bangladesh Council of Scientific and Industrial Research, Dhaka-1205, Bangladesh
| |
Collapse
|
9
|
Ai Z, Li C, Yang Y, Xi X, Li L, Duan S, Jia B, Yang MF, Zhang X. Preclinical Evaluation of a Nanobody-based SPECT Radiotracer 99mTc-AFN with Clinical Potential for Noninvasive Monitoring of Fibroblast Activation after Myocardial Infarction. Radiol Cardiothorac Imaging 2025; 7:e240204. [PMID: 40402089 DOI: 10.1148/ryct.240204] [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] [Indexed: 05/23/2025]
Abstract
Purpose To explore the feasibility of a new technetium 99m-labeled nanobody (99mTc-antifibroblast activation protein nanobody [AFN]) with high affinity to fibroblast activation protein for noninvasive monitoring of fibroblast activation after myocardial infarction. Materials and Methods 99mTc-AFN was prepared by site-specific labeling. Dynamic SPECT/CT imaging from 0.5 to 4 hours after injection of 99mTc-AFN was performed in male C57/BL6 mice at 8 weeks of age with the ischemia-reperfusion (IR) injury on day 7 (n = 4). Furthermore, IR mice underwent 99mTc-AFN SPECT/CT imaging on postoperative days 3, 7, 14, and 28 at 1 hour following injection. Ex vivo SPECT/CT imaging, blocking imaging, and biodistribution studies were performed on day 7 and compared with a 99mTc-labeled fibroblast activation protein inhibitor (FAPI)-04-derived radiotracer (99mTc-HFAPi). Preclinical evaluation of 99mTc-AFN in a swine model of myocardial infarction was performed on day 7. Biodistributions and quantitative results were compared between groups using analysis of variance and t tests. Results The accumulation of 99mTc-AFN at the infarcted region was most clearly visible at 1 hour following injection. 99mTc-AFN uptake on SPECT/CT images in the infarcted myocardium was visible from day 3, and maximum uptake occurred on day 7. At 1 hour after injection, the 99mTc-AFN and 99mTc-HFAPi uptake ratios (means ± SDs) were 2.23 ± 0.82 versus 1.34 ± 0.21 (P = .05), respectively, for infarcted to noninfarcted regions and 1.08 ± 0.49 versus 0.89 ± 0.42 (P = .34) for infarcted region to blood, respectively. Immunohistochemical examinations confirmed that maximum myocardium fibrosis and FAP expression was observed on day 7. 99mTc-AFN uptake in the infarcted myocardium was also clearly visualized with the swine model of myocardial infarction. Conclusion This study demonstrated the potential of 99mTc-AFN for noninvasive monitoring of fibroblast activation after myocardial infarction, which may be a new option for clinical imaging. Keywords: Fibroblast Activation Protein, Nanobody, 99mTc-AFN, 99mTc-HFAPi, Myocardial Infarction Supplemental material is available for this article. © RSNA, 2025.
Collapse
Affiliation(s)
- Zixuan Ai
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Road, Chaoyang District, Beijing 100020, China
| | - Chenzhen Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | | | - Xiaoying Xi
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Road, Chaoyang District, Beijing 100020, China
| | - Lina Li
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Road, Chaoyang District, Beijing 100020, China
| | - Shida Duan
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Road, Chaoyang District, Beijing 100020, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Min-Fu Yang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Road, Chaoyang District, Beijing 100020, China
| | - Xin Zhang
- Department of Nuclear Medicine, Beijing Chaoyang Hospital, Capital Medical University, 8th Gongtinanlu Road, Chaoyang District, Beijing 100020, China
| |
Collapse
|
10
|
Park T, Forbush K, Li Y, Vivas O, Rosenthal KJ, Falcone J, Wong CJ, Bruce JE, Moreno C, Dessauer CW, Scott JD. Long AKAP18 isoforms anchor ubiquitin specific proteinases and coordinate calcium reuptake at the sarcoplasmic reticulum. J Biol Chem 2025:110317. [PMID: 40449590 DOI: 10.1016/j.jbc.2025.110317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 05/20/2025] [Accepted: 05/22/2025] [Indexed: 06/03/2025] Open
Abstract
Subcellular targeting of signaling enzymes influences where and when various modes of intracellular communication operate. Macromolecular complexes of signal transduction and signal termination elements favor reversable control of repetitive processes. This includes adrenergic stimulation of excitation-contraction coupling in the heart. Long isoforms of A-kinase anchoring protein 18 (AKAP18γ and δ) modulate this process via regulation of calcium uptake into the sarcoplasmic reticulum through the Ca2+ATPase 2a (SERCA2a). AKAP18 proximity-proteomic screening in cardiomyocytes identifies networks for protein kinase A (PKA) and ubiquitin-specific proteinases (USP's). A 2'phosphoesterase domain on AKAP18 interfaces with the USP4 isoform at the Z bands of sarcomeres. PKA stimulates USP4 activity in the presence of the anchoring protein. AKAP18 anchored PKA phosphorylates serine 829 on USP4, a conserved residue near the active site of this deubiquitinase. Antibodies against the pSer829 motif show that adrenergic stimulation enhances phosphorylation of USP4 in mouse adult cardiomyocytes. In related studies, elevated USP4 phosphorylation at Ser829 is detected in human post-myocardial infraction tissue as compared to healthy tissue. Thus, phosphorylation of sarcoplasmic USP4 may be a cardioprotective response. Pharmacological inhibition of PKA or deletion of the AKAP7/18 gene in mice decreases calcium flux through the exchanger. This suggests that loss of the anchoring protein impacts SERCA2 action. Thus, AKAP18/PKA/USP4 complexes are well positioned to influence the rate and magnitude of calcium reuptake during the cardiac cycle.
Collapse
Affiliation(s)
- Taeyeop Park
- Department of Integrative Biology and Pharmacology, McGovern Medical School at University of Texas Health Science Center Houston, 6431 Fannin St. Houston, TX, 77030
| | - Katherine Forbush
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195
| | - Yong Li
- Department of Integrative Biology and Pharmacology, McGovern Medical School at University of Texas Health Science Center Houston, 6431 Fannin St. Houston, TX, 77030
| | - Oscar Vivas
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195
| | - Kacey J Rosenthal
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195
| | - Jerome Falcone
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195
| | - Cassandra J Wong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario
| | - James E Bruce
- Department of Genome Sciences, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195
| | - Claudia Moreno
- Howard Hughes Medical Institute, Department of Neurobiology and Biophysics, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at University of Texas Health Science Center Houston, 6431 Fannin St. Houston, TX, 77030
| | - John D Scott
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St. Seattle, WA, 98195.
| |
Collapse
|
11
|
Noorabadi P, Shahabi Rabori V, Jamali S, Jafari N, Saberiyan M. An overview on cardiac regeneration revolution: exploring the promise of stem cell therapies. Mol Biol Rep 2025; 52:511. [PMID: 40434692 DOI: 10.1007/s11033-025-10580-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of global mortality, with myocardial infarction (MI) and subsequent heart failure (HF) posing significant clinical challenges. Despite advancements in pharmacological and surgical interventions, the limited regenerative capacity of the adult human heart necessitates innovative therapeutic strategies. Stem cell-based therapies have emerged as a promising approach to cardiac regeneration, aiming to restore damaged myocardial tissue through cell replacement and paracrine-mediated repair mechanisms. This review provides a comprehensive overview of the current landscape of stem cell therapies for cardiac regeneration, focusing on the molecular mechanisms, cell types, delivery techniques, and recent clinical advancements. We highlight the roles of key signaling pathways, including NOTCH, PI3K/Akt, Wnt/β-catenin, Hippo/YAP, and MAPK, in regulating cardiomyocyte proliferation, angiogenesis, fibrosis, and inflammation. Additionally, we discuss the therapeutic potential of various stem cell types, such as mesenchymal stem cells (MSCs), cardiac progenitor cells (CPCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs), in promoting cardiac repair. Despite promising preclinical results, challenges such as low cell retention, immune rejection, and inconsistent clinical outcomes persist. Recent advancements in genetic engineering, and innovative delivery methods, including transendocardial and intracoronary injections, offer new avenues for enhancing therapeutic efficacy. This review underscores the need for further research to optimize stem cell-based therapies, improve clinical trial design, and translate these innovative approaches into effective treatments for heart disease. By addressing these challenges, stem cell therapy holds the potential to revolutionize cardiac regeneration and improve outcomes for patients with ischemic heart disease and heart failure.
Collapse
Affiliation(s)
- Parisa Noorabadi
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Venus Shahabi Rabori
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sara Jamali
- Department of Medical Genetics, School of Medical Sciences, Faculty of Medicine, Hormozgan University of Medical Sciences, P.O.Box: 7919693116, Bandar Abbas, Iran
| | - Negar Jafari
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, School of Medical Sciences, Faculty of Medicine, Hormozgan University of Medical Sciences, P.O.Box: 7919693116, Bandar Abbas, Iran.
| |
Collapse
|
12
|
Zhang K, Yang K, Zeng L, Xiao Y, Yu J. Rab32 protects mitochondrial function by anchoring Fancd2 and mediates the protective effect of penehyclidine hydrochloride against myocardial ischemia-reperfusion injury. Int Immunopharmacol 2025; 156:114697. [PMID: 40286784 DOI: 10.1016/j.intimp.2025.114697] [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: 12/02/2024] [Revised: 03/14/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Myocardial cell injury resulting from myocardial ischemia and reperfusion is one of the primary drivers behind the onset and progression of heart disease. Penehyclidine hydrochloride (PHC), a novel selective anti-cholinergic agent, exerts a protective effect against myocardial ischemia-reperfusion injury (MIRI). Rab32 belongs to the family of small GTPase proteins. The present study aimed to investigate whether PHC improved MIRI by regulating Rab32 and to explore the underlying mechanisms. Oxygen-glucose deprivation/reoxygenation (OGD/R) and left anterior descending coronary artery ligation were used to establish MIRI models in vitro and in vivo. Here, we showed that PHC upregulated the expression of Rab32 in OGD/R treated HL-1 cells. PHC alleviated OGD/R-induced cell apoptosis and intracellular and mitochondrial ROS levels, while the downregulation of Rab32 exacerbated cell injury. Rab32 was upregulated in MIRI mice and downregulated in OGD/R-induced HL-1 cells. Rab32 overexpression improved cardiac function, reduced the myocardial infarct size, and inhibited cell apoptosis and mitochondrial dysfunction, either in MIRI mice or OGD/R-induced HL-1 cells. On the mechanism, Rab32 interacted with Fancd2 in HL-1 cells. Rab32 downregulation decreased Fancd2 protein expression in mitochondria. Rab32 anchored Fancd2 to mitochondria in OGD/R treated HL-1 cells. Fancd2 knockdown reversed the protective effect of Rab32 on OGD/R-induced HL-1 cells and aggravated cardiomyocyte injury. Finally, the protective role of PHC on MIRI-caused cardiomyocyte injury was confirmed in MIRI mice. Overall, we demonstrated that Rab32 protects mitochondrial function by anchoring Fancd2 and mediates the protective effect of PHC against MIRI.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, PR China
| | - Kehui Yang
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, PR China
| | - Lingyuan Zeng
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, PR China
| | - Yang Xiao
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, PR China
| | - JinGui Yu
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China.
| |
Collapse
|
13
|
Wang C, Teng T, Ma Y, Xiao Y, Zeng M, Fu L, Tang Q. Liquid Metal: A New Approach to Diagnosis and Treatment of Cardiovascular Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2505540. [PMID: 40420682 DOI: 10.1002/adma.202505540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Indexed: 05/28/2025]
Abstract
Cardiovascular diseases (CVDs) remains a leading cause of high mortality and imposes a significant health burden globally. The biocompatibility between materials and the cardiovascular system, encompassing biological safety, modulus matching, and anti-fatigue performance in dynamic physiological environments, has been a critical challenge in the diagnosis and treatment of CVDs. The emergence of liquid metal (LM) offers promising opportunities to develop diagnostic and therapeutic methods that exhibit excellent biocompatibility with the cardiovascular system. In this perspective, the progress of LM applications in contrast agents, nanomedicine, implantable and wearable bioelectronic devices, and bionic materials is evaluated, providing a comprehensive and in-depth discussion of the role and advantages of LM in CVDs management. Finally, the current challenges and future prospects of LM in the field of CVDs diagnosis and treatment are outlined.
