1
|
Zhang P, Xin Y, Yuan H, Liu Z. Identification of the crucial roles of BAX high NK cells in human derived mesenchymal stem cell therapy for chronic heart failure patients. Pathol Res Pract 2025; 269:155924. [PMID: 40174277 DOI: 10.1016/j.prp.2025.155924] [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: 03/03/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
Mesenchymal stem cells (MSCs) have demonstrated significant potential in heart failure (HF) treatment, but the exact mechanisms are still not fully understood. This research utilized single-cell RNA sequencing to examine alterations in peripheral blood mononuclear cells from heart failure patients pre- and post-MSC therapy. Moreover, we utilized Mendelian randomization (MR) analysis to identify causal genes linked to HF. Specifically, through scRNA-seq, we observed a progressive increase in Natural Killer (NK) cells within peripheral blood mononuclear cells (PBMCs) following MSC treatment. Furthermore, MR analysis identified the differentially expressed gene (DEG) BAX as a potential target gene for HF. Notably, the expression of BAX was significantly downregulated after MSC treatment, suggesting its potential as a therapeutic response biomarker. Cell-cell communication analysis revealed that BAXhigh NK cells displayed reduced cell-cell communication and increased apoptotic activity. Enrichment analysis indicated an association between BAXhigh NK cells and the "coagulant" pathway. Taken together, our findings suggest that BAX may contribute to the pathogenesis of HF by promoting coagulation and apoptotic pathways. In contrast, MSCs appear to suppress BAX expression, thereby inhibiting these pathways. MSC treatment increases the proportion of NK cells and reduces BAXhigh NK cells, ultimately improving NK cell function, and ameliorating HF.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200092, China; Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200092, China; Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China; Shanghai Engineering Research Center for Stem Cell Clinical Treatment, Shanghai 200123, China
| | - Hui Yuan
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200092, China; Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China; Shanghai Engineering Research Center for Stem Cell Clinical Treatment, Shanghai 200123, China.
| |
Collapse
|
2
|
Yang S, Yan L, Chen L, Su G, Yang L, Gong L, Liu L. Cardiac PDK4 promotes neutrophilic PFKL methylation and drives the innate immune response in diabetic myocardial infarction. Pharmacol Res 2025; 215:107731. [PMID: 40222696 DOI: 10.1016/j.phrs.2025.107731] [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: 12/13/2024] [Revised: 03/20/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
NETosis plays a pivotal role in the innate immune response after diabetic myocardial infarction (MI), exerting a profound influence on the overall pathological process and potential recovery outcomes. The metabolism of diabetic cardiomyocyte actively creates a specialized micro environment for the innate immune response after MI. However, the mechanism by which cardiac metabolism drives NETosis remains unclear. Utilizing public databases of human MI sc-RNA datasets, we discovered that cardiomyocyte PDK4 expression mediates the intensification of glycolysis, which is strongly correlated with NETosis. Through mass spectrometry imaging and phenotype assessment, we ascertained that specific knockout of PDK4 in cardiomyocytes (PDK4fl/flMyh6Cre, male, 6 weeks) led to a reduction in NETosis by restraining micro environmental lactate (LA) production. In addition, the role of LA in promoting NETosis has been further corroborated by in vivo/in vitro experiments involving LA supplementation and its absence. Moreover, LA redirects neutrophil metabolic flux from glycolysis to the pentose-phosphate pathway (PPP). Mechanistically, LA triggers metabolic remodeling through the PRMT9-mediated methylation of PFKL at the R301 residue, resulting in PFKL inactivation and the consequent restriction of glycolysis. Our findings reveal the crucial role of cardiomyocyte metabolism in NETosis, shedding light on the role of LA as a vital signaling molecule in the crosstalk between cardiomyocytes and neutrophils. Importantly, we screened pitavastatin, a potential inhibitor of PDK4 among the FDA-approved drugs, and verified that it can alleviate NETosis in diabetic MI, which provides a rationale for drug selection in diabetic MI patients.
Collapse
Affiliation(s)
- Song Yang
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Longxin Yan
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Lang Chen
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Gaijuan Su
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, China
| | - Long Yang
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Lili Gong
- China-Japan Friendship Hospital, Beijing 100029, China.
| | - Lihong Liu
- China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
3
|
Xue C, Yan Z, Cheng W, Zhang D, Zhang R, Duan H, Zhang L, Ma X, Hu J, Kang J, Ma X. Curcumin ameliorates aging-induced blood-testis barrier disruption by regulating AMPK/mTOR mediated autophagy. PLoS One 2025; 20:e0321752. [PMID: 40273194 PMCID: PMC12021166 DOI: 10.1371/journal.pone.0321752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/08/2025] [Indexed: 04/26/2025] Open
Abstract
The blood-testis barrier (BTB) is composed of tight junctions (TJ) between adjacent Sertoli cells (SCs) and is crucial for sperm growth and development. Aging-induced TJ impairment is closely related to testicular dysfunction. Curcumin, a natural compound, has been widely demonstrated to have a wide range of pharmacological activities, but its regulatory effects on tight junction damage in the testis remain unclear. We here explored the effect of curcumin on TJ function and its underlying molecular mechanism by using D-galactose (D-gal)-induced mouse testis and mouse testicular SCs (TM4) aging models in vitro. In this study, D-gal increased the expression of aging-related proteins p16 and p21, whereas significantly decreased the expression of TJ proteins (ZO-1, Claudin-4, Claudin-7, and Occludin). In addition, curcumin restored the adverse effects of D-gal in the SCs. Autophagy is a degradation system for maintaining cell renewal and homeostasis. D-gal significantly decreased the autophagy level, whereas curcumin restored the effect of D-gal. Using chloroquine (CQ), an inhibitor of autophagy, and rapamycin (RAPA), an activator of autophagy, it was demonstrated that autophagy plays a key role in curcumin amelioration of TJ injury in testicular SCs. Further studies unveiled that autophagy activation was mediated through the AMPK/mTOR pathway. In conclusion, curcumin ameliorates aging-induced TJ damage through AMPK/mTOR signaling pathway-regulated autophagy. This study thus clearly identifies a novel action mechanism of curcumin in the treatment of age-related male reproductive disorders.
Collapse
Affiliation(s)
- Chen Xue
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Zhenxing Yan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Wenjing Cheng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Dong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Rong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Hongwei Duan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Lihong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Xiaofei Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Junjie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| | - Jian Kang
- School of Animal Science and technology, Guangdong polytechnic of science and trade, Guangzhou, Guangdong, China
| | - Xiaojun Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation in Gansu Province, Lanzhou, Lanzhou, Gansu, China
| |
Collapse
|
4
|
Yang SH, Zhang SN, Li XZ. Advances in Therapeutic Targets and Traditional Chinese Medicine for Cardiomyopathy. Phytother Res 2025. [PMID: 40219655 DOI: 10.1002/ptr.8494] [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: 01/05/2025] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 04/14/2025]
Abstract
Cardiomyopathy is a kind of heart disease caused by multiple factors of myocardial structure and function disorders. In this paper, we summarized and found the targets and mechanisms with therapeutic potential by querying the relevant literature on cardiomyopathy in the past 10 years from databases. Numerous pieces of literature have proven the significant efficacy of traditional Chinese medicine (TCM) in the treatment of cardiomyopathy. Through effective screening methods, we quickly identified a variety of commonly used Chinese herbs such as Astragalus, Danggui, Danshen, Pueraria Root, and ginseng, and further analyzed the active ingredients that play key roles in the treatment of cardiomyopathy. Specifically, our study revealed significant interaction activity at the molecular level of active ingredients such as calycosin, formononetin, and beta-sitosterol, which were strongly validated by sophisticated molecular docking experiments. These active ingredients can be precisely combined with 14 core targets (such as AKT1, TP53, IL6, and other key proteins), which not only reveals their potential therapeutic mechanisms but also provides direct and solid scientific support for the application of TCM in the treatment of cardiomyopathy. It is helpful to develop new TCM preparations further and provide more treatment options for patients with cardiomyopathy.
Collapse
Affiliation(s)
- Si-Hui Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, People's Republic of China
| | - Shuai-Nan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, People's Republic of China
| | - Xu-Zhao Li
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, People's Republic of China
| |
Collapse
|
5
|
Lin L, Wei J, Xue J, Fan G, Zhu W, Zhu Y, Wu R. Drp1 Promotes Macrophage M1 Polarization and Inflammatory Response in Autoimmune Myocarditis by Driving Mitochondrial Fission. J Cardiovasc Transl Res 2025; 18:237-246. [PMID: 39388091 DOI: 10.1007/s12265-024-10570-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Autoimmune myocarditis (AM) is characterized by an intricate inflammatory response within the myocardium. Dynamin-related protein 1 (Drp1), a pivotal modulator of mitochondrial fission, plays a role in the pathogenesis of various diseases. A myosin-induced experimental autoimmune myocarditis (EAM) mouse model was successfully established. Flow cytometry was employed to detect M1/M2-like macrophages. Mitochondrial fragmentation was assessed using Mito-Tracker Red CMXRos. Drp1 was upregulated and activated in EAM mice. Depletion of Drp1 was observed to mitigate inflammation, macrophage infiltration and M1 polarization within the cardiac tissue of EAM mice. In M1-like macrophages derived from the hearts of EAM mice, Drp1 was found to promote mitochondrial fission and diminish mitochondrial fusion. Furthermore, the depletion of Drp1 reduced the NF-κB-related pro-inflammatory response in EAM-associated M1-like macrophages. Drp1 drives mitochondrial fission in macrophages, driving their M1 polarization and the subsequent inflammatory response. Drp1 may represent an effective target for the prevention and treatment of AM.
Collapse
Affiliation(s)
- Lin Lin
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China.
| | - Jin Wei
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Jiahong Xue
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Gang Fan
- Second Department of Cardiology, Xianyang First People's Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Wenjing Zhu
- Department of Cardiovascular Medicine, Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Daminggong Campus, No. 5 Jianqiang Road, Xi'an, 710016, Weiyang District, China
| | - Yanhe Zhu
- Institute of Endiquidiopathies, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruiyun Wu
- Department of Internal Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| |
Collapse
|
6
|
Li S, Xiao X, Chang Y, Xu Z, Zheng X, Zhou H, Ding H, Lu W, Li T, Tao Y. Berberine Inhibits Abdominal Aortic Aneurysm Formation and Vascular Smooth Muscle Cell Phenotypic Switching by Regulating the Nrf2 Pathway. J Cell Mol Med 2025; 29:e70509. [PMID: 40193135 PMCID: PMC11974455 DOI: 10.1111/jcmm.70509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/12/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease featuring extensive membrane destruction in the vascular wall, which is closely associated with the phenotypic switching of vascular smooth muscle cells (VSMC). A thorough understanding of the changes in regulatory factors during the pathogenesis of VSMC phenotypic switching is essential for medical treatments in AAA. NRF2 was deemed to hold a pivotal position in developing AAA, especially as it can regulate VSMC phenotypic switching. In this study, we found that berberine prevents the formation of AAA by regulating the phenotypic switching of VSMC, which was well validated in both in vitro and in vivo functional experiments. Mechanically, we found that berberine regulates VSMC phenotypic switching by promoting the expression of downstream VSMC contraction genes through the deubiquitination of Keap1, in which the deubiquitinating enzyme USP15 plays an important mediating role in this process.
Collapse
MESH Headings
- Berberine/pharmacology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/prevention & control
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Animals
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Signal Transduction/drug effects
- Phenotype
- Mice
- Humans
- Male
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Sanjun Li
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Xiaoyong Xiao
- Department of Emergency MedicineThe First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's HospitalShenzhenGuangdongChina
| | - Yuechen Chang
- Experimental Center of Medical School of Shihezi UniversityShiheziChina
| | - Ziyao Xu
- Senior Department of General SurgeryThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | | | - Haiwen Zhou
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Haiqiang Ding
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Weiling Lu
- Department of CardiologyGuangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Prevention and Treatment of Coronary Heart Disease,Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease‐Associated Organ Injury and ITCWM RepairInstitute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical UniversityTianjinChina
| | - Yu Tao
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| |
Collapse
|
7
|
Xie R, Vlaski T, Sha S, Brenner H, Schöttker B. Sex-specific proteomic signatures improve cardiovascular risk prediction for the general population without cardiovascular disease or diabetes. J Adv Res 2025:S2090-1232(25)00194-8. [PMID: 40154735 DOI: 10.1016/j.jare.2025.03.034] [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: 01/11/2025] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
INTRODUCTION Accurate prediction of 10-year major adverse cardiovascular events (MACE) is critical for effective disease prevention and management. Although the SCORE2 model introduced sex-specific algorithms, opportunities remain to further refine prediction. OBJECTIVES To evaluate whether adding sex-specific proteomic profiles to the SCORE2 model enhances 10-year MACE risk prediction in the large UK Biobank (UKB) cohort. METHODS Data from 47,382 UKB participants, aged 40 to 69 years without prior cardiovascular disease or diabetes, were utilized. Proteomic profiling of plasma samples was conducted using the Olink Explore 3072 platform, measuring 2,923 unique proteins, of which 2,085 could be used. Sex-specific Least Absolute Shrinkage and Selection Operator (LASSO) regression was used for biomarker selection. Model performance was assessed by changes in Harrell's C-index (a measure of discrimination), net reclassification index (NRI), and integrated discrimination index (IDI). RESULTS During 10-year follow-up, 2,163 participants experienced MACE. Overall, 18 proteins were selected by LASSO regression, with 5 of them identified in both sexes, 7 only in males, and 6 only in females. Incorporating these proteins significantly improved the C-index of the SCORE2 model from 0.713 to 0.778 (P < 0.001) in the total population. The improvement was greater in males (C-index increase from 0.684 to 0.771; Δ = +0.087) than in females (from 0.720 to 0.769; Δ = +0.049). The WAP four-disulfide core domain protein (WFDC2) and the growth/differentiation factor 15 (GDF15) were the proteins contributing the strongest C-index increase in both sexes, even more than the N-terminal prohormone of brain natriuretic peptide (NTproBNP). CONCLUSION The derived sex-specific 10-year MACE risk prediction models, combining 12 protein concentrations among men and 11 protein concentrations among women with the SCORE2 model, significantly improved the discriminative abilities of the SCORE2 model. This study shows the potential of sex-specific proteomic profiles for enhanced cardiovascular risk stratification and personalized prevention strategies.
Collapse
Affiliation(s)
- Ruijie Xie
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69115 Heidelberg, Germany
| | - Tomislav Vlaski
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69115 Heidelberg, Germany
| | - Sha Sha
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.
| |
Collapse
|
8
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [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: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
9
|
Bai R, Yue X, Tian X, Zhao H, Liu Y, Li T, Wu J. Lysophosphatidic acid 2 alleviates deep vein thrombosis via protective endothelial barrier function. Open Med (Wars) 2025; 20:20241137. [PMID: 39927163 PMCID: PMC11806235 DOI: 10.1515/med-2024-1137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 02/11/2025] Open
Abstract
Background The specific role of lysophosphatidic acid 2 (LPA2) in deep vein thrombosis (DVT) remains unclear. Methods An inferior vena cava annulus retraction model of DVT was established in wild-type (WT) and global LPA2 knockout (Lpar2 -/- ) mice. We examined the incidence of DVT, wet weight of thrombus, length of thrombus, assessed endothelial permeability through Evans blue dye assay in vivo, cell viability, and endothelial cell (EC) permeability of mouse inferior vena cava ECs in vitro. Proteomics, histopathology, immunohistochemistry, and western blotting were employed to investigate the role of LPA2 in DVT. Results Lpar2 deficiency increased vascular endothelial permeability and promoted the progression of DVT. Histological examination revealed aggravated inflammation in the thrombus of Lpar2 -/- DVT mice. In vitro, Lpar2 -/- resulted in increased permeability of ECs. Proteomic results indicated that DVT after Lpar2 -/- may be related to tight junction (TJ) protein. LPA2 agonist, 2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl] benzoic acid, significantly reduced vascular endothelial permeability as well as increased expression of the vascular endothelial TJ protein zonula occludens-1. Conclusion These data provide a novel mechanism of endothelial barrier protection of LPA2 in DVT.
Collapse
Affiliation(s)
- Ruifeng Bai
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Xinyang Yue
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, 100035, China
| | - Xuan Tian
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Huiru Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Ying Liu
- Blood Transfusion Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin300100, China
| | - Jun Wu
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Capital Medical University, 31 Xinjiekou East St., Beijing, 100035, China
| |
Collapse
|
10
|
Xu Z, Lai Y, Zhou Y, Qin L, Hao X, Li T, Gao L, Wang X. The timing of surgical interventions following the implantation of coronary drug-eluting stents in patients undergoing gastrointestinal cancer surgery: a multicenter retrospective cohort study. Int J Surg 2025; 111:1724-1734. [PMID: 39715165 DOI: 10.1097/js9.0000000000002199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/12/2024] [Indexed: 12/25/2024]
Abstract
AIM We aim to investigate the optimal timing for surgical interventions to maximize patient benefit. BACKGROUND The guidelines recommending a minimum deferral of 6 months for non-cardiac surgeries following drug-eluting stent percutaneous coronary intervention (DES-PCI) do not adequately address the requirements for individuals undergoing gastrointestinal cancer surgery (GCS). METHODS The study encompassed 2501 patients treated from January 2017 to December 2021, all of whom underwent GCS within 1 year after DES-PCI. We conducted an analysis by comparing the occurrence of major adverse cardiovascular events (MACEs) within 30 days post-surgery at different time points. RESULTS This study enrolled a total of 2501 participants with meticulously recorded data who underwent DES-PCI and subsequently underwent GCS within 1 year post-implantation. The incidence rate of MACEs is 14.2%, including MI (5.1%), HF (5.8%), IS (3.2%), and cardiac death (0.2%), across all patients in this study. The threshold probability was determined using the Youden Index, resulting in a value of 0.320, corresponding to a "time-to-surgery value" of 87. Significant statistical differences were observed in the occurrence rates of MACEs for adjacent time intervals at 30 days ( P < 0.001), 90 days ( P < 0.009), and 180 days ( P < 0.001). CONCLUSIONS The timing of surgical intervention following DES-PCI significantly influences the occurrence of MACEs at 1, 3, and 6 months. GCS may be appropriately advanced within the 6-month timeframe, but with the exception of emergency, efforts should be made to defer them beyond the initial month.
Collapse
Affiliation(s)
- Ziyao Xu
- Senior Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yingying Lai
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Yan Zhou
- Department of Urology, Hebei Province Hospital of Chinese Medicine,Shijiazhuang, China
| | - Lipeng Qin
- Department of Neurosurgery, Hebei Province Hospital of Chinese Medicine, Shijiazhuang, China
| | - Xinyu Hao
- Department of Anesthesiology, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Lei Gao
- Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Xinxin Wang
- Senior Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Yang B, Jue XY, Luo SF, Tan ZB, Yang LN, Feng YT, Tan YZ, Liu B, Zhang JZ, Deng B, Wu WW, Zhang SW. Ilexgenin A Alleviates Myocardial Ferroptosis in Response to Ischemia Reperfusion Injury via the SIRT1 Pathway. Phytother Res 2025; 39:938-956. [PMID: 39698933 DOI: 10.1002/ptr.8414] [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: 09/07/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024]
Abstract
Myocardial ischemia-reperfusion (I/R) injury has emerged as an increasingly serious cardiovascular health concern worldwide, with ferroptosis playing a pivotal role as the underlying pathogenic process. This study aimed to investigate the pharmacological effect and mechanism of Ilexgenin A on cardiomyocyte ferroptosis induced by myocardial I/R injury. In vivo, we established a murine anterior descending artery ligation/recanalization model to evaluate the cardioprotective effect of Ilexgenin A. Bioinformatics analysis, molecular docking, and Surface Plasmon Resonance imaging were conducted to predict the pharmacological targets of Ilexgenin A. In vitro experiments, the neonatal rat cardiomyocytes (NRCMs) were utilized to further explore the mechanism of Ilexgenin A in inhibiting ferroptosis using chemiluminescence and immunofluorescence staining, electron microscopy, biochemical assay, RT-qPCR, western blotting, and so on. The results showed that Ilexgenin A protected against cardiac dysfunction, ameliorated myocardial ferroptosis and mitochondrial damage induced by murine myocardial I/R injury via the silence information regulator 1 (SIRT1) pathway, the trend was consistently observed in NRCMs. Additionally, the SIRT1 knockdown by siRNA delivery partially abrogated the beneficial effects of Ilexgenin A on ameliorating mitochondrial damage, and then aggravated erastin-induced ferroptosis in NRCMs. Overall, Our research demonstrated that the inhibition of ferroptosis via the SIRT1 pathway was one of the mechanisms by which Ilexgenin A exerted cardioprotective effect.