Collapse
Affiliation(s)
- Chenlu Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Yuhan Ma
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yang Xiao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Mengqi Zeng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Lei Fu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| |
Collapse
|
14
|
Zhang C, Zhao X, Zhang H, Wang T, Zhang Z, Yin Y, Wang H, Tong X, Xue Y, Zhou Y, Yuan F, Bian X, Wei H, Huang Y, Liu T. Gut Microbiota Modulation by Lysozyme as a Key Regulator of Vascular Inflammatory Aging. RESEARCH (WASHINGTON, D.C.) 2025; 8:0704. [PMID: 40416362 PMCID: PMC12099208 DOI: 10.34133/research.0704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/27/2025]
Abstract
Vascular inflammatory aging is strongly associated with multimorbidity, including immunosenescence. Here, bioinformatic analysis indicated elevated expression of the lysozyme (LYZ) gene in age-dependent vascular diseases. Lyz1 deficiency led to vascular inflammatory aging, including damage to indicators related to oxidative stress, vascular function, and inflammation in the serum and vascular tissues of wild-type (WT) and Lyz1-/- mice. The 16S ribosomal RNA sequencing of intestinal contents revealed increased Bifidobacterium and its metabolism of acetate, butyrate, omega-muricholic acid, propionate, and valeric acid in Lyz1-/- mice compared with that in WT mice. Additionally, RNA sequencing of vascular tissues identified differentially expressed genes in Lyz1-/- mice compared with those in WT mice, as well as enrichment of the common phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. Vascular inflammatory aging phenotypes were detected in the blood vessels of antibiotic-treated and germ-free mice, and the PI3K-Akt signaling pathway was inhibited. Importantly, intravenous LYZ administration worsened the pathological conditions, whereas oral LYZ administration successfully restored the gut microbial balance and reversed the vascular inflammatory aging phenotypes. Collectively, this study establishes LYZ as a novel biomarker for age-related vascular diseases and the gut microbiota-PI3K-Akt axis as a promising therapeutic target.
Collapse
Affiliation(s)
- Chenyang Zhang
- Institute of Integrated Traditional Chinese and Western Medicine,
Affiliated Hospital of Jiangnan University, Wuxi 214122, China
- Wuxi School of Medicine,
Jiangnan University, Wuxi 214122, China
| | - Xin Zhao
- Department of Rehabilitation Treatment,
Jiangsu Rongjun Hospital, Wuxi 214062, China
| | - Hang Zhang
- Department of Pathology,
Army Medical University, Chongqing 400038, China
- Yu-Yue Pathology Scientific Research Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Tongtong Wang
- Wuxi School of Medicine,
Jiangnan University, Wuxi 214122, China
| | - Zhenyu Zhang
- Wuxi School of Medicine,
Jiangnan University, Wuxi 214122, China
| | - Yilin Yin
- Wuxi School of Medicine,
Jiangnan University, Wuxi 214122, China
| | - Hui Wang
- Wuxi School of Medicine,
Jiangnan University, Wuxi 214122, China
| | - Xiao Tong
- Department of Pediatrics,
Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Yuzheng Xue
- Department of Gastroenterology,
Affiliated Hospital of Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yahong Zhou
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, Jiangsu, China
| | - Fenglai Yuan
- Institute of Integrated Traditional Chinese and Western Medicine,
Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Xiuwu Bian
- Department of Pathology,
Army Medical University, Chongqing 400038, China
- Yu-Yue Pathology Scientific Research Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Hong Wei
- Yu-Yue Pathology Scientific Research Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Yuan Huang
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences,
Peking Union Medical College, Beijing 100037, China
| | - Tianhao Liu
- Institute of Integrated Traditional Chinese and Western Medicine,
Affiliated Hospital of Jiangnan University, Wuxi 214122, China
- Wuxi School of Medicine,
Jiangnan University, Wuxi 214122, China
- Department of Pathology,
Army Medical University, Chongqing 400038, China
- Yu-Yue Pathology Scientific Research Center, Jinfeng Laboratory, Chongqing 401329, China
- Department of Gastroenterology,
Affiliated Hospital of Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
15
|
Xu R, Yan C, Cao G. RPS27A as a potential clock-related diagnostic biomarker for myocardial infarction: Comprehensive bioinformatics analysis and experimental validation. Clinics (Sao Paulo) 2025; 80:100677. [PMID: 40409241 DOI: 10.1016/j.clinsp.2025.100677] [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: 11/08/2024] [Revised: 02/05/2025] [Accepted: 04/13/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND The circadian system plays a crucial role in managing cardiovascular functions, with disturbances in this system associated with Myocardial Infarction (MI). Despite this connection, the exact mechanisms by which clock genes influence MI occurrence are not well-defined. This research focused on investigating the link between clock genes and MI. METHODS The authors examined MI microarray datasets (GSE151412 and GSE60993) from the GEO database, concentrating on Differentially Expressed Genes (DEGs) associated with the circadian system. To clarify critical biological functions and pathways, the authors performed enrichment analyses using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Through Lasso regression, the authors pinpointed hub genes and confirmed their relevance using both the GSE66360 dataset and quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR). Furthermore, the authors conducted single-Gene Set Enrichment Analysis (GSEA) to reveal pathways linked to the hub gene. The analysis extended to exploring drug interactions and networks involving competing endogenous RNA (ceRNA). RESULTS The present analysis identified ten clock genes associated with circadian rhythms that showed differential expression between MI patients and healthy controls. Enrichment analysis suggested these genes' roles in pathways like the Gap junction and circadian rhythm pathways. Following Lasso regression and validation, RPS27A was identified as the main hub gene. GSEA further highlighted enriched pathways, such as mismatch repair. Additionally, immune infiltration analysis revealed notable differences in B-cell and CD4+ T-cell populations between the MI group and the control group. CONCLUSION The present findings suggest that the clock-related gene RPS27A is associated with MI, potentially influencing its development through circadian rhythm regulation. These results enhance the understanding of MI pathogenesis and may offer new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Rui Xu
- Department of Cardiology, Fifth Affiliated Hospital of Xinjiang Medical University, China
| | - Changshun Yan
- Department of Cardiology, Fifth Affiliated Hospital of Xinjiang Medical University, China
| | - GuiQiu Cao
- Department of Cardiology, Fifth Affiliated Hospital of Xinjiang Medical University, China.
| |
Collapse
|
16
|
Tian Y, Liu X, Chen L, Zeng T, Gu T, Xu W, Ren J, Lu L. Dietary resveratrol alleviates liver and intestinal injury in ducks under cage rearing system. Poult Sci 2025; 104:105330. [PMID: 40449104 DOI: 10.1016/j.psj.2025.105330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 05/21/2025] [Accepted: 05/21/2025] [Indexed: 06/02/2025] Open
Abstract
Cage rearing is a promising farming method. However, our previous studies have demonstrated that changes in farming practices induce oxidative stress and inflammation in the liver and duodenum of ducks. Resveratrol (RES), a natural plant polyphenol, possesses antioxidant, anti-inflammatory, and cytoprotective properties. This study evaluated the alleviating effects of RES against cage-rearing-induced duck health problems, emphasizing the involvement of redox imbalance, inflammatory response, endoplasmic reticulum (ER) stress, apoptosis, and PI3K/AKT and MAPK/ERK pathways. A total of 120 healthy 12-week-old female ducks were transferred to a cage system and randomly assigned to two dietary RES groups with 6 replicates each (10 ducks per replicate), including basal diet + 0 mg/kg RES (control group, CON), and basal diet + 500 mg/kg RES (RES-treated group, RES). During the early stages (within 10 days) of cage rearing, blood, liver, and duodenal samples were collected for analysis. The results demonstrated that RES reduced histopathological damage in the liver and duodenum of cage-reared ducks. It also reduced serum albumin levels, increased serum aspartate aminotransferase and alanine aminotransferase levels, and enhanced antioxidant (increased CAT, GSH-Px, SOD, and T-AOC activities in the serum, liver, and duodenum, and reduced the increase in MDA) and anti-inflammatory properties (reduced pro-inflammatory cytokines interleukin (IL)-1β and IL-6 secretion and increased anti-inflammatory cytokine IL-4 levels). Additionally, quantitative real-time polymerase chain reaction revealed that RES intervention reversed the abnormal mRNA abundance of biomarkers associated with inflammatory injury (iNOS and COX2) in the liver, and ER stress (GRP78) and apoptosis (Bax and Bcl2) in the liver and duodenum of cage-reared ducks. Further analysis of key proteins in the PI3K/AKT and ERK MAPK signaling pathways revealed that RES promoted AKT phosphorylation in the liver and duodenum of cage-reared ducks and reduced cleaved caspase-3 protein content. Overall, RES prevents cage-rearing stimuli-induced liver and intestinal injury in ducks by enhancing liver function, improving antioxidant properties, inhibiting inflammation, ER stress, and apoptosis, and activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China
| | - Xiangshan Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Li Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China
| | - Jindong Ren
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China; Zhejiang Provincial Key Laboratory of Livestock and Poultry Biotech Breeding, Hangzhou, 310021, China.
| |
Collapse
|
17
|
Zhang Z, Zheng L, Chen Y, Chen Y, Hou J, Xiao C, Zhu X, Zhao SM, Xiong JW. AARS2 ameliorates myocardial ischemia via fine-tuning PKM2-mediated metabolism. eLife 2025; 13:RP99670. [PMID: 40371904 PMCID: PMC12080999 DOI: 10.7554/elife.99670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
AARS2, an alanyl-tRNA synthase, is essential for protein translation, but its function in mouse hearts is not fully addressed. Here, we found that cardiomyocyte-specific deletion of mouse AARS2 exhibited evident cardiomyopathy with impaired cardiac function, notable cardiac fibrosis, and cardiomyocyte apoptosis. Cardiomyocyte-specific AARS2 overexpression in mice improved cardiac function and reduced cardiac fibrosis after myocardial infarction (MI), without affecting cardiomyocyte proliferation and coronary angiogenesis. Mechanistically, AARS2 overexpression suppressed cardiomyocyte apoptosis and mitochondrial reactive oxide species production, and changed cellular metabolism from oxidative phosphorylation toward glycolysis in cardiomyocytes, thus leading to cardiomyocyte survival from ischemia and hypoxia stress. Ribo-Seq revealed that Aars2 overexpression increased pyruvate kinase M2 (PKM2) protein translation and the ratio of PKM2 dimers to tetramers that promote glycolysis. Additionally, PKM2 activator TEPP-46 reversed cardiomyocyte apoptosis and cardiac fibrosis caused by AARS2 deficiency. Thus, this study demonstrates that AARS2 plays an essential role in protecting cardiomyocytes from ischemic pressure via fine-tuning PKM2-mediated energy metabolism, and presents a novel cardiac protective AARS2-PKM2 signaling during the pathogenesis of MI.
Collapse
Affiliation(s)
- Zongwang Zhang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityBeijingChina
| | - Lixia Zheng
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityBeijingChina
| | - Yang Chen
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityBeijingChina
| | - Yuanyuan Chen
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityBeijingChina
| | - Junjie Hou
- School of Basic Medical Sciences and The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Chenglu Xiao
- School of Basic Medical Sciences and The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityBeijingChina
| | - Shi-Min Zhao
- Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan UniversityShanghaiChina
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking UniversityBeijingChina
- School of Basic Medical Sciences and The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| |
Collapse
|
18
|
Gao Y, Meng X, Zhu H, Zao X, Wu D, Guo Q, Li X, Dong H, Zhang D. Exosomes Derived from the Serum of Mice That Received a Huoxue Yiqi Recipe Promote Angiogenesis Following Myocardial Infarction. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40358553 DOI: 10.1021/acsami.5c02784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Proangiogenic therapy offers a promising strategy for treating and preventing heart failure and cardiac remodeling following a myocardial infarction (MI). Although exosome-based proangiogenic therapy has significant potential in regenerative medicine and MI treatment, its application remains limited by suboptimal therapeutic efficacy. Here, we present exosomes (HXYQR-Exo) derived from the serum of mice treated with the Huoxue Yiqi Recipe (HXYQR) to promote angiogenesis and repair cardiac tissue post-MI, with a systematic elucidation of the underlying mechanisms. Our findings show that HXYQR-Exo incorporates pharmaceutically active components of HXYQR, enhancing the proliferation, invasion, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) under hypoxic conditions. In vivo studies demonstrate significant improvements in cardiac function and angiogenesis. Mechanistic investigations reveal that these effects are mediated through the activation of the HIF-1α/VEGF, Focal Adhesion Kinase (FAK), and p38/Mitogen-Activated Protein Kinase-Activated Protein Kinase (MAPKAPK)/Heat Shock Protein 27 (HSP27) pathways. This study introduces an exosome-based approach for MI treatment and cardiac repair, offering an effective strategy to enhance exosome biological activities and functions via traditional Chinese medicine preconditioning.