Collapse
Affiliation(s)
- Bo Yang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Yu Jue
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shang-Fei Luo
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhang-Bin Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li-Ning Yang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun-Ting Feng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong-Zhen Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing-Zhi Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bo Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei-Wei Wu
- Department of Rehabilitation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang-Wei Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Hu Y, Su D, Zhang Y, Fu Y, Li S, Chen X, Zhang X, Zheng S, Ma X, Hu S. Genome-wide DNA methylation and transcriptome sequencing analyses of lens tissue in an age-related mouse cataract model. PLoS One 2025; 20:e0316766. [PMID: 39883715 PMCID: PMC11781636 DOI: 10.1371/journal.pone.0316766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/15/2024] [Indexed: 02/01/2025] Open
Abstract
DNA methylation is known to be associated with cataracts. In this study, we used a mouse model and performed DNA methylation and transcriptome sequencing analyses to find epigenetic indicators for age-related cataracts (ARC). Anterior lens capsule membrane tissues from young and aged mice were analyzed by MethylRAD-seq to detect the genome-wide methylation of extracted DNA. The young and aged mice had 76,524 and 15,608 differentially methylated CCGG and CCWGG sites, respectively. The Pearson correlation analysis detected 109 and 33 differentially expressed genes (DEGs) with negative methylation at CCGG and CCWGG sites, respectively, in their promoter regions. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses showed that DEGs with abnormal methylation at CCGG sites were primarily associated with protein kinase C signaling (Akap12, Capzb), protein threonine kinase activity (Dmpk, Mapkapk3), and calcium signaling pathway (Slc25a4, Cacna1f), whereas DEGs with abnormal methylation at CCWGG sites were associated with ribosomal protein S6 kinase activity (Rps6ka3). These genes were validated by pyrosequencing methylation analysis. The results showed that the ARC group (aged mice) had lower Dmpk and Slc25a4 methylation levels and a higher Rps6ka3 methylation than the control group (young mice), which is consistent with the results of the joint analysis of differentially methylated and differentially expressed genes. In conclusion, we confirmed the genome-wide DNA methylation pattern and gene expression profile of ARC based on the mouse cataract model with aged mice. The identified methylation molecular markers have great potential for application in the future diagnosis and treatment of ARC.
Collapse
Affiliation(s)
- Yuzhu Hu
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Dongmei Su
- Department of Genetics, Health Department, National Research Institute for Family Planning, Beijing, China
- Graduate School, Peking Union Medical College, Beijing, China
| | - Yue Zhang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yanjiang Fu
- Daqing Ophthalmology Hospital, Daqing, Heilongjiang, China
| | - Sijia Li
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaoya Chen
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiao Zhang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Shunfei Zheng
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xu Ma
- Department of Genetics, Health Department, National Research Institute for Family Planning, Beijing, China
- Graduate School, Peking Union Medical College, Beijing, China
| | - Shanshan Hu
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| |
Collapse
|
13
|
Yu Z, Zhao Z, Ding C, Liu Q, Ma T, Han X, Lu D, Zhang L. The association between sarcopenia and cardiovascular disease: An investigative analysis from the NHANES. Nutr Metab Cardiovasc Dis 2025:103864. [PMID: 40016026 DOI: 10.1016/j.numecd.2025.103864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/30/2024] [Accepted: 01/09/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND AND AIMS Sarcopenia, a multifaceted chronic condition, exhibits an ambiguous association with cardiovascular disease (CVD). This study aimed to elucidate the connection between sarcopenia and CVD, as well as its implications for cardiovascular prognosis, by analyzing globally representative data from the National Health and Nutrition Examination Survey (NHANES). The findings aim to provide valuable insights for clinical practice and future research endeavors. METHODS AND RESULTS We used data from the NHANES database covering the period from 2011 to 2018. Sarcopenia was defined according to the Foundation for the National Institutes of Health criteria, while CVD was determined through self-reports. The association between sarcopenia and CVD was assessed via logistic regression analysis, with a nonlinear relationship explored using a restricted cubic spline curve. Cox proportional hazards regression was used to examine the associations between sarcopenia and both all-cause and cardiovascular mortality.Among the 7702 participants, the association between sarcopenia and CVD remained significant after adjusting for confounding factors such as age, sex, smoking status, alcohol consumption, and comorbidities (odds ratio, 1.89; 95 % confidence interval [CI], 1.04-3.43; P = 0.030). The hazard ratios for cardiovascular and all-cause mortality in this fully adjusted model were 1.95 (95 % CI, 0.62-6.12; P = 0.252) and 1.43 (95 % CI, 0.71-2.87; P = 0.319), respectively. DISCUSSION Sarcopenia is significantly associated with CVD in the general population, even after adjusting for confounding factors but is not associated with cardiovascular or all-cause mortality.
Collapse
Affiliation(s)
- Zhenze Yu
- Cardiac Department, Aerospace center hospital, Beijing, 100049, China; Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, 10049, China.
| | - Zihan Zhao
- Cardiac Department, Aerospace center hospital, Beijing, 100049, China; Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, 10049, China.
| | - Chujing Ding
- Peking University Health Science Center, Beijing, 10049, China.
| | - Qilin Liu
- China Resources Double-crane Pharmaceutical Co., Ltd, China.
| | - Teng Ma
- Cardiac Department, Aerospace center hospital, Beijing, 100049, China; Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, 10049, China.
| | - Xiongyi Han
- Cardiac Department, Aerospace center hospital, Beijing, 100049, China; Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, 10049, China.
| | - Dan Lu
- Cardiac Department, Aerospace center hospital, Beijing, 100049, China; Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, 10049, China.
| | - Lili Zhang
- Cardiac Department, Aerospace center hospital, Beijing, 100049, China; Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, 10049, China.
| |
Collapse
|
14
|
Xiong J, Chen G, Lin B, Zhong L, Jiang X, Lu H. Integrative analysis of single-Cell RNA sequencing and experimental validation in the study of abdominal aortic aneurysm progression. Gene 2024; 929:148820. [PMID: 39103059 DOI: 10.1016/j.gene.2024.148820] [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: 04/08/2024] [Revised: 07/13/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a complex vascular disorder characterized by the progressive dilation of the abdominal aorta, with a high risk of rupture and mortality. Understanding the cellular interactions and molecular mechanisms underlying AAA development is critical for identifying potential therapeutic targets. METHODS This study utilized datasets GSE197748, GSE164678 and GSE183464 from the GEO database, encompassing bulk and single-cell RNA sequencing data from AAA and control samples. We performed principal component analysis, differential expression analysis, and functional enrichment analysis to identify key pathways involved in AAA. Cell-cell interactions were investigated using CellPhoneDB, focusing on fibroblasts, vascular smooth muscle cells (VSMCs), and macrophages. We further validated our findings using a mouse model of AAA induced by porcine pancreatic enzyme infusion, followed by gene expression analysis and co-immunoprecipitation experiments. RESULTS Our analysis revealed significant alterations in gene expression profiles between AAA and control samples, with a pronounced immune response and cell adhesion pathways being implicated. Single-cell RNA sequencing data highlighted an increased proportion of pro-inflammatory macrophages, along with changes in the composition of fibroblasts and VSMCs in AAA. CellPhoneDB analysis identified critical ligand-receptor interactions, notably collagen type I alpha 1 chain (COL1A1)/COL1A2-CD18 and thrombospondin 1 (THBS1)-CD3, suggesting complex communication networks between fibroblasts and VSMCs. In vivo experiments confirmed the upregulation of these genes in AAA mice and demonstrated the functional interaction between COL1A1/COL1A2 and CD18. CONCLUSION The interaction between fibroblasts and VSMCs, mediated by specific ligand-receptor pairs such as COL1A1/COL1A2-CD18 and THBS1-CD3, plays a pivotal role in AAA pathogenesis.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Animals
- Mice
- Single-Cell Analysis/methods
- Humans
- Sequence Analysis, RNA/methods
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Macrophages/metabolism
- Disease Progression
- Fibroblasts/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Male
- Mice, Inbred C57BL
- Gene Expression Profiling/methods
- Cell Communication/genetics
- Collagen Type I/genetics
- Collagen Type I/metabolism
Collapse
Affiliation(s)
- Jie Xiong
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Guojun Chen
- Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Beiyou Lin
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Lintao Zhong
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China.
| | - Hongyun Lu
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China.
| |
Collapse
|
15
|
Lei M, Wu J, Tan Y, Shi Y, Yang W, Tu H, Tan W. β-asarone protects against age-related motor decline via activation of SKN-1/Nrf2 and subsequent induction of GST-4. Pharmacol Res 2024; 209:107450. [PMID: 39366648 DOI: 10.1016/j.phrs.2024.107450] [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: 05/25/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Decelerating motor decline is important for promoting healthy aging in the elderly population. Acorus tatarinowii Schott is a traditional Chinese medicine that contains β-asarone as a pharmacologically active constituent. We found that β-asarone can decelerate motor decline in various age groups of Caenorhabditis elegans, while concurrently prolonging their lifespan and modulating synaptic transmission. To understand the mechanisms of its efficacy in motor improvement, we investigated and discovered that mitochondrial fragmentation, a marker for aging, is delayed after β-asarone treatment. Moreover, their efficacy is blocked by dysfunctional mitochondria. Corresponding to their role in regulating mitochondrial homeostasis, we found that SKN-1/Nrf2 and GST-4 are critical in the β-asarone treatment, and they appear to be activated via the insulin/IGF-1 signaling pathway. Well-developed intestinal microvilli are required for this process. Our study demonstrates the efficacy and mechanism of β-asarone treatment in age-related motor decline, contributing to the discovery of drugs for achieving healthy aging.
Collapse
Affiliation(s)
- Ming Lei
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Jiayu Wu
- College of Biology and Environmental Science, Jishou, Jishou University, Jishou, Hunan, China.
| | - Yanheng Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China.
| | - Yang Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China.
| | - Wuyan Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China.
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China.
| | - Weihong Tan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
Cao J, Su Z, Zhang B, Yang J, Wang Y, Huang L, Cao G, Xie H, Zhong X, Zhu H, Jiang R, Li T, Xie Z, Lu W. Deciphering heart failure: an integrated proteomic and transcriptomic approach with experimental validation. Funct Integr Genomics 2024; 24:196. [PMID: 39441209 DOI: 10.1007/s10142-024-01475-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: 07/30/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
This study analyzed transcriptomic and proteomic data to identify molecular changes during heart failure (HF). Additionally,we embarked on an exploration of the prospect of therapeutic intervention through the manipulation of proteins implicated in ferroptosis. Three publicly available microarray datasets (GSE135055, GSE147236, GSE161472) profiling left ventricular samples from HF patients and healthy controls were obtained. Differentially expressed genes were identified in each dataset and cross-analyzed to determine shared gene signatures. Enrichment analysis of Gene Ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and gene set enrichment analysis were performed. Differentially expressed proteins were obtained from published proteomic studies and integrated with the transcriptomic results. To validate findings, a HF mouse model was generated and ferroptosis-related proteins were evaluated. Additionally, the effect of suppression of ferroptosis on hypoxia-induced ischemia model in HL-1 cardiomyocytes was assessed by knocking down Acyl-CoA synthetase long-chain family member 4 (ACSL4) using small interfering RNA (siRNA).Cross-analysis of differentially expressed genes (DEGs) in the GSE135055, GSE147236 and GSE161472 datasets revealed 224 up-regulated and 187 down-regulated potential genes which showed high enrichment in immune, inflammatory and metabolic pathways. Notably, four proteins, among them ACSL4, displayed consistent alterations at both the transcriptional and protein levels. In the HF mouse model, ACSL4 exhibited an elevation, whereas negative regulators of ferroptosis witnessed a decrement. Subsequently, knockdown of ACSL4 in a hypoxia-induced ischemic HL-1 cardiomyocyte cell model upregulated the expression of ferroptosis inhibitory protein and decreased the levels of reactive oxygen species (ROS), malondialdehyde (MDA)., and free iron and increased cell viability. Comprehensive multi-omics analysis revealed that the expression of the molecular target ACSL4 was increased in HF. Targeting ACSL4 to inhibit ferroptosis may represent a novel therapeutic strategy for HF treatment.
Collapse
Affiliation(s)
- Jun Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Zhaohai Su
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Bilong Zhang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Jiangyong Yang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Yueting Wang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Ling Huang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Gang Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Hui Xie
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Xiutong Zhong
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Hengqing Zhu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Rengui Jiang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 710032, Xi'an, China
| | - Zheng Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China.
| | - Weiling Lu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, 341000, Ganzhou, China.
| |
Collapse
|
17
|
Xie Z, Xie H, Xie C, Yang S, Feng Y, Su Z, Tang T, Zhang B, Yang J, Wang Y, Huang L, Zhu H, Cao J, Jiang R, Li T, Lu W. A combined analysis of bulk RNA-seq and scRNA-seq was performed to investigate the molecular mechanisms associated with the occurrence of myocardial infarction. BMC Genomics 2024; 25:921. [PMID: 39363266 PMCID: PMC11448016 DOI: 10.1186/s12864-024-10813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI) induces complex transcriptional changes across diverse cardiac cell types. Single-cell RNA sequencing (scRNA-seq) provides an unparalleled ability to discern cellular diversity during infarction, yet the veracity of these discoveries necessitates confirmation. This investigation sought to elucidate MI mechanisms by integrating scRNA-seq and bulk RNA-seq data. METHODS Publicly available scRNA-seq (GSE136088) and bulk RNA-seq (GSE153485) data from mice MI models were analyzed. Cell types were annotated, and differential expression analysis conducted. Bulk RNA-seq underwent quality control, principal component analysis, and differential expression analysis. RESULTS In scRNA-seq data, the comparison between MI and sham groups unveiled a reduction in endothelial cell populations, but macrophages and monocytes increased. Within fibroblast subgroups, three distinct categories were discerned, with two exhibiting upregulation in MI. Notably, endothelial cells exhibited an elevated expression of genes associated with apoptosis and ferroptosis. In bulk RNA-seq analysis, distinct patterns emerged when comparing MI and sham groups. Specifically, six genes linked to endothelial ferroptosis exhibited heightened expression in MI group, thereby corroborating the scRNA-seq findings. Moreover, the examination of isolated cardiac macrophages from mice MI model revealed increased expression of Spp1, Col1a2, Col3a1, Ctsd, and Lgals3 compared to sham group, thus substantiating the dysregulation of macrophage apoptosis-related proteins following MI. CONCLUSION MI altered the transcriptomic landscapes of cardiac cells with increased expression of apoptotic genes. Moreover, the upregulation of macrophage apoptosis marker was confirmed within MI models. The presence of endothelial cell depletion and ferroptosis in MI has been demonstrated.
Collapse
Affiliation(s)
- Zheng Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Huicong Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Chen Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Saichao Yang
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Yun Feng
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Zhaohai Su
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Tao Tang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Bilong Zhang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Jiangyong Yang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Yueting Wang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Ling Huang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Hengqing Zhu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Jun Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Rengui Jiang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Weiling Lu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China.
| |
Collapse
|
18
|
Liu L, Yu L, Wang Y, Zhou L, Liu Y, Pan X, Huang J. Unravelling the impact of RNA methylation genetic and epigenetic machinery in the treatment of cardiomyopathy. Pharmacol Res 2024; 207:107305. [PMID: 39002868 DOI: 10.1016/j.phrs.2024.107305] [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: 04/19/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Cardiomyopathy (CM) represents a heterogeneous group of diseases primarily affecting cardiac structure and function, with genetic and epigenetic dysregulation playing a pivotal role in its pathogenesis. Emerging evidence from the burgeoning field of epitranscriptomics has brought to light the significant impact of various RNA modifications, notably N6-methyladenosine (m6A), 5-methylcytosine (m5C), N7-methylguanosine (m7G), N1-methyladenosine (m1A), 2'-O-methylation (Nm), and 6,2'-O-dimethyladenosine (m6Am), on cardiomyocyte function and the broader processes of cardiac and vascular remodelling. These modifications have been shown to influence key pathological mechanisms including mitochondrial dysfunction, oxidative stress, cardiomyocyte apoptosis, inflammation, immune response, and myocardial fibrosis. Importantly, aberrations in the RNA methylation machinery have been observed in human CM cases and animal models, highlighting the critical role of RNA methylating enzymes and their potential as therapeutic targets or biomarkers for CM. This review underscores the necessity for a deeper understanding of RNA methylation processes in the context of CM, to illuminate novel therapeutic avenues and diagnostic tools, thereby addressing a significant gap in the current management strategies for this complex disease.
Collapse
Affiliation(s)
- Li Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Youjiang Medical University for Nationalities, Baise 533000, China; Laboratory of the Atherosclerosis and Ischemic Cardiovascular Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Linxing Yu
- Graduate School of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Yubo Wang
- Graduate School of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Liufang Zhou
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Yan Liu
- Laboratory of the Atherosclerosis and Ischemic Cardiovascular Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Xingshou Pan
- Laboratory of the Atherosclerosis and Ischemic Cardiovascular Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China.
| | - Jianjun Huang
- Youjiang Medical University for Nationalities, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China.
| |
Collapse
|
19
|
Wang H, Li S, Zhang J, Peng W, Li T, Zhang J. Efficacy of selective serotonin reuptake inhibitors-related antidepressants in Alzheimer's disease: a meta-analysis. Eur J Med Res 2024; 29:438. [PMID: 39210432 PMCID: PMC11360319 DOI: 10.1186/s40001-024-02006-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE To study the effects of selective serotonin reuptake inhibitors (SSRIs) on cognitive functions, mental improvements, and adverse effects in patients with Alzheimer's disease (AD). METHODS Registered in INPLASY (INPLASY202450004), five drugs (citalopram, s-citalopram, quetiapine, olanzapine, and sertraline) were selected as representatives. A comprehensive search was conducted in PubMed, EMBASE, Web of Science, and the Cochrane Library up to May 15, 2024. Search terms were combined using Boolean operators, specifically 'AND' between different categories (e.g., 'Alzheimer's Disease' AND 'SSRIs') and 'OR' within the same category (e.g., 'citalopram OR s-citalopram OR quetiapine OR olanzapine OR sertraline'), to ensure a thorough retrieval of relevant studies. The selection followed rigorous inclusion and exclusion criteria for meta-analysis. RESULTS Fourteen articles from 1118 were selected for meta-analysis. The indicators, including Neuropsychiatric Inventory (NPI), Mini-Mental State Examination (MMSE), Brief Psychiatric Rating Scale (BPRS), and Cornell Scale for Depression in Dementia (CSDD), were used to assess the effects of the drugs on AD treatment. According to the results of NPI, CSDD, BPRS, MMSE, and security assessments, the five antidepressants have significant advantages in AD treatment compared with placebo, while the MMSE of the patient treated with the antidepressants did not show notable changes compared with patients treated only with placebo. Statistical analyses were conducted using Review Manager 5.3, employing random-effects models to account for study heterogeneity and sensitivity analyses to test the robustness of our findings. CONCLUSION This study suggests that SSRI-related antidepressants have great potential values in AD treatment, and further research on the application of SSRI-related antidepressants in AD treatment is necessary.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, 247 Beiyuan St, Jinan, 250033, China
| | - Siyi Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Jiwei Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wei Peng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianxin Zhang
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, 247 Beiyuan St, Jinan, 250033, China.