Collapse
Affiliation(s)
- Yijie Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
- Beijing Key Laboratory for Bioengineering and Sensing Technology Research Centre for Bioengineering and Sensing Technology School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 10083, P. R. China
- National Center for Integrative Medicine; Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiangdan Meng
- Beijing Key Laboratory for Bioengineering and Sensing Technology Research Centre for Bioengineering and Sensing Technology School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 10083, P. R. China
| | - Haiyan Zhu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Xiaobin Zao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Dandan Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Qianqian Guo
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Xianlun Li
- National Center for Integrative Medicine; Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Haifeng Dong
- Beijing Key Laboratory for Bioengineering and Sensing Technology Research Centre for Bioengineering and Sensing Technology School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 10083, P. R. China
- Marshall Laboratory of Biomedical Engineering, Shenzhen Key Laboratory for Nano-Biosensing Technology, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, P. R. China
| | - Dongmei Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| |
Collapse
|
19
|
Handa K, Kawamura M, Sasai M, Matsuzaki T, Harada A, Fujimura L, Whitehouse J, Saito S, Komukai S, Kitamura T, Fujishiro A, Hirano K, Miki K, Miyagawa S. Effective transcatheter intracoronary delivery of mRNA-lipid nanoparticles targeting the heart. J Control Release 2025; 381:113623. [PMID: 40073941 DOI: 10.1016/j.jconrel.2025.113623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Messenger RNA (mRNA) has great potential to provide innovative medical solutions in the treatment of heart failure. Although lipid nanoparticles (LNPs) are an established mRNA delivery system, effectively delivering LNPs to the heart remains a significant challenge. Here, we evaluated the efficacy of transcatheter intracoronary (IC) administration compared to intravenous (IV) and intramyocardial (IM) administration in normal and ischemia-reperfusion (I/R) model rabbit hearts using LNPs encapsulating Firefly Luciferase (FLuc) mRNA. In the normal model, IVIS spectrum data showed that FLuc expression was widespread throughout the heart in the IC group and was significantly higher than in the IV group, and comparable to the IM group, where it was highly expressed only at the injection sites. Histological analysis revealed that FLuc-expressing cells were observed in cardiomyocytes, endothelial cells, smooth muscle cells, and fibroblasts. In the I/R model, FLuc expression was also significantly higher in the IC group than the IV group, and comparable to the IM group. Although FLuc expression was strongly observed in the infarct area in all three delivery groups, the IC group demonstrated the most widespread FLuc expression in the remote area. Histological analysis revealed significantly higher FLuc-expressing cells in the remote area in the IC group than in the other groups. IC administration effectively delivered mRNA-LNPs not only to the infarct area (damaged area) but also to the remote area (non-damaged area) in the diseased heart. Moreover, VEGF mRNA-LNP administration via the IC method to I/R model rabbit hearts significantly reduced the infarct area and attenuated the impairment of cardiac function caused by I/R injury compared to the other methods. Considering the invasiveness and clinically limited applications of IM administration, our study suggests that less invasive IC administration is a clinically safe and useful method for mRNA-LNP delivery to a wider range of myocardial tissue in the heart.
Collapse
Affiliation(s)
- Kazuma Handa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masashi Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Masao Sasai
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Matsuzaki
- Department of DDS Pharmaceutical Development, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Lisa Fujimura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Julia Whitehouse
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shunsuke Saito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sho Komukai
- Division of Biomedical Statistics, Department of Integrated Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuhisa Kitamura
- Department of Social and Environmental Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Kunio Hirano
- Terumo Corporation, Innovation Center, Kanagawa, Japan
| | - Kenji Miki
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan.
| |
Collapse
|
20
|
Ma E, Zhang J, Tang Y, Fang X, Wang C, Wu C, Zhu W, Wo D, Ren DN. Liensinine Prevents Acute Myocardial Ischemic Injury via Inhibiting the Inflammation Response Mediated by the Wnt/β-Catenin Signaling Pathway. Int J Mol Sci 2025; 26:4566. [PMID: 40429711 PMCID: PMC12110967 DOI: 10.3390/ijms26104566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Myocardial infarction (MI) is characterized by the sudden reduction in myocardial blood flow and remains the leading cause of death worldwide. Because MI causes irreversible damage to the heart, discovering drugs that can limit the extent of ischemic damage is crucial. Liensinine (LSN) is a natural alkaloid that has exhibited beneficial effects in various cardiovascular diseases, including MI; however, its molecular mechanisms of action remain largely unelucidated. In this study, we constructed murine models of MI to examine the potential beneficial effects and mechanisms of LSN in myocardial ischemic injury. Murine models of MI in wild-type and cardiomyocyte-specific β-catenin knockout mice were used to explore the role of LSN and Wnt/β-catenin signaling in MI-induced cardiac injuries and inflammatory responses. The administration of LSN markedly improved cardiac function and decreased the extent of ischemic damage and infarct size following MI. LSN not only prevented excessive inflammatory responses but also inhibited the aberrant activation of Wnt/β-catenin signaling, two factors that are critically involved in the exacerbation of MI-induced injury. Our findings provide important new mechanistic insight into the beneficial effect of LSN in MI-induced cardiac injury and suggest the therapeutic potential of LSN as a novel drug in the treatment of MI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Da Wo
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatric, 1 Qiuyang Road, Minhou, Fuzhou 350122, China; (E.M.); (J.Z.); (Y.T.); (X.F.); (C.W.); (C.W.); (W.Z.)
| | - Dan-ni Ren
- Academy of Integrative Medicine, College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatric, 1 Qiuyang Road, Minhou, Fuzhou 350122, China; (E.M.); (J.Z.); (Y.T.); (X.F.); (C.W.); (C.W.); (W.Z.)
| |
Collapse
|
21
|
Wang C, Zhu F, Zhou L, Zhang S, Wang R, Tian H, Zhang B, Wu J, Xu X, Jiang R, Hou X, Liu J, Tian W. Krüppel like factor 7 regulates mitochondrial dynamics balance in myocardial infarction. Commun Biol 2025; 8:722. [PMID: 40346382 PMCID: PMC12064834 DOI: 10.1038/s42003-025-08139-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
Targeting the balance of mitochondrial fission and fusion can effectively alleviate the cardiac energy supply efficiency, to restore cardiac systolic dysfunction and reduce mortality. We previously found that Klf7 is closely related to cardiac energy metabolism. Here we generated cardiomyocyte-specific Klf7 knockout and overexpression mice that underwent myocardial infarction (MI) surgery. Klf7 expression increased in the ischemic myocardium of mice, and cardiomyocyte-specific knockout Klf7 significantly lowered the mortality of MI-inflicted mice and improved ATP insufficiency in MI. Subsequently, Klf7 overexpression aggravated adverse cardiac remodeling and mitochondrial fission and fusion imbalance after MI. Our results also demonstrated that Klf7 inhibited mitochondrial fusion and promoted mitochondrial fission by targeting prohibitin 2 (Phb2) and mitofusin 2 (Mfn2). Our study revealed a crucial role in upholding the overall balance of mitochondrial fission and fusion during MI. Furthermore, our findings indicated that the Klf7/Mfn2/Phb2 axis holds promise as a potential target for therapeutic interventions of MI.
Collapse
Affiliation(s)
- Cao Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Fuxing Zhu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Lan Zhou
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 150001, China, Harbin
| | - Situo Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Ruiqi Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Hui Tian
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Bosong Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Jiahui Wu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Xiang Xu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Ruixian Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Xiaolu Hou
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Jian Liu
- School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin, 150080, China
| | - Weiming Tian
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China.
| |
Collapse
|
22
|
Xu S, Li H, Han J, Xu Y, Li N, Che W, Liu F, Yue W. Klf9 promotes the repair of myocardial infarction by regulating macrophage recruitment and polarization. JCI Insight 2025; 10:e187072. [PMID: 40198141 DOI: 10.1172/jci.insight.187072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
The inflammatory response after myocardial infarction (MI) is a precisely regulated process that greatly affects subsequent wound healing and remodeling. However, understanding about the process is still limited. Macrophages are critically involved in inflammation resolution after MI. Krüppel-like factor 9 (Klf9) is a C2H2 zinc finger-containing transcription factor that has been implicated in glucocorticoid regulation of macrophages. However, the contribution of Klf9 to macrophage phenotype and function in the context of MI remains unclear. Our study revealed that KLF9 deficiency resulted in higher mortality and cardiac rupture rate, as well as a considerable exacerbation in cardiac function. Single-cell RNA sequencing and flow cytometry analyses revealed that, compared with WT mice, Klf9-/- mice displayed excessive neutrophil infiltration, insufficient macrophage infiltration, and a reduced proportion of monocyte-derived CD206+ macrophages after MI. Moreover, the expression of IFN-γ/STAT1 pathway genes in Klf9-/- cardiac macrophages was dysregulated, characterized by insufficient expression at 1 day post-MI and excessive expression at day 3 post-MI. Mechanistically, Klf9 directly binds to the promoters of Stat1 gene, regulating its transcription. Overall, these findings indicate that Klf9 beneficially influences wound healing after MI by modulating macrophage recruitment and differentiation by regulating the IFN-γ/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Sheng Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Han
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Niannian Li
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Liu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenhui Yue
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Zhang R, Wang Y, Jiang H, Aheniyazi A, Tao J, Li J, Yang Y. Therapeutic Angiogenesis Mediated by Traditional Chinese Medicine: Advances in Cardiovascular Disease Treatment. JOURNAL OF ETHNOPHARMACOLOGY 2025:119871. [PMID: 40345269 DOI: 10.1016/j.jep.2025.119871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/26/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicine (TCM) shows growing potential as an adjunct or alternative therapy for vascular occlusion diseases (e.g., stroke, peripheral artery disease) by promoting therapeutic angiogenesis to restore blood flow in ischemic regions while minimizing side effects. AIMS OF THE STUDY This review examines TCM-mediated angiogenesis mechanisms and therapeutic advances in vascular occlusion management, establishing a theoretical foundation for clinical translation and precision medicine development. MATERIALS AND METHODS We systematically analyzed PubMed articles on TCM-induced angiogenesis in vascular occlusion diseases, focusing on herbal formulations, single herbs, bioactive compounds, and their associated signaling pathways. Search PubMed for studies investigating the role of Chinese herbal medicine (TCM), natural compounds, and herbal medicine in angiogenesis, while excluding research related to cancer, tumor, or oncological contexts. RESULTS TCM formulas, individual herbs, and monomeric compounds enhance endothelial cell proliferation, migration, and tube formation via pathways such as HIF/VEGF, PI3K/AKT, NOTCH, BMP/ALK, and Apelin/APJ, improving ischemic blood flow. CONCLUSION This review highlights angiogenesis as a novel strategy for vascular occlusive diseases and underscores TCM's efficacy through multi-target angiogenic regulation mechanism.However, further research using modern medical technologies is needed to optimize clinical application and advance precision medicine.