| |
Collapse
|
20
|
Yang W, Lei X, Liu F, Sui X, Yang Y, Xiao Z, Cui Z, Sun Y, Yang J, Yang X, Lin X, Bao Z, Li W, Ma Y, Wang Y, Luo Y. Meldonium, as a potential neuroprotective agent, promotes neuronal survival by protecting mitochondria in cerebral ischemia-reperfusion injury. J Transl Med 2024; 22:771. [PMID: 39148053 PMCID: PMC11325598 DOI: 10.1186/s12967-024-05222-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/19/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Stroke is a globally dangerous disease capable of causing irreversible neuronal damage with limited therapeutic options. Meldonium, an inhibitor of carnitine-dependent metabolism, is considered an anti-ischemic drug. However, the mechanisms through which meldonium improves ischemic injury and its potential to protect neurons remain largely unknown. METHODS A rat model with middle cerebral artery occlusion (MCAO) was used to investigate meldonium's neuroprotective efficacy in vivo. Infarct volume, neurological deficit score, histopathology, neuronal apoptosis, motor function, morphological alteration and antioxidant capacity were explored via 2,3,5-Triphenyltetrazolium chloride staining, Longa scoring method, hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay, rotarod test, transmission electron microscopy and Oxidative stress index related kit. A primary rat hippocampal neuron model subjected to oxygen-glucose deprivation reperfusion was used to study meldonium's protective ability in vitro. Neuronal viability, mitochondrial membrane potential, mitochondrial morphology, respiratory function, ATP production, and its potential mechanism were assayed by MTT cell proliferation and cytotoxicity assay kit, cell-permeant MitoTracker® probes, mitochondrial stress, real-time ATP rate and western blotting. RESULTS Meldonium markedly reduced the infarct size, improved neurological function and motor ability, and inhibited neuronal apoptosis in vivo. Meldonium enhanced the morphology, antioxidant capacity, and ATP production of mitochondria and inhibited the opening of the mitochondrial permeability transition pore in the cerebral cortex and hippocampus during cerebral ischemia-reperfusion injury (CIRI) in rats. Additionally, meldonium improved the damaged fusion process and respiratory function of neuronal mitochondria in vitro. Further investigation revealed that meldonium activated the Akt/GSK-3β signaling pathway to inhibit mitochondria-dependent neuronal apoptosis. CONCLUSION Our study demonstrated that meldonium shows a neuroprotective function during CIRI by preserving the mitochondrial function, thus prevented neurons from apoptosis.
Collapse
Affiliation(s)
- Weijie Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiuxing Lei
- Lu'An Hospital of Traditional Chinese Medicine, Anhui, China
| | - Fengying Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhenyu Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ziqi Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yangyang Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinyi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xueyang Lin
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhenghao Bao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Weidong Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yingkai Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| |
Collapse
|
21
|
Yang Q, Sun S, Cui LB, Gao S, Gu Z, Fang Z, Zhang Y, Chen S, Sun N, Wang Y, Cao F. Ischemic cardio-cerebrovascular disease and all-cause mortality in Chinese elderly patients: a propensity-score matching study. Eur J Med Res 2024; 29:330. [PMID: 38879523 PMCID: PMC11179225 DOI: 10.1186/s40001-024-01929-x] [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: 03/20/2024] [Accepted: 06/06/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND Ischemic cardio-cerebrovascular disease is the leading cause of mortality worldwide. However, studies focusing on elderly and very elderly patients are scarce. Hence, our study aimed to characterize and investigate the long-term prognostic implications of ischemic cardio-cerebrovascular diseases in elderly Chinese patients. METHODS This retrospective cohort study included 1026 patients aged ≥ 65 years who were categorized into the mono ischemic cardio-cerebrovascular disease (MICCD) (either coronary artery disease or ischemic stroke/transient ischemic attack) (n = 912) and the comorbidity of ischemic cardio-cerebrovascular disease (CICCD) (diagnosed with both coronary artery disease and ischemic stroke/transient ischemic attack at admission) (n = 114). The primary outcome was all-cause death. The mortality risk was evaluated using the Cox proportional hazards risk model with multiple adjustments by conventional and propensity-score-based approaches. RESULTS Of the 2494 consecutive elderly patients admitted to the hospital, 1026 (median age 83 years [interquartile range]: 76.5-86.4; 94.4% men) met the inclusion criteria. Patients with CICCD consisted mostly of very elderly (79.2% vs. 66.1%, P < 0.001) individuals with a higher burden of comorbidities. Over a median follow-up of 10.4 years, 398 (38.8%) all-cause deaths were identified. Compared with the MICCD group, the CICCD group exhibited a higher adjusted hazard ratio (HR) (95% confidential interval, CI) of 1.71 (1.32-2.39) for long-term mortality after adjusting for potential confounders. The sensitivity analysis results remained robust. After inverse probability of treatment weighting (IPTW) modeling, the CICCD group displayed an even worse mortality risk (IPTW-adjusted HR: 2.07; 95% CI 1.47-2.90). In addition, anemia (adjusted HR: 1.48; 95% CI 1.16-1.89) and malnutrition (adjusted HR: 1.43; 95% CI 1.15-1.78) are also independent risk factors for all-cause mortality among elderly and very elderly patients. CONCLUSIONS Our results thus suggest that elderly patients with ischemic cardio-cerebrovascular disease and anemia or malnutrition may have higher mortality, which may be predicted upon admission. These findings, however, warrant further investigation.
Collapse
Affiliation(s)
- Qian Yang
- Medical School of Chinese PLA, Beijing, 100039, China
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Shasha Sun
- The Fifth Department of Cadre Health Care, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Long-Biao Cui
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Shan Gao
- Medical School of Chinese PLA, Beijing, 100039, China
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhenghui Gu
- Medical School of Chinese PLA, Beijing, 100039, China
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiyi Fang
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 30071, China
| | - Yingjie Zhang
- Medical School of Chinese PLA, Beijing, 100039, China
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Sijia Chen
- Medical School of Chinese PLA, Beijing, 100039, China
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Naiyuan Sun
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 30071, China
| | - Yabin Wang
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Feng Cao
- Department of Cardiology, The Second Medical Center & National Clinical Research, Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
22
|
Vedunova M, Borysova O, Kozlov G, Zharova AM, Morgunov I, Moskalev A. Candidate molecular targets uncovered in mouse lifespan extension studies. Expert Opin Ther Targets 2024; 28:513-528. [PMID: 38656034 DOI: 10.1080/14728222.2024.2346597] [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: 09/22/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Multiple interventions have demonstrated an increase in mouse lifespan. However, non-standardized controls, sex or strain-specific factors, and insufficient focus on targets, hinder the translation of these findings into clinical applications. AREAS COVERED We examined the effects of genetic and drug-based interventions on mice from databases DrugAge, GenAge, the Mouse Phenome Database, and publications from PubMed that led to a lifespan extension of more than 10%, identifying specific molecular targets that were manipulated to achieve the maximum lifespan in mice. Subsequently, we characterized 10 molecular targets influenced by these interventions, with particular attention given to clinical trials and potential indications for each. EXPERT OPINION To increase the translational potential of mice life-extension studies to clinical research several factors are crucial: standardization of mice lifespan research approaches, the development of clear criteria for control and experimental groups, the establishment of criteria for potential geroprotectors, and focusing on targets and their clinical application. Pinpointing the targets affected by geroprotectors helps in understanding species-specific differences and identifying potential side effects, ensuring the safety and effectiveness of clinical trials. Additionally, target review facilitates the optimization of treatment protocols and the evaluation of the clinical feasibility of translating research findings into practical therapies for humans.
Collapse
Affiliation(s)
- Maria Vedunova
- Institute of Biomedicine, Institute of Biogerontology, National Research Lobachevsky State University of Nizhni Novgorod (Lobachevsky University), Nizhny Novgorod, Russia
| | | | - Grigory Kozlov
- Institute of Biomedicine, Institute of Biogerontology, National Research Lobachevsky State University of Nizhni Novgorod (Lobachevsky University), Nizhny Novgorod, Russia
| | - Anna-Maria Zharova
- Institute of Biomedicine, Institute of Biogerontology, National Research Lobachevsky State University of Nizhni Novgorod (Lobachevsky University), Nizhny Novgorod, Russia
| | | | - Alexey Moskalev
- Institute of Biomedicine, Institute of Biogerontology, National Research Lobachevsky State University of Nizhni Novgorod (Lobachevsky University), Nizhny Novgorod, Russia
- Longaevus Technologies LTD, London, United Kingdom
- Russian Gerontology Research and Clinical Centre, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
23
|
Song XW, He WX, Su T, Li CJ, Jiang LL, Huang SQ, Li SH, Guo ZF, Zhang BL. Abnormal expression of PRKAG2-AS1 in endothelial cells induced inflammation and apoptosis by reducing PRKAG2 expression. Noncoding RNA Res 2024; 9:536-546. [PMID: 38511052 PMCID: PMC10950609 DOI: 10.1016/j.ncrna.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
PRKAG2 is required for the maintenance of cellular energy balance. PRKAG2-AS1, a long non-coding RNA (lncRNA), was found within the promoter region of PRKAG2. Despite the extensive expression of PRKAG2-AS1 in endothelial cells, the precise function and mechanism of this gene in endothelial cells have yet to be elucidated. The localization of PRKAG2-AS1 was predominantly observed in the nucleus, as revealed using nuclear and cytoplasmic fractionation and fluorescence in situ hybridization. The manipulation of PRKAG2-AS1 by knockdown and overexpression within the nucleus significantly altered PRKAG2 expression in a cis-regulatory manner. The expression of PRKAG2-AS1 and its target genes, PRKAG2b and PRKAG2d, was down-regulated in endothelial cells subjected to oxLDL and Hcy-induced injury. This finding suggests that PRKAG2-AS1 may be involved in the mechanism behind endothelial injury. The suppression of PRKAG2-AS1 specifically in the nucleus led to an upregulation of inflammatory molecules such as cytokines, adhesion molecules, and chemokines in endothelial cells. Additionally, this nuclear suppression of PRKAG2-AS1 facilitated the adherence of THP1 cells to endothelial cells. We confirmed the role of nuclear knockdown PRKAG2-AS1 in the induction of apoptosis and inhibition of cell proliferation, migration, and lumen formation through flow cytometry, TUNEL test, CCK8 assay, and cell scratching. Finally, it was determined that PRKAG2-AS1 exerts direct control over the transcription of PRKAG2 by its binding to their promoters. In conclusion, downregulation of PRKAG2-AS1 suppressed the proliferation and migration, promoted inflammation and apoptosis of endothelial cells, and thus contributed to the development of atherosclerosis resulting from endothelial cell injury.