Collapse
Affiliation(s)
- Rong Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Yunze Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Haoyan Jiang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Aliyanmu Aheniyazi
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Jin Tao
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China.
| | - Yining Yang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China.
| |
Collapse
|
24
|
Xian R, Xian H, Dong H, Lin J, Zhuang X, Zou Y, Xie Q, Liang Y, Li S. Black Phosphorus-Loaded Gelatin Methacryloyl Hydrogels Enhance Angiogenesis via Activation of the PEAK1-MAPK Pathway. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26371-26385. [PMID: 40272250 DOI: 10.1021/acsami.5c02054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Repair and regeneration of oral and maxillofacial tissue defects remain significant challenges, mainly due to the limitations of existing treatment approaches. Conventional methods such as transplantation, tissue scaffolds, growth factors, and stem cell therapies often face obstacles, including donor shortages, insufficient vascularization, and safety concerns. There is an urgent need for innovative therapeutic strategies to effectively promote vascular regeneration while minimizing complications. Black phosphorus nanosheets (BPNSs) and hydrogels present significant advantages and broad application potential as tissue regeneration carriers due to their biocompatibility, degradability, and controlled drug release properties. By combining various characterization techniques and detection methods, we conducted a thorough analysis of BPNSs and gelatin methacryloyl (GelMA) scaffolds loaded with BPNSs (BP-GelMA). The results indicate that this study successfully prepared BPNSs with uniform size, good dispersion, and intact structure. Moreover, the BP-GelMA composite demonstrated excellent swelling behavior and structural stability while effectively enabling the controlled release of BPNSs. This study investigated the angiogenic effects of BP-GelMA at concentrations of 0, 12.5, and 25.0 μg/mL. In vitro experiments showed that BP-GelMA significantly enhanced endothelial cell proliferation, migration, and tube formation. In vivo results demonstrated that 12.5 μg/mL and 25.0 μg/mL BP-GelMA did not induce significant developmental toxicity in zebrafish and effectively promoted neovascularization. RNA-Seq analysis revealed that BP-GelMA activates angiogenesis-related biological processes. Mechanistic studies identified PEAK1 as a central regulator, driving vascular formation through activation of the MAPK signaling pathway. These findings highlight the potential of BP-GelMA as a therapeutic strategy for promoting angiogenesis and underscore the importance of optimizing BP-GelMA concentrations to achieve maximum therapeutic efficacy and safety in clinical applications.
Collapse
Affiliation(s)
- Ruoting Xian
- Center of Oral Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Hongyi Xian
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hao Dong
- The Department of Pathology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Jiating Lin
- Center of Oral Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Xianxian Zhuang
- The Department of Stomatology Center, The People's Hospital of Baoan Shenzhen, Shenzhen, Guangdong 518100, China
| | - Yue Zou
- Center of Oral Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Qinkai Xie
- Center of Oral Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Youde Liang
- The Department of Stomatology Center, The People's Hospital of Baoan Shenzhen, Shenzhen, Guangdong 518100, China
| | - Shaobing Li
- Center of Oral Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
25
|
Zheng H, Fan S, Zang H, Luo J, Shu L, Peng J. A comprehensive analysis identified an autophagy-related risk model for predicting recurrence and immunotherapy response in stage I lung adenocarcinoma. PeerJ 2025; 13:e19366. [PMID: 40330698 PMCID: PMC12051938 DOI: 10.7717/peerj.19366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is characterized by early recurrence and poor prognosis. Autophagy is a double-edged sword in tumor development and anti-tumor therapy resistance. However, the prediction of relapse and therapeutic response in LUAD patients with stage I based on the signature of autophagy remains unclear. Methods Gene expression data were obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) database. Autophagy-associated genes were extracted from the Human Autophagy Moderator Database. The autophagy score was established by Least Absolute Shrinkage and Selection Operator (LASSO) regression. Real-time PCR was used to detect gene expression of hub genes in LUAD patients. Protein-protein interaction (PPI) was analyzed to identify crucial genes. Gene set enrichment analysis (GSEA) was used to reveal the molecular features of patients. ESTIMATE algorithm was applied to estimate the tumor immune infiltration. TIDE score and Genomics of Drug Sensitivity in Cancer (GDSC) database were used to assess therapeutic response. Results We established an autophagy score based on 19 autophagy genes. Among these genes, MAP1LC3B played a crucial role in PPI network and was down-regulated in tumor tissues both in TCGA and local cohort. Receiver operating characteristic (ROC) curve showed that the risk model effectively predict RFS of stage I LUAD (area under the curve (AUC) at 1, 2, 3 years = 0.701, 0.836, and 0.818, respectively). Multivariate regression analysis indicated that the autophagy score was an independent predictor for relapse (P < 0.001, HR = 4.8, 95% CI [3.25-7.2]). The autophagy score also showed great predictive efficacy in the external validation GEO cohorts. GSEA revealed gene sets significantly enriched in immunity, cell cycle, and adhesion, etc. Meanwhile, we found the autophagy score was negatively related to KRAS mutation (P = 0.017) but positively associated with TP53 mutation (P = 6.4e-11). The autophagy score had a negative relationship with CD8+, CD4+ T cell, and dendritic cell, and positively correlated with immune checkpoint molecule CD276. Patients with a high autophagy score were sensitive to chemotherapy and targeted therapy, while resistant to immune checkpoint inhibitors. Conclusion We constructed an effective recurrence risk predictive model for stage I LUAD patients based on autophagy related genes. High autophagy score predicted a higher recurrence risk and suppressing tumor immune microenvironment.
Collapse
Affiliation(s)
- Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Long Shu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Kar A, Gupta S, Matilal A, Sarkar S. Tissue engineering with targeted delivery of nanotized S-nitrosyl mutant of NEMO ameliorates myocardial infarction. Nanomedicine (Lond) 2025; 20:1085-1099. [PMID: 40235354 PMCID: PMC12068353 DOI: 10.1080/17435889.2025.2491989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Myocardial infarction (MI) is characterized by an elevated nitrosative and hypoxic microenvironment due to reduced coronary blood flow. NEMO (IKKγ) regulates the formation of the IKK holo-complex to activate NFκB-p65 signaling. This study reports successful restoration of MI through cardiomyocyte-targeted nanotized S-nitrosyl mutant of NEMO under elevated nitrosative stress. METHODS The MI model was generated in male Wistar rats. S-nitrosyl mutant of NEMO (R- NEMO) was selectively delivered to the cardiomyocytes via targeted chitosan nano-vehicle. RESULTS Nano-conjugated R- NEMO delivery to diseased cardiomyocytes resulted in downregulation of nitrosative stress and cellular apoptosis leading to regressed infarct area with improved cardiac pathophysiology. Mechanistically, NEMO-p300 binding in R- NEMO expressed cells destabilized p65-p300 complex leading to regressed nitrosative stress and cellular apoptosis. The NEMO mutant inhibits the PGC1α-p65 complex-mediated degradation of PGC1α, leading to upregulation of VEGF. A shift in the binding preference of p65 from PGC1α/p300 to HDAC1 results in the downregulation of the cell-cycle inhibitor and the induction of cell-cycle re-entry markers during MI. CONCLUSION Tissue-targeted R- NEMO nanoconjugates show potential to ameliorate MI insult by downregulating apoptosis and promoting the proliferative prowess of the resident cardiomyocytes with potential revascularization at infarct sites; thus, repairing the damaged myocardium.
Collapse
Affiliation(s)
- Abhik Kar
- Department of Zoology, University of Calcutta, Kolkata, India
| | | | | | | |
Collapse
|
27
|
Zhang G, Dong X, Sun B, Zhou Z, Xu Y, Huang Y, Meng S, Cao Z, Qin N, Zhu Y, Yu L, Wang H. Matrine Inhibits the Wnt3a/β-Catenin Signalling to Attenuate Pressure Overload-Induced Atrial Remodelling and Vulnerability to Atrial Fibrillation. J Cell Mol Med 2025; 29:e70617. [PMID: 40407710 PMCID: PMC12101071 DOI: 10.1111/jcmm.70617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/17/2025] [Accepted: 05/12/2025] [Indexed: 05/26/2025] Open
Abstract
Atrial fibrillation (AF) is closely associated with atrial electrical and structural remodelling, yet effective drug strategies remain limited. Matrine (MAT), the active compound in Sophora flavescens, has shown anti-AF effects, but its mechanisms are unclear. This study explored MAT's impact on pressure overload-induced AF using clinical samples, bioinformatics, network pharmacology and murine models, focusing on the canonical Wnt signalling. A murine pressure overload model was established via transverse aortic constriction (TAC) surgery for 4 weeks. Programmed electrical stimulation, langendorff perfusion, echocardiography, Masson's trichrome staining and western blotting were used to evaluate the potential effects and mechanisms of MAT. The results demonstrated that TAC-induced atrial electrical and structural remodelling significantly increased susceptibility to AF in mice while also up-regulated atrial Wnt3a/β-catenin signalling as well as markers for remodelling and inflammation, which were partially supported by clinical samples. MAT dose-dependently mitigated atrial structural and electrical remodelling. Furthermore, MAT intervention inhibited Wnt3a/β-catenin signalling. However, co-administration of SKL2001, a Wnt/β-catenin agonist, counteracted MAT's benefits. The overall findings suggest that MAT treatment may serve as a potential therapeutic approach for inhibiting TAC-induced atrial electrical and structural remodelling by suppressing Wnt3a/β-catenin signalling pathways, thereby reducing susceptibility to AF.
Collapse
Affiliation(s)
- Guoxin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Xue Dong
- The Third Outpatient DepartmentGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Boxuan Sun
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Zijun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Yinli Xu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Yuting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Shan Meng
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
- Jinzhou Medical UniversityJinzhouLiaoningChina
| | - Zijun Cao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Nana Qin
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
- Liaoning University of Traditional Chinese MedicineShenyangLiaoningChina
| | - Yan Zhu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Huishan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| |
Collapse
|
28
|
Meng F, Bai H, Ke K, Fang L, Huang H, Liang X, Li W, Chen X, Chen C. tRF5-22-SerGCT-1 protects the heart against myocardial injury by targeting MSK1. Epigenomics 2025; 17:439-451. [PMID: 40269521 PMCID: PMC12026222 DOI: 10.1080/17501911.2025.2495544] [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: 12/01/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
AIM This study aims to explore the expression profiles and potential functions of tsRNAs in MI. METHODS Using a mouse model of MI induced by coronary artery ligation, we used smallRNA array to obtain tsRNAs expression profiles. Reverse transcription quantitative polymerase chain reaction(RT-qPCR), Western Blot, tRF5-22-SerGCT-1 mimics and inhibitors, cell proliferation and apoptosis detection, luciferase reporter assay, and bioinformatics analysis were employed to screen differentially expressed tsRNAs and identify the functions of tsRNAs after MI. RESULTS A total of 175 significantly different tsRNAs (FC > 1.5, p < 0.05) were identified in MI mice, including 98 upregulated and 77 downregulated tsRNAs. Bioinformatics and target gene prediction revealed that two differentially expressed tsRNAs (5'tiRNA-34-GlnCTG-4, tRF5-22-SerGCT-1) may be involved in processes like autophagy and apoptosis, as well as in key signaling pathways such as MAPK and autophagy. Further investigation of tRF5-22-SerGCT-1 revealed that its overexpression or inhibition in vitro affected MSK1 levels and cardiomyocytes apoptosis following oxygen-glucose deprivation, providing a protective effect. Dual-luciferase assays confirmed that tRF5-22-SerGCT-1 targets MSK1. CONCLUSION We found differentially expressed tsRNAs in MI. In addition, our research showed first that tRF5-22-SerGCT-1 might be involved in the MAPK pathways by targeting the MSK1, modulating apoptosis.