Collapse
Affiliation(s)
- Xiao-Wei Song
- Department of Anesthesiology, Shidong Hospital of Shanghai, University of Shanghai for Science and Technology, Shanghai, China
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wen-Xia He
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ting Su
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chang-Jin Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li-Li Jiang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Song-Qun Huang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Song-Hua Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhi-Fu Guo
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Bi-Li Zhang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
24
|
Luan Y, Zhu X, Jiao Y, Liu H, Huang Z, Pei J, Xu Y, Yang Y, Ren K. Cardiac cell senescence: molecular mechanisms, key proteins and therapeutic targets. Cell Death Discov 2024; 10:78. [PMID: 38355681 PMCID: PMC10866973 DOI: 10.1038/s41420-023-01792-5] [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: 09/04/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Cardiac aging, particularly cardiac cell senescence, is a natural process that occurs as we age. Heart function gradually declines in old age, leading to continuous heart failure, even in people without a prior history of heart disease. To address this issue and improve cardiac cell function, it is crucial to investigate the molecular mechanisms underlying cardiac senescence. This review summarizes the main mechanisms and key proteins involved in cardiac cell senescence. This review further discusses the molecular modulators of cellular senescence in aging hearts. Furthermore, the discussion will encompass comprehensive descriptions of the key drugs, modes of action and potential targets for intervention in cardiac senescence. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of cardiac senescence, this review seeks to provide a fresh perspective and important theoretical foundations for the development of drugs targeting this condition.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Xiaofan Zhu
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Jinyan Pei
- Quality Management Department, Henan No.3 Provincial People's Hospital, Zhengzhou, 450052, P. R. China
| | - Yawei Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
25
|
Salminen A. AMPK signaling inhibits the differentiation of myofibroblasts: impact on age-related tissue fibrosis and degeneration. Biogerontology 2024; 25:83-106. [PMID: 37917219 PMCID: PMC10794430 DOI: 10.1007/s10522-023-10072-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023]
Abstract
Disruption of the extracellular matrix (ECM) and an accumulation of fibrotic lesions within tissues are two of the distinctive hallmarks of the aging process. Tissue fibroblasts are mesenchymal cells which display an impressive plasticity in the regulation of ECM integrity and thus on tissue homeostasis. Single-cell transcriptome studies have revealed that tissue fibroblasts exhibit a remarkable heterogeneity with aging and in age-related diseases. Excessive stress and inflammatory insults induce the differentiation of fibroblasts into myofibroblasts which are fusiform contractile cells and abundantly secrete the components of the ECM and proteolytic enzymes as well as many inflammatory mediators. Detrimental stresses can also induce the transdifferentiation of certain mesenchymal and myeloid cells into myofibroblasts. Interestingly, many age-related stresses, such as oxidative and endoplasmic reticulum stresses, ECM stiffness, inflammatory mediators, telomere shortening, and several alarmins from damaged cells are potent inducers of myofibroblast differentiation. Intriguingly, there is convincing evidence that the signaling pathways stimulated by the AMP-activated protein kinase (AMPK) are potent inhibitors of myofibroblast differentiation and accordingly AMPK signaling reduces fibrotic lesions within tissues, e.g., in age-related cardiac and pulmonary fibrosis. AMPK signaling is not only an important regulator of energy metabolism but it is also able to control cell fate determination and many functions of the immune system. It is known that AMPK signaling can delay the aging process via an integrated signaling network. AMPK signaling inhibits myofibroblast differentiation, e.g., by suppressing signaling through the TGF-β, NF-κB, STAT3, and YAP/TAZ pathways. It seems that AMPK signaling can alleviate age-related tissue fibrosis and degeneration by inhibiting the differentiation of myofibroblasts.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
26
|
Du J, Wu J, Zhang Y, Liu Q, Luo X, Huang X, Wang X, Yang F, Hou J. CTRP13 Mitigates Endothelial Cell Ferroptosis via the AMPK/KLF4 Pathway: Implications for Atherosclerosis Protection. Med Sci Monit 2024; 30:e942733. [PMID: 38273650 PMCID: PMC10826208 DOI: 10.12659/msm.942733] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/11/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND C1q/tumor necrosis factor-related protein 13 (CTRP13) preserves endothelial function and possesses anti-oxidation activity. However, its effects on ferroptosis of human umbilical vein endothelial cells (HUVECs) remain unclear. We investigated the effects of CTRP13 on HUVEC ferroptosis induced by oxidized low-density lipoprotein (ox-LDL) and explored the underlying mechanisms of CTRP13 against ferroptosis via the AMPK/KLF4 pathway. MATERIAL AND METHODS Cell Counting Kit-8 assay was used to evaluate cell viability. Lactate dehydrogenase activity and malondialdehyde content analysis were performed to evaluate the cell membrane integrity and lipid peroxidation. Mito-Tracker, JC-1, and 2',7'-dichlorofluorescein di-acetate were used to evaluate the biological activity of mitochondria, mitochondrial membrane potential, and reactive oxygen species (ROS) in endothelial cells. The ferroptosis indicator expressions, recombinant solute carrier family 7, member 11, glutathione peroxidase 4 (GPX4), and acyl-CoA synthetase long-chain family member 4 were examined using real-time reverse transcription-polymerase chain reaction and Western blot. Immunofluorescence staining detected GPX4 location in endothelial cells. RESULTS The results demonstrate that CTRP13 (450 ng/mL) prevented HUVEC ferroptosis by inhibiting ROS overproduction and mitochondrial dysfunction, and CTRP13 accelerated antioxidant enzyme expression levels, such as heme oxygenase 1, superoxide dismutase 1, and superoxide dismutase 2, compared with the ox-LDL (100 µg/mL) group for 48 h. Additionally, CTRP13 treatment increased p-AMPK/AMPK expression by 47.65% (P<0.05) while decreasing Krüppel-like factor 4 expression by 37.43% (P<0.05) in ox-LDL-induced HUVECs and elucidated the protective effect on endothelial dysfunction from ferroptosis. CONCLUSIONS These findings provide new insights for understanding the effects and mechanism of CTRP13 on preventing endothelial cell ferroptosis.
Collapse
Affiliation(s)
- Jie Du
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Jianjun Wu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Youqi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Qi Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xing Luo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xingtao Huang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xuedong Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Fan Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - JingBo Hou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| |
Collapse
|
27
|
Song XW, Su T, Li B, Huang YJ, He WX, Jiang LL, Li CJ, Huang SQ, Li SH, Guo ZF, Wu H, Zhang BL. Abnormal expression of PRKAG2-AS results in dysfunction of cardiomyocytes through regulating PRKAG2 transcription by interacting with PPARG. Clin Epigenetics 2023; 15:178. [PMID: 37932845 PMCID: PMC10629191 DOI: 10.1186/s13148-023-01591-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023] Open
Abstract
The role of PRKAG2 in the maintenance of heart function is well established, but little is known about how PRKAG2 is regulated in cardiomyocytes. In this study, we investigated the role of the lncRNA PRKAG2-AS, which is present at the PRKAG2 promoter, in the regulation of PRKAG2 expression. PRKAG2-AS expression was predominantly nuclear, as determined by RNA nucleoplasmic separation and fluorescence in situ hybridization. Knockdown of PRKAG2-AS in the nucleus, but not the cytoplasm, significantly decreased the expression of PRKAG2b and PRKAG2d. Interestingly, we found that PRKAG2-AS and its target genes, PRKAG2b and PRKAG2d, were reduced in the hearts of patients with ischemic cardiomyopathy, suggesting a potential role for PRKAG2-AS in myocardial ischemia. Indeed, knockdown of PRKAG2-AS in the nucleus resulted in apoptosis of cardiomyocytes. We further elucidated the mechanism by which PRKAG2-AS regulates PRKAG2 transcription by identifying 58 PRKAG2-AS interacting proteins. Among them, PPARG was selected for further investigation based on its correlation and potential interaction with PRKAG2-AS in regulating transcription. Overexpression of PPARG, or its activation with rosiglitazone, led to a significant increase in the expression of PRKAG2b and PRKAG2d in cardiomyocytes, which could be attenuated by PRKAG2-AS knockdown. This finding suggests that PRKAG2-AS mediates, at least partially, the protective effects of rosiglitazone on hypoxia-induced apoptosis. However, given the risk of rosiglitazone in heart failure, we also examined the involvement of PRKAG2-AS in this condition and found that PRKAG2-AS, as well as PRKAG2b and PRKAG2d, was elevated in hearts with dilated cardiomyopathy (DCM) and that overexpression of PRKAG2-AS led to a significant increase in PRKAG2b and PRKAG2d expression, indicating that up-regulation of PRKAG2-AS may contribute to the mechanism of heart failure by promoting transcription of PRKAG2. Consequently, proper expression of PRKAG2-AS is essential for maintaining cardiomyocyte function, and aberrant PRKAG2-AS expression induced by hypoxia or other stimuli may cause cardiac dysfunction.
Collapse
Grants
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
- 82000283, 82070419, 82170275 and 82170233 National Natural Science Foundation of China
Collapse
Affiliation(s)
- Xiao-Wei Song
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Ting Su
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Bo Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yun-Jie Huang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Wen-Xia He
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Li-Li Jiang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Chang-Jin Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Song-Qun Huang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Song-Hua Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhi-Fu Guo
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Hong Wu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Bi-Li Zhang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
28
|
Yang X, Wang J, Dai X, Ma N, Cheng H, Guo H, Chen S, Huang Y, Wu J. The mechanism and targeted intervention of the HIF-1 pathway in improving atherosclerotic heart's sensitivity to ischemic postconditioning. Free Radic Biol Med 2023; 208:494-509. [PMID: 37660838 DOI: 10.1016/j.freeradbiomed.2023.08.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND IPoC possesses a preventive effect against IR injury in healthy myocardium, but IPoC's protective effect on atherosclerotic myocardium is controversial. The current investigation aims to determine whether IPoC remains protective in atherosclerotic myocardium subjected to ischemia-reperfusion (IR) injury; to explore the specific mechanisms by which IPoC exerts cardioprotection; to explore whether HIF-1 upregulation combined with IPoC could further the provide cardioprotection; and to gaze at the specific mechanism whereby combined treatment expert the cardioprotection. METHODS ApoE-/- mice fed with a high-fat diet (HFD) were used to develop a model of atherosclerosis. The myocardial IR model was induced by occlusion of the left anterior descending (LAD) artery for 45 min, followed by reperfusion for 120 min. The protection of IPoC in both healthy and atherosclerotic myocardium was evaluated by measuring oxidative stress, apoptosis, infarct size, pathology, mitochondrial dysfunction and morphology of myocardium. The specific mechanism by which IPoC exerts cardioprotection in healthy and atherosclerotic myocardium was observed by measuring the expression of proteins involved in HIF-1, APMK and RISK pathways. The effect of HIF-1α overexpression on the cardioprotection by IPoC was observed by intravenous AAV9 -HIF-1α injection. RESULTS In healthy ischemic myocardium, IPoC exerted myocardial protective effects (antioxidant, anti-apoptosis, and improved mitochondrial function) through the activation of HIF-1, AMPK and RISK pathways. In atherosclerotic ischemic myocardium, IPoC exerted cardioprotection only through the activation of HIF-1 pathway; however, HIF-1 overexpression combined IPoC restored the activation of AMPK and RISK pathways, thereby further alleviating the myocardial IR injury. CONCLUSIONS In the atherosclerotic state, the HIF-1 pathway is the intrinsic mechanism by which IPoC exerts cardioprotective effects. The combination of HIF-1 upregulation and IPoC has a significant effect in reducing myocardial injury, which is worth being promoted and advocated. In addition, HIF-1-AMPK and HIF-1-RISK may be two endogenous cardioprotective signalling pathways with great value, which deserve to be thoroughly investigated in the future.
Collapse
Affiliation(s)
- Xue Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaowen Dai
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ning Ma
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hu Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hai Guo
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Siyu Chen
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yidan Huang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jianjiang Wu
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
29
|
Tang B, Luo Z, Zhang R, Zhang D, Nie G, Li M, Dai Y. An update on the molecular mechanism and pharmacological interventions for Ischemia-reperfusion injury by regulating AMPK/mTOR signaling pathway in autophagy. Cell Signal 2023; 107:110665. [PMID: 37004834 DOI: 10.1016/j.cellsig.2023.110665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
AMP-activated protein kinase (5'-adenosine monophosphate-activated protein kinase, AMPK)/mammalian target of rapamycin (mTOR) is an important signaling pathway maintaining normal cell function and homeostasis in vivo. The AMPK/mTOR pathway regulates cellular proliferation, autophagy, and apoptosis. Ischemia-reperfusion injury (IRI) is secondary damage that frequently occurs clinically in various disease processes and treatments, and the exacerbated injury during tissue reperfusion increases disease-associated morbidity and mortality. IRI arises from multiple complex pathological mechanisms, among which cell autophagy is a focus of recent research and a new therapeutic target. The activation of AMPK/mTOR signaling in IRI can modulate cellular metabolism and regulate cell proliferation and immune cell differentiation by adjusting gene transcription and protein synthesis. Thus, the AMPK/mTOR signaling pathway has been intensively investigated in studies focused on IRI prevention and treatment. In recent years, AMPK/mTOR pathway-mediated autophagy has been found to play a crucial role in IRI treatment. This article aims to elaborate the action mechanisms of AMPK/mTOR signaling pathway activation in IRI and summarize the progress of AMPK/mTOR-mediated autophagy research in the field of IRI therapy.
Collapse
Affiliation(s)
- Bin Tang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Zhijian Luo
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Rong Zhang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Dongmei Zhang
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Guojun Nie
- The First Outpatient Department of People's Liberation Army Western Theater General Hospital, Cheng Du, Sichuan Province 61000, China
| | - Mingxing Li
- Department of Ultrasound, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Yan Dai
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
30
|
Tian X, Huang Y, Zhang X, Fang R, Feng Y, Zhang W, Li L, Li T. Salidroside attenuates myocardial ischemia/reperfusion injury via AMPK-induced suppression of endoplasmic reticulum stress and mitochondrial fission. Toxicol Appl Pharmacol 2022; 448:116093. [PMID: 35659894 DOI: 10.1016/j.taap.2022.116093] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 12/19/2022]
Abstract
Ischemic heart disease (IHD) is the primary cause of death worldwide. Salidroside (Sal), the major active compound derived from Rhodiola rosea, is believed to have cardioprotective effects. AMP-activated protein kinase (AMPK), is a pivotal AMP-activated protein kinase in energy metabolism. Whether Sal plays an anti-endoplasmic reticulum stress/mitochondrial fission role through AMPK remains elusive. In this study, we established a myocardial ischemia/reperfusion (I/R) rat model. Rat hearts exposed to Sal with or without compound C were then subjected to I/R. Further, H9c2 cardiomyocytes were subjected to simulated ischemia/reperfusion (SIR) by hypoxia-reoxygenation. The rats and cardiomyocytes were pretreated with Sal, followed by Compound C and AMPK-siRNA to block AMPK activity. We found that Sal significantly ameliorated cardiac function, mitigated infarct size and serum content of lactate dehydrogenase and creatine kinase, improved mitochondrial function, and reduced mitochondrial fission and apoptosis. Furthermore, in cultured H9c2 cardiomyocytes, Sal increased the cell viability and inhibited SIR-induced myocardial apoptosis and mitochondrial fission. Furthermore, the translocation of Drp1 from the cytoplasm to mitochondria induced by salidroside was confirmed both in vivo and in vitro. However, the use of Compound C or AMPK siRNA to block AMPK activity leads to blockade of the protective effects of Sal. In summary, protects against myocardial I/R by activating the AMPK signaling pathway, inhibiting ER stress, and reducing mitochondrial fission and apoptosis.