Collapse
Affiliation(s)
- Fanji Meng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics and School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Hemanyun Bai
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Kangling Ke
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Lingyan Fang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Haitao Huang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Xiao Liang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Weiyan Li
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Xiongwen Chen
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics and School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Can Chen
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
29
|
Song C, Yang J, Gu Z. Latest developments of microphysiological systems (MPS) in aging-related and geriatric diseases research: A review. Ageing Res Rev 2025; 107:102728. [PMID: 40058462 DOI: 10.1016/j.arr.2025.102728] [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: 12/02/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Aging is a gradual and irreversible process accompanied by the decline in tissue function and a significantly increased risk of various aging-related and geriatric diseases. Especially in the paradoxical context of accelerated global aging and the widespread emergence of pandemics, aging-related and geriatric diseases have become leading causes of individual mortality and disability, drawing increasing attention from researchers and investors alike. Despite the utility of current in vitro systems and in vivo animal models for studying aging, these approaches are limited by insurmountable inherent constraints. In response, microphysiological systems (MPS), leveraging advances in tissue engineering and microfluidics, have emerged as highly promising platforms. MPS are capable of replicating key features of the tissue microenvironment within microfabricated devices, offering biomimetic tissue culture conditions that enhance the in vitro simulation of intact or precise human body structure and function. This capability improves the predictability of clinical trial outcomes while reducing time and cost. In this review, we focus on recent advancements in MPS used to study age-related and geriatric diseases, with particular emphasis on the application of organoids and organ-on-a-chip technologies in understanding cardiovascular diseases, cerebrovascular diseases, neurodegenerative diseases, fibrotic diseases, locomotor and sensory degenerative disorders, and rare diseases. And we aim to provide readers with critical guidelines and an overview of examples for modeling age-related and geriatric diseases using MPS, exploring mechanisms, treatments, drug screening, and other subsequent applications, from a physiopathological perspective, emphasizing the characteristic of age-related and geriatric diseases and their established correlations with the aging process. We also discuss the limitations of current models and propose future directions for MPS in aging research, highlighting the potential of interdisciplinary approaches to address unresolved challenges in the field.
Collapse
Affiliation(s)
- Chao Song
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jiachen Yang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
30
|
Li X, Li Y, Jin Z, Chen Z, Yang X, He L. Association between dietary oxidative balance scores and myocardial infarction in diabetic patients: insights from NHANES 1999-2018. BMC Public Health 2025; 25:1613. [PMID: 40312300 PMCID: PMC12044811 DOI: 10.1186/s12889-025-22742-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Myocardial infarction (MI) poses a serious health threat to diabetic patients, who are particularly vulnerable due to heightened oxidative stress. The dietary oxidative balance score (DOBS) quantifies the overall oxidative profile of the diet and may reflect diet-related cardiovascular risk. OBJECTIVE This study aimed to evaluate the association between DOBS and the risk of MI among diabetic individuals using a nationally representative U.S. POPULATION METHODS We analyzed data from 5,002 diabetic participants in the NHANES 1999-2018 cycles. DOBS was calculated based on 16 pro- and antioxidant nutrients using two 24-hour dietary recalls. Logistic regression models and 1:1 propensity score matching (PSM) were employed to assess the association between DOBS and self-reported history of MI, adjusting for demographic, clinical, and lifestyle covariates. Restricted cubic spline (RCS) models were used to evaluate potential nonlinear relationships. RESULTS A one-point increase in DOBS was associated with a 3% lower odds of MI in both unadjusted and fully adjusted models (adjusted OR = 0.97, 95% CI: 0.95-0.99). Participants in the highest DOBS tertile had a 38% lower odds of MI compared to the lowest tertile (OR = 0.62, 95% CI: 0.43-0.87), and this association remained consistent in the matched cohort (OR = 0.72, 95% CI: 0.48-0.88). While formal tests for nonlinearity were not significant, RCS curves suggested a threshold effect with diminishing benefits at higher DOBS levels. Subgroup and sensitivity analyses confirmed the robustness of the findings. CONCLUSION Higher DOBS is associated with a lower likelihood of MI among diabetic patients. These findings highlight the potential value of antioxidant-rich dietary patterns in cardiovascular risk assessment. However, given geographic and cultural variability in diet, further validation is needed in diverse populations and prospective study settings.
Collapse
Affiliation(s)
- Xu Li
- TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Yashi Li
- TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Zehao Jin
- TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Zitong Chen
- Chinese Institutes for Medical Research (CIMR), Capital Medical University, Beijing, China
| | - Xinyi Yang
- Department of Gastroenterology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Lan He
- Department of Ultrasound, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, Shanghai, China.
| |
Collapse
|
31
|
Liu X, Zhao H, Liu S, Wen S, Fan W, Xie Q, Cui B, Zhou L, Peng J, Pan H, Zheng Z, Zhang Q. Comparison of the effects of metformin and empagliflozin on cardiac function in heart failure with preserved ejection fraction mice. Front Cardiovasc Med 2025; 12:1533820. [PMID: 40364820 PMCID: PMC12069290 DOI: 10.3389/fcvm.2025.1533820] [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: 11/25/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose Recent evidence suggests that empagliflozin (EMPA) and metformin (MET) may improve prognosis in heart failure with preserved ejection fraction (HFpEF) patients. This study aims to compare their effects on cardiac structure and function in HFpEF. Methods Male C57BL/6J mice were fed a high-fat diet with L-NAME for 8 weeks to induce HFpEF, followed by 4 weeks of MET or EMPA treatment. Cardiac structure and function were assessed. Network pharmacology and bioinformatics identified key targets, validated by RT-qPCR and WB. Results EMPA-treated mice lost weight, unlike MET-treated ones. MET reduced systolic blood pressure significantly. Both treatments improved glucose tolerance; MET enhanced insulin sensitivity. EMPA increased exercise tolerance by extending exhaustion distance. Both treatments improved diastolic function, reduced heart weight, and attenuated myocardial fibrosis and hypertrophy. Plasma NT-proBNP levels were slightly elevated but not significant. EMPA downregulated HSP90 mRNA and protein expression; both drugs downregulated TGFβ. Conclusion MET and EMPA improve cardiac fibrosis, diastolic function, and pulmonary congestion in HFpEF mice. MET acts by downregulating TGFβ, while EMPA affects collagen metabolism and downregulates HSP90 and TGFβ. These findings offer insights into HFpEF treatment.
Collapse
Affiliation(s)
- Xiehong Liu
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Department of Emergency, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Huiqi Zhao
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Sisi Liu
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Siao Wen
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Wenjuan Fan
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Qiong Xie
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Bo Cui
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Lin Zhou
- College of Clinical Laboratory, Changsha Medical University, Changsha, Hunan, China
| | - Jianqiang Peng
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Hongwei Pan
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Zhaofen Zheng
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| | - Qinghai Zhang
- Department of Cardiology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Clinical Medicine Research Center of Heart Failure of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
32
|
Chen M, Zhao N, Shi W, Xing Y, Liu S, Meng X, Li L, Zhang H, Meng Y, Xie S, Deng W. Glucose-dependent insulinotropic polypeptide/glucagon-like peptide 1 receptor agonist tirzepatide promotes branched chain amino acid catabolism to prevent myocardial infarction in non-diabetic mice. Cardiovasc Res 2025; 121:454-467. [PMID: 39928435 DOI: 10.1093/cvr/cvaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/18/2024] [Accepted: 11/03/2024] [Indexed: 02/12/2025] Open
Abstract
AIMS A novel dual glucose-dependent insulinotropic polypeptide and glucagon-like peptide 1 receptor agonist, tirzepatide (LY3298176, TZP), has been developed to treat Type 2 diabetes mellitus (T2DM). In ischaemic heart diseases, TZP is involved in cardiac metabolic processes. However, its efficacy and safety in treating heart failure (HF) following myocardial infarction (MI) remain uncertain. METHODS AND RESULTS Herein, 12 week C57BL/6J mice were subjected to MI surgery, followed by administration of TZP. The effects of TZP on cardiac function and metabolism were thoroughly assessed by physiological, histological, and cellular analyses. Downstream effectors of TZP were screened through untargeted metabolomics analysis and molecular docking. Construct a lower branched chain amino acid (BCAA) diet model to determine whether TZP's cardioprotective effect is associated with reducing BCAA levels. Our results demonstrated that TZP reduced mortality following MI, decreased the infarct area, and attenuated cardiomyocyte necrosis. Pathological evaluation of cardiac tissues demonstrated increased fibrosis repair and decreased inflammatory infiltration. Mechanistically, untargeted metabolomics analysis uncovered a positive correlation between TZP and the BCAA catabolism pathway. The molecular docking verified that TZP could bind with branched-chain keto acid dehydrogenase E1 subunit α (BCKDHA). TZP reduced BCKDHA phosphorylation at S293, enhanced BCAA catabolism, and inhibited the activation of metabolism by activating rapamycin (mTOR) signalling pathway. Furthermore, mice fed a low-BCAA diet post-MI demonstrated reduced cardiomyocyte necrosis, increased fibrosis repair, and decreased inflammatory infiltration. These cardioprotective effects were further enhanced when used synergistically with TZP. CONCLUSION Taken together, our findings provide new perspectives on the unrecognized role of TZP in cardiac protection. TZP enhanced BCAA catabolism and attenuated BCAA/mTOR signalling pathway in MI mice. Consequently, this study may present novel therapeutic options for patients with HF.
Collapse
Affiliation(s)
- Mengya Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Wenke Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Yun Xing
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Shiqiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Xianxian Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Yanyan Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| |
Collapse
|
33
|
Feng Y, Mao T, Yi J, Zhang N, Gu Y, Shen H, Chen J. Runt-related transcription factors: from pathogenesis to therapeutic targets in multiple-organ fibrosis. Front Cell Dev Biol 2025; 13:1528645. [PMID: 40356603 PMCID: PMC12066561 DOI: 10.3389/fcell.2025.1528645] [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: 11/15/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Fibrosis is a partially manageable process that leads to scarring and tissue hardening by prompting myofibroblasts to deposit significant amounts of extracellular matrix (ECM) following injury. It results in detrimental consequences and pathological characteristics, which hinder the functioning of associated organs and increase mortality rates. Runt-related transcription factors (RUNX) are part of a highly conserved family of heterodimer transcription factors, comprising RUNX1, RUNX2, and RUNX3. They are involved in several biological processes and undergo various forms of post-translational modification. RUNX regulates multiple targets and pathways to impact fibrosis, indicating promise for clinical application. Therefore, its significance in the fibrosis process should not be disregarded. The review begins with an objective description of the structure, transcriptional mechanism, and biological function of RUNX1, RUNX2, and RUNX3. A subsequent analysis is made of their physiological relationship with heart, lung, kidney, and liver, followed by a focus on the signaling mechanism of RUNX in regulating fibrosis of these organs. Furthermore, potential agents or drugs targeting RUNX for treating organ fibrosis are summarized, along with an evaluation of the therapeutic prospects and potential value of RUNX in fibrosis. Further research into RUNX could contribute to the development of novel therapeutic approaches for fibrosis.
Collapse
Affiliation(s)
- Yuan Feng
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Tianshi Mao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jifei Yi
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Na Zhang
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Yinying Gu
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Huifen Shen
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Jie Chen
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
34
|
Zhang X, Fang Y, Qin X, Zhang Y, Kang B, Zhong L, Liu B, Luo J, Wei Y. The Role of MCPIP1 in Macrophage Polarization and Cardiac Function Post-Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500747. [PMID: 40285621 DOI: 10.1002/advs.202500747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Macrophages play a critical role in both initiating and resolving inflammation following MI (myocardial infarction). Their polarization is essential for maintaining cardiac function. This study aims to explore the role of MCPIP1(Monocyte chemotactic protein-induced protein 1) in regulating macrophage polarization and its impact on heart-spleen interactions during MI recovery. The role of MCPIP1 was investigated using histological staining, RNA sequencing of bone marrow-derived macrophages, co-culture experiments, and validated by western blot. Compared to controls, myeloid MCPIP1-deficient mice had lower survival rates, larger infarction areas, and more severe inflammatory responses after MI. This was due to increased M1 polarization and impaired conversion to the M2 phenotype. Ferroptosis activation in MCPIP1-deficient macrophages was inhibited by Fer-1 and PFT-α, which promoted M2 polarization and fibroblast activation into myofibroblasts. MCPIP1-deficient MI mice also showed splenomegaly and elevated levels of circulating macrophages, indicating excessive extramedullary hematopoiesis. Splenectomy improved survival rates and reduced infarction size in MCPIP1-deficient mice. MCPIP1 suppresses the P53/ferroptosis pathway to regulate macrophage polarization and TGF-β/SMAD3-mediated fibroblast activation. Its deficiency exacerbates inflammation through abnormal splenic macrophage output, impairing cardiac repair. MCPIP1 is a promising therapeutic target for modulating ferroptosis and heart-spleen communication to protect cardiac function following MI.
Collapse
Affiliation(s)
- Xingxu Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Yuan Fang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xiaoming Qin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Yiwei Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Bo Kang
- Department of Health Policy and Management, Fielding School of Public Health, University of California, 650 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Li Zhong
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, United States
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| |
Collapse
|
35
|
Russell-Hallinan A, Tonry C, Kerrigan L, Edgar K, Collier P, McDonald K, Ledwidge M, Grieve D, Karuna N, Watson C. Proteome Alterations in Cardiac Fibroblasts: Insights from Experimental Myocardial Infarction and Clinical Ischaemic Cardiomyopathy. Int J Mol Sci 2025; 26:3846. [PMID: 40332511 PMCID: PMC12028142 DOI: 10.3390/ijms26083846] [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: 03/04/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Ischaemic heart disease (IHD) is a chronic condition that can cause pathological cardiac remodelling and heart failure (HF). In this study, we sought to determine how cardiac fibroblasts were altered post-experimental myocardial infarction (MI). Female C57BL6 mice underwent experimental MI by permanent left coronary artery ligation. Cardiac fibroblasts were isolated from extracted heart tissue of experimental MI mice and subsequently treated with the pro-fibrotic cytokine, TGF-β, for 24 h and analysed using high throughput LC-MS/MS analysis. Findings were validated using mass spectrometry data generated from human left ventricular tissue analysis, which were collected from patients with ischaemic cardiomyopathy (ISCM) and age/sex-matched patients without clinical HF (NF). Proteomic analysis revealed significant protein expression changes in mouse cardiac fibroblasts after MI. These changes were most pronounced at 1 month post-MI, compared to earlier time points (3 days and 1 week). TGF-β treatment profoundly affected fibroblast cells extracted from MI mice, indicating a heightened sensitivity to pro-fibrotic factors after myocardial injury. Extracellular matrix (ECM) proteins significantly altered in MI fibroblasts following TGF-β treatment were significantly associated with cardiac remodelling. Notably, Lox was significantly changed in both isolated fibroblasts treated with TGF-β from experiment MI mice and human ISCM. Isolated cardiac fibroblasts from MI mice are more susceptible to developing pathogenic traits following TGF-β treatment than isolated fibroblasts from normal heart tissue. ECM proteins associated with these enhanced fibroblast activities and functions are evident. These altered proteins may play a functional role in MI-associated cardiac dysfunction.
Collapse
Affiliation(s)
- Adam Russell-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
| | - Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
| | - Lauren Kerrigan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
| | - Kevin Edgar
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
| | - Patrick Collier
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ken McDonald
- STOP-HF Unit, St Vincent’s University Hospital Healthcare Group, D04 T6F4 Dublin, Ireland; (K.M.); (M.L.)
- UCD Conway Institute, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Mark Ledwidge
- STOP-HF Unit, St Vincent’s University Hospital Healthcare Group, D04 T6F4 Dublin, Ireland; (K.M.); (M.L.)
- UCD Conway Institute, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - David Grieve
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
| | - Narainrit Karuna
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chris Watson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.R.-H.); (C.T.); (L.K.); (K.E.); (D.G.)
- UCD Conway Institute, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
36
|
Wang S, Pei G, Shen J, Fang Z, Chen T, Wang L, Cheng H, Li H, Pei H, Feng Q, Fan Q, He C, Fu C, Guo Y, Wei Q. Pulsed electromagnetic fields treatment ameliorates cardiac function after myocardial infarction in mice and pigs. J Adv Res 2025:S2090-1232(25)00263-2. [PMID: 40250556 DOI: 10.1016/j.jare.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/21/2024] [Accepted: 04/15/2025] [Indexed: 04/20/2025] Open
Abstract
INTRODUCTION Ischemic heart disease (IHD) is a prominent contributor to mortality worldwide, with myocardial infarction (MI) representing its most severe manifestation. Pulsed electromagnetic fields (PEMF) treatment shows promise for treating IHD. Nevertheless, the therapeutic impact and underlying mechanism of PEMF in MI are not fully understood. OBJECTIVES To investigate the efficacy, safety, and mechanisms of PEMF for MI. METHODS We established MI models in both mice and pigs and performed serial echocardiography and cardiac magnetic resonance follow-up to demonstrate the benefit of PEMF treatment after MI. The pathological environment after myocardial infarction was simulated in vitro to observe changes in various cells exposed to PEMF. Gene knockout (TLR4-/-) mice and inhibitors were used to compare the differences in the efficacy of PEMF treatment relative to that of gene knockout/inhibitor treatments. Agonists were used to further explore the mechanism of PEMF treatment. RESULTS In post-MI mice, PEMF treatment enhanced cardiac function and reduced scar formation. PEMF reduced the macrophage inflammatory response, improved cardiomyocyte survival in an inflammatory environment, and decreased collagen secretion by fibroblasts in vitro. Importantly, in the clinically relevant porcine model, PEMF treatment inhibited the inflammatory response and alleviated adverse left ventricular remodeling. Moreover, PEMF could exert therapeutic effects similar to those of gene knockout or inhibitor treatments. In the presence of TLR4 knockout or pyrrolidine dithiocarbamate (an NF-κB inhibitor) administration, PEMF could still improve cardiac function in post-MI mice. Mechanistically, the anti-inflammatory effect of PEMF was reversed when RS09 (a TLR4 agonist) was administered, and the antifibrotic effect of PEMF was attenuated after treatment with SRI-011381 (a TGF-β signaling pathway agonist). CONCLUSIONS PEMF treatment exhibits considerable promise as a noninvasive physical therapy modality, warranting further investigation into its potential implications for managing patients with IHD.
Collapse
Affiliation(s)
- Shiqi Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gaiqin Pei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiayu Shen
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhi Fang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tingyu Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongxin Cheng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanbin Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongliang Pei
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Qipu Feng
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingwen Fan
- School of Mechanical Engineering, Sichuan University, Chengdu 610041, China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenying Fu
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
37
|
Chen S, Zeng X, Wu M, Zhu J, Wu Y. Sodium Alginate Hydrogel Infusion of Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles and p38α Antagonistic Peptides in Myocardial Infarction Fibrosis Mitigation. J Am Heart Assoc 2025; 14:e036887. [PMID: 40178108 DOI: 10.1161/jaha.124.036887] [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: 07/03/2024] [Accepted: 11/27/2024] [Indexed: 04/05/2025]
Abstract
BACKGROUND Myocardial fibrosis is a pathological hallmark of heart failure post infarction, emphasizing the need for innovative treatment strategies. This research assesses the antifibrotic potential of a sodium alginate (SA) hydrogel loaded with extracellular vesicles (EVs) from bone marrow mesenchymal stem cells and PAP (p38α antagonistic peptides), aiming to interfere with fibrosis-inducing pathways in myocardial tissue after infarction. METHODS We induced fibrosis in mouse cardiac fibroblasts through hypoxia and disrupted the Mapk14 gene to study its contribution to fibrosis. Mesenchymal stem cell-derived EVs, loaded with PAP, were encapsulated in the SA hydrogel (EVs-PAP@SA). The formulation was tested in vitro for its effect on fibrotic marker expression and cell behavior, and in vivo in a murine model of myocardial infarction for its therapeutic efficacy. RESULTS Map k14 silencing showed a decrease in the fibrotic response of cardiac fibroblasts. Treatment with the EVs-PAP@SA hydrogel notably reduced profibrotic signaling, increased cell proliferation and migration, and lowered apoptosis rates. The in vivo treatment with the hydrogel post myocardial infarction significantly diminished myocardial fibrosis and improved cardiac performance. CONCLUSIONS The study endorses the SA hydrogel as an effective vehicle for delivering mesenchymal stem cell-derived EVs and PAP to the heart post myocardial infarction, providing a novel approach for modulating myocardial fibrosis and promoting cardiac healing.
Collapse
Affiliation(s)
- Siyao Chen
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Xiaodong Zeng
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Meifeng Wu
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Jiade Zhu
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Yijin Wu
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| |
Collapse
|
38
|
Liu Z, Chen C, Zhang Y, Ji F, Liu H, Du H, Guo Y, Dong X, Yang Z, Han M, Tang C, Yang K, Zhang J, Zhao K, Chen Y, Jiang X, Xu F. Legumain In Situ Engineering Promotes Efferocytosis of CAR Macrophage to Treat Cardiac Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2417831. [PMID: 40223483 DOI: 10.1002/adma.202417831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/13/2025] [Indexed: 04/15/2025]
Abstract
Uncontrolled and excessive cardiac fibrosis after myocardial infarction (MI) is a primary contributor to mortality by heart failure. Chimeric antigen receptor macrophage (CAR-MΦ) therapy shows great promise in cardiac fibrosis, however, the overwhelming apoptotic cells after MI results in an overburdened efferocytosis in CAR-MΦ, which compromises their antifibrotic potency. This work here reports an in situ engineered legumain (Lgmn) to elevate the cargo degradation of phagolysosome for promoting the efferocytosis of CAR-MΦs, restoring their antifibrotic capability. Specifically, with the in-house customized macrophages-targeting lipid nanoparticles, this work first creates an efferocytosis-boosted fibrosis-specific CAR-MΦs by introducing dual mRNAs that encode Lgmn, an endolysosomal cysteine protease, along with an anti-fibroblast activation protein (FAP) CAR, respectively. This data demonstrate these CAR-MΦs displayed a significantly increased phagocytic capacity as well as improved efferocytosis and enhanced antifibrotic capability. Treatment with the in situ reprogrammed CAR-MΦs in MI mice obviously reduced the infarct size and mitigated cardiac fibrosis, leading to significant restoration of cardiac function. In sum, these findings establish that promoting efferocytosis through Lgmn engineering effectively relieved the overburdened efferocytosis of CAR-MΦs, and enhanced their treatment efficacy of cardiac fibrosis with broad application in other fibrotic diseases.
Collapse
Affiliation(s)
- Zejuan Liu
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Chen Chen
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Fengping Ji
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Hehui Liu
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Han Du
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yunyun Guo
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Xianghui Dong
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Zhenmei Yang
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Maosen Han
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Chunwei Tang
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Kehui Yang
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Jian Zhang
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Kun Zhao
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Yuguo Chen
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Xinyi Jiang
- Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key laboratory for technology Research and evaluation of drug Products and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 cultural West Road, Jinan, Shandong Province, 250012, China
| | - Feng Xu
- Department of Emergency Medicine, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| |
Collapse
|
39
|
Diao MN, Lv YJ, Xin H, Zhang YF, Zhang R. A comprehensive review of m6 A methylation in coronary heart disease. J Mol Med (Berl) 2025:10.1007/s00109-025-02540-1. [PMID: 40208302 DOI: 10.1007/s00109-025-02540-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
The morbidity and mortality rates of coronary heart disease (CHD) are high worldwide. The primary pathological changes in CHD involve stenosis and ischemia caused by coronary atherosclerosis (AS). Extensive research on the pathogenesis of AS has revealed chronic immunoinflammatory processes and cell proliferation in all layers of coronary vessels, including endothelial cells (ECs), vascular smooth muscle cells, and macrophages. m6 A methylation is a common posttranscriptional modification of RNA that is coordinated by a variety of regulators (writers, readers, erasers) to maintain the functional stability of modified mRNAs and ncRNAs. In recent years, there has been increasing focus on the involvement of m6 A methylation in the incidence and progression of CHD, which starts with atherosclerotic plaque formation, leads to myocardial ischemia, and ultimately results in the occurrence of myocardial infarction (MI). m6 A regulators modulate relevant signaling pathways to participate in the inflammatory response, programmed death of cardiomyocytes, and fibrosis. Therefore, diagnostic models based on m6 A profiling are helpful for the early detection of CHD, and m6 A methylation shows promise as a sensitive target for new drugs to treat CHD in the future.
Collapse
Affiliation(s)
- Mei-Ning Diao
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Yi-Jv Lv
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China.
| | - Rui Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China.
| |
Collapse
|
40
|
Qiao J, Xu X, Zhou X, Wu Y, Wang J, Xi H, Liu C, Wang Y, Zhou L, Zhou X, Jiang H, Wu J, Deng H, Yu L. Targeted Ganglion Delivery of CaV2.2-Mediated Peptide by DNA Nanoflowers for Relieving Myocardial Infarction and Neuropathic Pain. ACS NANO 2025; 19:13037-13052. [PMID: 40128122 DOI: 10.1021/acsnano.4c17325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
N-type calcium channel (CaV2.2) protein contributes to neuronal excitability and overactivation in sympathetic ganglion (SG) following myocardial infarction (MI), thereby easily triggering cardiac remodeling and ventricular arrhythmias (VAs). Despite much advances in the understanding of CaV2.2, a neuron-targeted modifying treatment is, yet, infrequently realized. Moreover, establishing a specific delivery strategy and stable probe architecture with an extensive molecular structure in pursuit of the complex CaV2.2 regulation still remains a challenge. Herein, we develop a smart DNA nanoflower (sDNF) composite by utilizing the customizable design and scalable production from a multifunctionality-encoded template that self-assembles into a biomimetic nanoarchitecture. The nanoarchitecture contains a neuron-targeting aptamer and a decorated CaV2.2 mediator peptide-DNA bioconjugate. The combined targeted delivery and the release of the CaV2.2 mediator peptide synergistically led to an ∼31% reduction of the peak calcium current in neuron cells. Moreover, sDNF alleviated MI-induced SG hyperactivity and improved in vivo outcomes, such as decreasing susceptibility to VAs and relieving neuropathic pain for 10 h. The infarct size treated with sDNF is reduced to approximately 11.1%. It is envisioned that the DNF-based nanostructure for cardiac remodeling suppression and VAs inhibition along with pain relief provides a potential approach for the clinical treatment of sympathetic-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Jiaming Qiao
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Xiao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Yang Wu
- Hubei Key Laboratory of Biomass Resource Chemistry. and Environmental Biotechnology, Hubei Engineering Center of Natural Polymers-Based Medical Materials, School of Resource and Environmental Science, Wuhan University, Wuhan 430079, China
| | - Jiale Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Haosong Xi
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Chengzhe Liu
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Yuhong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Xue Zhou
- Key Laboratory of Environment and Health, Ministry of Education, Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Jun Wu
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| | - Hongbing Deng
- Hubei Key Laboratory of Biomass Resource Chemistry. and Environmental Biotechnology, Hubei Engineering Center of Natural Polymers-Based Medical Materials, School of Resource and Environmental Science, Wuhan University, Wuhan 430079, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan 430060, P. R. China
| |
Collapse
|
41
|
Moon KZ, Rahman MH, Alam MJ, Hossain MA, Hwang S, Kang S, Moon S, Park MN, Ahn CH, Kim B. Unraveling the interplay between cardiovascular diseases and alcohol use disorder: A bioinformatics and network-based exploration of shared molecular pathways and key biomarkers validation via western blot analysis. Comput Biol Chem 2025; 115:108338. [PMID: 39778286 DOI: 10.1016/j.compbiolchem.2024.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/02/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Clinical observations indicate a pronounced exacerbation of Cardiovascular Diseases (CVDs) in individuals grappling with Alcohol Use Disorder (AUD), suggesting an intricate interplay between these maladies. Pinpointing shared risk factors for both conditions has proven elusive. To address this, we pioneered a sophisticated bioinformatics framework and network-based strategy to unearth genes exhibiting aberrant expression patterns in both AUD and CVDs. In heart tissue samples from patients battling both AUD and CVDs, our study identified 76 Differentially Expressed Genes (DEGs) further used for retrieving important Gene Ontology (GO) keywords and metabolic pathways, highlighting mechanisms like proinflammatory cascades, T-cell cytotoxicity, antigen processing and presentation. By using Protein-Protein Interaction (PPI) analysis, we were able to identify key hub proteins that have a significant impact on the pathophysiology of these illnesses. Several hub proteins were identified include PTGS2, VCAM1, CCL2, CXCL8, IL7R, among these only CDH1 was covered in 10 algorithms of cytoHubba plugin. Furthermore, we pinpointed several Transcription Factors (TFs), including SOD2, CXCL8, THBS2, GREM1, CCL2, and PTGS2, alongside potential microRNAs (miRNAs) such as hsa-mir-203a-3p, hsa-mir-23a-3p, hsa-mir-98-5p, and hsa-mir-7-5p, which exert critical regulatory control over gene expression… In vitro study investigates the effect of alcohol on E-cadherin (CDH1) expression in HepG2 and Hep3B cells, showing a significant decrease in expression following ethanol treatment. These findings suggest that alcohol exposure may disrupt cell adhesion, potentially contributing to cellular changes associated with cardiovascular diseases. Our innovative approach has unveiled distinctive biomarkers delineating the dynamic interplay between AUD and various cardiovascular conditions for future therapeutic exploration.
Collapse
Affiliation(s)
- Kamelia Zaman Moon
- Department of Computer Science and Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh.
| | - Md Jahangir Alam
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh
| | - Md Arju Hossain
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Sungho Hwang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chi-Hoon Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
42
|
Feng L, Sun R, Zhang H, Zhang J, Peng Z, Li J, Gao Y, Xu Y, Cui J, Liu J, Yan J, Guo L, Yang L, Shen Y, Qi Z. Exploring the protective mechanisms of syringaresinol against myocardial infarction by experimental validation and network pharmacology. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167728. [PMID: 39985987 DOI: 10.1016/j.bbadis.2025.167728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Myocardial Infarction (MI) is a leading cause of mortality worldwide. Currently, effective treatments are still lacking. Increasing evidence supports the benefits of Syringaresinol (SYR) for the treatment of cardiovascular disease is accumulating. Nevertheless, whether SYR can alleviate MI is unknown. The study aims to investigate the protective effect of SYR against MI and elucidate its potential molecular mechanism. We found that SYR ameliorate MI-induced cardiac dysfunction, reduce infarct size, and alleviate myocardial hypertrophy, fibrosis, inflammation, as well as apoptosis. In addition, we collected targets related to SYR and MI through multiple databases, and obtained 281 potential therapeutic targets after intersection. GO and KEGG enrichment analysis found that these therapeutic targets were concentrated on inflammation, fibrosis, and apoptosis pathways. Based on the PPI network and combined with Centiscape2.2 and cytoHubba analysis, we obtained 10 hub proteins. The molecular docking results showed that SYR has strong bindings with 10 hub proteins. snRNA-seq data showed that CASP3 and NFKB1 were expressed in all cell types. In addition, the therapeutic targets of SYR are also mainly distributed in all cell types. Finally, we found that SYR could alleviate MI by partially reversing the expression of AKT1, EGFR, CASP3, SRC, NFKB1, HSP90AA1, HIF1A, MMP9 and ESR1 both in vivo and in vitro. Our findings suggested that SYR may protect against MI by reducing inflammatory, fibrotic and apoptotic effects via multiple targets and pathways, which provides a new theoretical foundation for the clinical therapy of MI.
Collapse
Affiliation(s)
- Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Runjia Sun
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Hanmo Zhang
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Junwei Zhang
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Zeyan Peng
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jianlin Cui
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China
| | - Lihong Guo
- Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China.
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China.
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin 300203, China.
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Beichen Hospital, Nankai University, Tianjin 300071, China; Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300122, China; The First Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University, Shihezi 832003, China.
| |
Collapse
|
43
|
Zhang X, Wang X, Wang Y, Ma X, Geng Y, Zang S, Ban Z, Jia Y, Gao Y. Norisoboldine Alleviates Isoproterenol-Induced Myocardial Ischemic Injury via the TLR4-MyD88-Dependent NF-κB Activation Pathway and Modulation of L-Type Calcium Channels. Clin Exp Pharmacol Physiol 2025; 52:e70033. [PMID: 40011076 DOI: 10.1111/1440-1681.70033] [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: 10/13/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Norisoboldine (NIB) displays beneficial effects on cardiovascular diseases, although its protective role and underlying mechanisms in myocardial ischemia (MI) injury remain elusive. The aim of this study is to explore the potential cardioprotective mechanism of NIB on MI injury caused by isoproterenol (ISO). We administered NIB to SD rats at 20 and 40 mg/kg daily for 7 days in this study; this was followed by an ISO injection to induce MI injury. Parameters such as electrocardiogram readings, heart rate, serum concentrations of creatine kinase (CK) and creatine kinase-MB (CK-MB), levels of inflammatory markers, some histopathological assessments and oxidative stress markers were evaluated. We conducted Western blot analyses to evaluate protein expression related to apoptosis and the TLR4-MyD88-mediated NF-κB activation pathway. The L-type Ca2+ current (ICa-L) and contraction of isolated ventricular cells from rats were identified using patch-clamp methods and the IonOptix detection system. The treatment with NIB resulted in improvements in heart rate and ST-segment changes, a reduction in CK and CK-MB levels, the restoration of superoxide dismutase, catalase and glutathione levels and a decrease in malondialdehyde accumulation. Furthermore, NIB reduced the expression of inflammatory markers, lowered Ca2+ levels and reactive oxygen species production and improved myocardial tissue morphology. It also countered ISO-induced alterations in apoptosis and the TLR4-MyD88-dependent NF-κB activation pathway. Additionally, NIB considerably attenuated ICa-L and reduced the contractile function of cardiomyocytes. These results suggest that NIB effectively mitigates ISO-induced MI injury through anti-inflammatory, antioxidative, and anti-apoptotic mechanisms, potentially involving the TLR4-MyD88-dependent NF-κB activation pathway and calcium balance.
Collapse
Affiliation(s)
- Xin Zhang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xinliu Wang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuanyuan Wang
- College of Basic Medicine Sciences, Hebei University of Chinese Medicine, Shijiazhuang, China
| | | | - Yunyun Geng
- College of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shuxian Zang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Ziyun Ban
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yugai Jia
- College of Basic Medicine Sciences, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, China
| | - Yonggang Gao
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
44
|
Li B, Yang WW, Yao BC, Chen QL, Zhao LL, Song YQ, Jiang N, Guo ZG. Liriodendrin alleviates myocardial ischemia‑reperfusion injury via partially attenuating apoptosis, inflammation and mitochondria damage in rats. Int J Mol Med 2025; 55:65. [PMID: 39981888 PMCID: PMC11875722 DOI: 10.3892/ijmm.2025.5506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 02/03/2025] [Indexed: 02/22/2025] Open
Abstract
Myocardial ischemia‑reperfusion (I/R) injury may lead to dysfunction of signaling pathways related to cell apoptosis, inflammation, oxidative stress, and mitochondrial damage. The present study investigated the defensive effect of liriodendrin, as a natural product isolated from Linaria vulgaris, on reperfusion injury in rats and the underlying mechanisms involved in this process. An in vivo rat model of I/R constructed by ligation of the left anterior descending artery, as well as an in vitro model using H9C2 cells under hypoxic conditions, was established to assess the cardioprotective effects of liriodendrin. The biomarkers of myocardial damage, oxidative stress, and inflammatory response were measured with enzyme‑linked immunosorbent assay (ELISA). Gene and protein expression were detected by reverse transcription‑quantitative PCR (RT‑qPCR) and western blotting. Mitochondrial morphology was observed by electron microscopy. The levels of creatine kinase isoenzymes and cardiac troponin T were significantly elevated in the I/R compared with the sham group; liriodendrin mitigated this elevation. The liriodendrin group exhibited a significant reduction in myocardial tissue apoptosis, as indicated by immunohistochemical staining and western blotting. Additionally, ELISA indicated that the I/R group had higher levels of reactive oxygen species (ROS) compared with the liriodendrin group, while the liriodendrin group had higher levels of superoxide dismutase. The in vitro experiments demonstrated that liriodendrin ameliorated hypoxia‑induced injury to mitochondria and suppressed the activation of nuclear factor-κB and B-cell lymphoma-2 associated X protein (Bax). Therefore, the present study demonstrated that liriodendrin impeded ROS‑associated metabolic disorders, maintained mitochondrial homeostasis and partially alleviated cardiomyocyte apoptosis by inhibiting the Bax signaling pathway.
Collapse
Affiliation(s)
- Bo Li
- Department of Intensive Care, Tianjin Chest Hospital, Tianjin 300001, P.R. China
| | - Wei-Wei Yang
- Department of Clinical Laboratory, Tianjin Central Hospital of Gynecology Obstetrics, Nankai University Maternity Hospital, Tianjin 300100, P.R. China
| | - Bo-Chen Yao
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300001, P.R. China
| | - Qing-Liang Chen
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300001, P.R. China
| | - Li-Li Zhao
- Tianjin Institute of Cardiovascular Diseases, Tianjin 300001, P.R. China
| | - Yan-Qiu Song
- Tianjin Institute of Cardiovascular Diseases, Tianjin 300001, P.R. China
| | - Nan Jiang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300001, P.R. China
| | - Zhi-Gang Guo
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300001, P.R. China
| |
Collapse
|
45
|
Wang L, Yu C, You T, Zhang X, Su H, Cao B, Anwaier S, Xiang H, Dai C, Long X, Han L, Zhang D, Wang J, Zhu P, Yan X, Liang J, Chen Z, Huang H, Zhu S, Sun T, Chen J, Zhu P. Injection of ROS-Responsive Hydrogel Loaded with IL-1β-targeted nanobody for ameliorating myocardial infarction. Bioact Mater 2025; 46:273-284. [PMID: 39811465 PMCID: PMC11732248 DOI: 10.1016/j.bioactmat.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The cardiac microenvironment profoundly restricts the efficacy of myocardial regeneration tactics for the treatment of myocardial infarction (MI). A prospective approach for MI therapeutics encompasses the combined strategy of scavenging reactive oxygen species (ROS) to alleviate oxidative stress injury and facilitating macrophage polarization towards the regenerative M2 phenotype. In this investigation, we fabricated a ROS-sensitive hydrogel engineered to deliver our previously engineered IL-1β-VHH for myocardial restoration. In mouse and rat models of myocardial infarction, the therapeutic gel was injected into the pericardial cavity, effectively disseminated over the heart surface, forming an in situ epicardial patch. The IL-1β-VHH released from the hydrogel exhibited penetrative potential into the myocardium. Our results imply that this infarct-targeting gel can adhere to the damaged cardiac tissue and augment the quantity of anti-IL-1β antibodies. Moreover, the anti-IL-1β hydrogel safeguards cardiomyocytes from apoptosis by neutralizing IL-1β and inducing M2-type polarization within the myocardial infarction regions, thereby facilitating therapeutic cardiac repair. Our results emphasize the effectiveness of this synergistic comprehensive treatment modality in the management of MI and showcase its considerable potential for promoting recovery in infarcted hearts.
Collapse
Affiliation(s)
- Lu Wang
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Changjiang Yu
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Ting You
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- The First Affiliated Hospital, Department of Emergency, Hengyang Medical School, University of South China, China
| | - Xinkui Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Haotao Su
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Bihui Cao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Sainiwaer Anwaier
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Hongmo Xiang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Chengming Dai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Xiang Long
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Linjiang Han
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Dengfeng Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Junwei Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Peng Zhu
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinjian Yan
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Jialiang Liang
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Zerui Chen
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Huanlei Huang
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial People’ S Hospital Ganzhou Hospital, Ganzhou, 341000, China
| | - Shuoji Zhu
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Tucheng Sun
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Jimei Chen
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Ping Zhu
- School of Medicine South China University of Technology, Guangzhou, Guangdong, 510006, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial People’ S Hospital Ganzhou Hospital, Ganzhou, 341000, China
| |
Collapse
|
46
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [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: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
47
|
Chen X, Wang Y, Huang J, Dou H, Zhang Z, Zheng Y, Long R, Zhang X, Xu F, Ye W, Xiao Q. Tamibarotene directly targets the NACHT domain of NLRP3 to alleviate acute myocardial infarction. Biochem Pharmacol 2025; 234:116801. [PMID: 39952330 DOI: 10.1016/j.bcp.2025.116801] [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: 11/08/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
The aberrant activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome has been implicated in the exacerbation of myocardial damage and the subsequent development of heart failure following myocardial infarction (MI). Inhibiting NLRP3 inflammasome activation offers a promising therapeutic strategy for mitigating MI-related injury, although no NLRP3 inhibitors have received Food and Drug administration (FDA) approval to date. To identify novel NLRP3 inflammasome inhibitors through the repurposing of FDA-approved drugs, Tamibarotene emerged as a potent inhibitor with a favorable safety profile. Mechanistically, Tamibarotene inhibits NLRP3 inflammasome activation independently of retinoic acid receptor activation, binding to Phe410 and Ile417 within the nucleotide-binding and oligomerization (NACHT) domain in an ATPase activity-dependent manner. This interaction further inhibits the assembly of the NLRP3 inflammasome. In a murine model of MI, Tamibarotene significantly reduced myocardial damage and improved cardiac function by inhibiting NLRP3 inflammasome activation. In summary, NLRP3 has been identified as a direct target of Tamibarotene for myocardial repair following MI, indicating that Tamibarotene could serve as a potential precursor for the development of innovative NLRP3 inhibitors.
Collapse
Affiliation(s)
- Xiuhui Chen
- Department of Pharmacy, the Eighth People' s Hospital of Dongguan, Dongguan Children' s Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China
| | - Yunjing Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Junjun Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Huaqian Dou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhe Zhang
- Department of Cardiovascular Medicine & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China
| | - Yutong Zheng
- Department of Pharmacy, the Eighth People' s Hospital of Dongguan, Dongguan Children' s Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China
| | - Rui Long
- Department of Neonatology, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China
| | - Xiaofeng Zhang
- Department of Pharmacy, the Eighth People' s Hospital of Dongguan, Dongguan Children' s Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China
| | - Fengdan Xu
- Department of Neonatology, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China.
| | - Weijun Ye
- Department of Pharmacy, the Eighth People' s Hospital of Dongguan, Dongguan Children' s Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China.
| | - Qing Xiao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
48
|
Zhao WJ, Qian Y, Zhang YF, Yang AH, Cao JX, Qian HY, Liu Y, Zhu WZ. Endothelial FOSL1 drives angiotensin II-induced myocardial injury via AT1R-upregulated MYH9. Acta Pharmacol Sin 2025; 46:922-939. [PMID: 39592734 PMCID: PMC11950184 DOI: 10.1038/s41401-024-01410-9] [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: 05/15/2024] [Accepted: 10/09/2024] [Indexed: 11/28/2024]
Abstract
Vascular remodeling represents a pathological basis for myocardial pathologies, including myocardial hypertrophy and myocardial infarction, which can ultimately lead to heart failure. The molecular mechanism of angiotensin II (Ang II)-induced vascular remodeling following myocardial infarction reperfusion is complex and not yet fully understood. In this study, we examined the effect of Ang II infusion on cardiac vascular remodeling in mice. Single-cell sequencing showed Ang II induced cytoskeletal pathway enrichment and that FOS like-1 (FOSL1) affected mouse cardiac endothelial dysfunction by pseudotime analysis. Myosin heavy chain 9 (MYH9) was predominantly expressed in primary cardiac endothelial cells. The Ang II type I receptor blocker telmisartan and the protein kinase C inhibitor staurosporine suppressed Ang II-induced upregulation of MYH9 and FOSL1 phosphorylation in human umbilical vein endothelial cells. Silencing MYH9 abolished Ang II-mediated inhibition of angiogenesis in human umbilical vein endothelial cells, and attenuated AngII-induced vascular hyperpermeability. We found that FOSL1 directly bound to the MYH9 promoter and thus activated transcription of MYH9 by the dual luciferase reporter and chromatin immunoprecipitation assays, leading to vascular dysfunction. In vivo, 6 weeks after injecting adeno-associated virus-ENT carrying the TEK tyrosine kinase (tie) promoter-driven short hairpin RNA for silencing FOSL1 (AAV-tie-shFOSL1), cardiac function represented by the ejection fraction and fractional shortening was improved, myocardial fibrosis was decreased, protein levels of phosphorylated FOSL1, MYH9, and collagen type I alpha were reduced, and cardiac vascular density was recovered in mice with endothelial Fosl1-specific knockdown in Ang II-infused mice. In ischemia-reperfusion mice, AAV-shFosl1 mice had a reduced infarct size and preserved cardiac function compared with control AAV mice. Our findings suggest a critical role of the FOSL1/MYH9 axis in hindering Ang II-induced vascular remodeling, and we identified FOSL1 as a potential therapeutic target in endothelial cell injuries induced by myocardial ischemia-reperfusion.
Collapse
Affiliation(s)
- Wen-Jing Zhao
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
- Cancer Research Center Nantong, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, 226006, China
| | - Yi Qian
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Yi-Feng Zhang
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Ai-Hua Yang
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Jia-Xin Cao
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Hong-Yan Qian
- Cancer Research Center Nantong, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, 226006, China
| | - Yi Liu
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Wei-Zhong Zhu
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China.
| |
Collapse
|
49
|
Chen H, Hu K, Tang Q, Wang J, Gu Q, Chen J, Hu J, Peng N, Guo M, Jiang Y, Xu Q, Xie J. CD248-targeted BBIR-T cell therapy against late-activated fibroblasts in cardiac repair after myocardial infarction. Nat Commun 2025; 16:2895. [PMID: 40148319 PMCID: PMC11950650 DOI: 10.1038/s41467-025-56703-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/24/2025] [Indexed: 03/29/2025] Open
Abstract
Excessive cardiac fibrosis is a key cause of heart failure and adverse ventricular remodeling after myocardial infarction. The abnormally activated fibroblasts after scar maturation are the chief culprit. Single-cell RNA sequencing of mouse cardiac interstitial cells after myocardial infarction depicts a late-activated fibroblast subpopulation F-Act and initially identifies its characteristic antigen CD248, which is also verified in human hearts. On this basis, we develop a CD248-targeted biotin-binding immune receptor T cell therapy against F-Act to correct cardiac repair disorders. In our study, the precise removal of F-Act after the scar matured effectively inhibits fibrotic expansion in the peri-infarct zone and improves cardiac function. This therapy provides an idea for the treatment of cardiac fibrosis and also promotes the application of engineered T cells to non-tumor diseases.
Collapse
Affiliation(s)
- Haiting Chen
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Ke Hu
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
| | - Qi Tang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, No.101 Longmian Road, Nanjing, Jiangsu, 211166, China
| | - Junzhuo Wang
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Qianyu Gu
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Jiayu Chen
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, No.158 Wuyang Road, Enshi, Hubei, 445099, China
| | - Ningxin Peng
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China
| | - Meng Guo
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Yaohui Jiang
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| | - Jun Xie
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, Anhui, 230022, China.
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, No.321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
50
|
Dabravolski SA, Kalmykov VA, Maksaeva AO, Rozhkova UV, Lapshina KO, Orekhov AN. Necroptosis in myocardial ischaemia-reperfusion injury: current update on mechanisms, therapeutic targets, and translational potential. Apoptosis 2025:10.1007/s10495-025-02108-x. [PMID: 40146485 DOI: 10.1007/s10495-025-02108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2025] [Indexed: 03/28/2025]
Abstract
Necroptosis is a programmed form of cell death that has gained significant attention in the field of cardiovascular research due to its involvement in myocardial infarction (MI) and myocardial ischaemia-reperfusion (I/R) injury. Unlike apoptosis, necroptosis elicits a pro-inflammatory response, contributing to myocardial injury, fibrosis, and adverse remodelling. This review aims to provide an overview of the molecular mechanisms underlying necroptosis, with a particular focus on its role in myocardial I/R injury. Key regulatory proteins such as Receptor-interacting protein kinase 3 (RIPK3) and Mixed lineage kinase domain-like protein (MLKL) are central to the necroptotic process, mediating cell death and inflammation. The review discusses the potential of targeting necroptosis as a therapeutic strategy for managing cardiovascular diseases, particularly post-MI. The RIPK3-CaMKII-mitochondrial permeability transition pore (mPTP) pathway is identified as a critical signalling axis in necroptosis and its inhibition may offer protective benefits in myocardial injury. The review also considers the role of natural and chemical inhibitors and other genes in necroptosis regulation. Overall, targeting necroptosis represents a promising avenue for therapeutic intervention to mitigate cardiac injury, promote recovery, and improve long-term patient outcomes in cardiovascular diseases.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, 2161002, Karmiel, Israel.
| | - Vladislav A Kalmykov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Anastasia O Maksaeva
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
- Sechenov First Moscow State Medical University, 8, Trubetskaya Street, Building 2, Moscow, Russia, 119991
| | - Ulyana V Rozhkova
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Ksenia O Lapshina
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, Moscow, Russia, 117418
| | - Alexander N Orekhov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, Moscow, Russia, 117418
| |
Collapse
|