Collapse
Affiliation(s)
- Xin Tian
- Department of Cardiologya, Traditional Chinese Medicine Hospital of Shaanxi Province, No. 4 Xihuamen Street, Xi'an 710003, China
| | - Ye Huang
- Department of Emergency, Xiyuan Hospital, China Academy of Chinese Medical Sciences, No. 1 Xiyuan playground Street, Beijing 100091, China
| | - Xiaofeng Zhang
- Department of Nephrology, Traditional Chinese Medicine Hospital of Shaanxi Province, No. 4 Xihuamen Street, Xi'an 710003, China
| | - Rong Fang
- School of Graduate, Shaanxi University of Traditional Chinese Medicine, No.1 Middle Weiyang Road, Xianyang 712046, China
| | - Yi Feng
- School of Graduate, Shaanxi University of Traditional Chinese Medicine, No.1 Middle Weiyang Road, Xianyang 712046, China
| | - Wanfang Zhang
- School of Graduate, Shaanxi University of Traditional Chinese Medicine, No.1 Middle Weiyang Road, Xianyang 712046, China
| | - Ling Li
- Department of Cardiologya, Traditional Chinese Medicine Hospital of Shaanxi Province, No. 4 Xihuamen Street, Xi'an 710003, China; Department of Geriatrics, Traditional Chinese Medicine Hospital of Shaanxi Province, No. 4 Xihuamen Street, Xi'an 710003, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
31
|
Cui Z, Zhao X, Amevor FK, Du X, Wang Y, Li D, Shu G, Tian Y, Zhao X. Therapeutic application of quercetin in aging-related diseases: SIRT1 as a potential mechanism. Front Immunol 2022; 13:943321. [PMID: 35935939 PMCID: PMC9355713 DOI: 10.3389/fimmu.2022.943321] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022] Open
Abstract
Quercetin, a naturally non-toxic flavonoid within the safe dose range with antioxidant, anti-apoptotic and anti-inflammatory properties, plays an important role in the treatment of aging-related diseases. Sirtuin 1 (SIRT1), a member of NAD+-dependent deacetylase enzyme family, is extensively explored as a potential therapeutic target for attenuating aging-induced disorders. SIRT1 possess beneficial effects against aging-related diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), Depression, Osteoporosis, Myocardial ischemia (M/I) and reperfusion (MI/R), Atherosclerosis (AS), and Diabetes. Previous studies have reported that aging increases tissue susceptibility, whereas, SIRT1 regulates cellular senescence and multiple aging-related cellular processes, including SIRT1/Keap1/Nrf2/HO-1 and SIRTI/PI3K/Akt/GSK-3β mediated oxidative stress, SIRT1/NF-κB and SIRT1/NLRP3 regulated inflammatory response, SIRT1/PGC1α/eIF2α/ATF4/CHOP and SIRT1/PKD1/CREB controlled phosphorylation, SIRT1-PINK1-Parkin mediated mitochondrial damage, SIRT1/FoxO mediated autophagy, and SIRT1/FoxG1/CREB/BDNF/Trkβ-catenin mediated neuroprotective effects. In this review, we summarized the role of SIRT1 in the improvement of the attenuation effect of quercetin on aging-related diseases and the relationship between relevant signaling pathways regulated by SIRT1. Moreover, the functional regulation of quercetin in aging-related markers such as oxidative stress, inflammatory response, mitochondrial function, autophagy and apoptosis through SIRT1 was discussed. Finally, the prospects of an extracellular vesicles (EVs) as quercetin loading and delivery, and SIRT1-mediated EVs as signal carriers for treating aging-related diseases, as well as discussed the ferroptosis alleviation effects of quercetin to protect against aging-related disease via activating SIRT1. Generally, SIRT1 may serve as a promising therapeutic target in the treatment of aging-related diseases via inhibiting oxidative stress, reducing inflammatory responses, and restoring mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zhifu Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiaxia Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Gang Shu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yaofu Tian
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Xiaoling Zhao,
| |
Collapse
|
32
|
Li W, Bai X, Hao J, Xu X, Lin F, Jiang Q, Ding C, Dai G, Peng F, Zhang M, Feng Y, Wang J, Chen X, Xue T, Guo X, Fu Z, Chen WH, Zhang L, Wang C, Jiao L. Thrombosis origin identification of cardioembolism and large artery atherosclerosis by distinct metabolites. J Neurointerv Surg 2022:neurintsurg-2022-019047. [PMID: 35654581 DOI: 10.1136/neurintsurg-2022-019047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/13/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND The diagnosis of cerebral thrombosis origin is challenging and remains unclear. This study aims to identify thrombosis due to cardioembolism (CE) and large artery atherosclerosis (LAA) from a new perspective of distinct metabolites. METHODS Distinct metabolites between 26 CE and 22 LAA origin thrombi, which were extracted after successful mechanical thrombectomy in patients with acute ischemic stroke in the anterior circulation, were analyzed with a ultra performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) system. Enriched metabolic pathways related to the metabolites were identified. Least absolute shrinkage selection operator regression analyses and a filtering method were used to select potential predictors. Furthermore, four machine learning classifiers, including decision tree, logistic regression, random forest (RF), and k means unsupervised classification model, were used to evaluate the predictive ability of the selected metabolites. RESULTS UPLC-QTOF-MS analysis revealed that levels of 88 and 55 metabolites were elevated in LAA and CE thrombi, respectively. Kyoto Encyclopedia of Genes and Genomes analysis revealed a significant difference between the pathways enriched in the two types of thrombi. Six metabolites (diglyceride (DG, 18:3/24:0), DG (22:0/24:0), phytosphingosine, galabiosylceramide (18:1/24:1), triglyceride (15:0/16:1/o-18:0), and glucosylceramide (18:1/24:0)) were finally selected to build a predictive model. The predictive RF model was confirmed to be the best, with a satisfactory stability and prediction capacity (area under the curve=0.889). CONCLUSIONS Six metabolites as potential predictors for distinguishing between cerebral thrombi of CE and LAA origin were identified. The results are useful for understanding the pathogenesis and for secondary stroke prevention.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China.,China International Neuroscience Institute (China-INI), Beijing, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,China International Neuroscience Institute (China-INI), Beijing, China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,China International Neuroscience Institute (China-INI), Beijing, China
| | - Feng Lin
- Department of Neurology, Sanming First Hospital and First Hospital of Sanming Affiliated to Fujian Medical University, Sanming City, Fujian Province, China
| | - Qunlong Jiang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Chunguang Ding
- National Center for Occupational Safety and Health, NHC, Beijing, China
| | - Gaolei Dai
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Fangda Peng
- National Center for Occupational Safety and Health, NHC, Beijing, China
| | - Meng Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Yao Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Xianyang Chen
- Zhongguancun Biological and Medical Big Data Center, Beijing, China.,Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Teng Xue
- Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China.,Zhongyuanborui Key Laborotory of Genetics and Metabolism, Guangdong-Macao In-depth Cooperation Zone in Hengqin, Zhuhai City, Guangdong Province, China
| | - Xiaofan Guo
- Department of Neurology, Loma Linda University Health, Loma Linda, California, USA
| | - Zhaolin Fu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,China International Neuroscience Institute (China-INI), Beijing, China
| | - Wen-Huo Chen
- Department of Neurology, Zhangzhou Affiliated Hospital, Fujian Medical University, Zhangzhou City, Fujian Province, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China .,China International Neuroscience Institute (China-INI), Beijing, China.,Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Jiang D, Liu C, Chen Y, Xing X, Zheng D. Whole body vibration activates AMPK/CPT1 signaling pathway of skeletal muscle in young and aging mice based on metabolomics study. Endocr J 2022; 69:585-596. [PMID: 34955464 DOI: 10.1507/endocrj.ej21-0343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Whole-body vibration (WBV) can improve skeletal muscle function in aging mice, but whether the effect on young and aging skeletal muscle is consistent has not been studied. We selected C57BL/6J mouse models, which were divided into young control group (YC), young vibration group (YV), aging control group (AC) and aging vibration group (AV). After 12 weeks of WBV, we found that compared with the YC group, the pathways of linoleic acid metabolism, biosynthesis of unsaturated fatty acids, arachidonic acid metabolism, nicotinate and nicotinamide metabolism, glycine, serine and threonine metabolism, and arginine and proline metabolism improved significantly in the YV group. Compared with the AC group, the pathways of arachidonic acid metabolism, alpha-linolenic acid metabolism, biosynthesis of unsaturated fatty acids, pentose and glucuronate interconversions and pentose phosphate pathway improved significantly in the AV group. Furthermore, we found that WBV decreased triglyceride (TG), total cholesterol (TC), and free fatty acid (FFA) levels in aging mice, improved mitochondrial membrane potential, and increased the expression of phosphorylated activated protein kinase (p-AMPK), peroxisome proliferator-activated receptor coactivator-1α (PGC-1α) and carnitine palmitoyl transferase 1B (CPT1B) in the skeletal muscle of young and aging mice. Our study revealed that WBV mainly improved lipid metabolism and amino acid metabolism pathways of skeletal muscle in young mice and mainly improved lipid metabolism and glucose metabolism pathways of skeletal muscle in aging mice. WBV can activate the AMPK/CPT1 signaling pathway and improve mitochondrial function in skeletal muscle in both young and aging mice.
Collapse
Affiliation(s)
- Dingwen Jiang
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chang Liu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ye Chen
- School of Mechanical Engineering and Automation, Liaoning University of Technology, Jinzhou, Liaoning, China
| | - Xuejiao Xing
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Danmeng Zheng
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
34
|
AMPK Activation Is Indispensable for the Protective Effects of Caloric Restriction on Left Ventricular Function in Postinfarct Myocardium. BIOLOGY 2022; 11:biology11030448. [PMID: 35336822 PMCID: PMC8945456 DOI: 10.3390/biology11030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022]
Abstract
Background: Caloric restriction (CR) extends lifespan in many species, including mammals. CR is cardioprotective in senescent myocardium by correcting pre-existing mitochondrial dysfunction and apoptotic activation. Furthermore, it confers cardioprotection against acute ischemia-reperfusion injury. Here, we investigated the role of AMP-activated protein kinase (AMPK) in mediating the cardioprotective CR effects in failing, postinfarct myocardium. Methods: Ligation of the left coronary artery or sham operation was performed in rats and mice. Four weeks after surgery, left ventricular (LV) function was analyzed by echocardiography, and animals were assigned to different feeding groups (control diet or 40% CR, 8 weeks) as matched pairs. The role of AMPK was investigated with an AMPK inhibitor in rats or the use of alpha 2 AMPK knock-out mice. Results: CR resulted in a significant improvement in LV function, compared to postinfarct animals receiving control diet in both species. The improvement in LV function was accompanied by a reduction in serum BNP, decrease in LV proapoptotic activation, and increase in mitochondrial biogenesis in the LV. Inhibition or loss of AMPK prevented most of these changes. Conclusions: The failing, postischemic heart is protected from progressive loss of LV systolic function by CR. AMPK activation is indispensable for these protective effects.
Collapse
|
35
|
The Role of Oxidative Stress in the Aging Heart. Antioxidants (Basel) 2022; 11:antiox11020336. [PMID: 35204217 PMCID: PMC8868312 DOI: 10.3390/antiox11020336] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022] Open
Abstract
Medical advances and the availability of diagnostic tools have considerably increased life expectancy and, consequently, the elderly segment of the world population. As age is a major risk factor in cardiovascular disease (CVD), it is critical to understand the changes in cardiac structure and function during the aging process. The phenotypes and molecular mechanisms of cardiac aging include several factors. An increase in oxidative stress is a major player in cardiac aging. Reactive oxygen species (ROS) production is an important mechanism for maintaining physiological processes; its generation is regulated by a system of antioxidant enzymes. Oxidative stress occurs from an imbalance between ROS production and antioxidant defenses resulting in the accumulation of free radicals. In the heart, ROS activate signaling pathways involved in myocyte hypertrophy, interstitial fibrosis, contractile dysfunction, and inflammation thereby affecting cell structure and function, and contributing to cardiac damage and remodeling. In this manuscript, we review recent published research on cardiac aging. We summarize the aging heart biology, highlighting key molecular pathways and cellular processes that underlie the redox signaling changes during aging. Main ROS sources, antioxidant defenses, and the role of dysfunctional mitochondria in the aging heart are addressed. As metabolism changes contribute to cardiac aging, we also comment on the most prevalent metabolic alterations. This review will help us to understand the mechanisms involved in the heart aging process and will provide a background for attractive molecular targets to prevent age-driven pathology of the heart. A greater understanding of the processes involved in cardiac aging may facilitate our ability to mitigate the escalating burden of CVD in older individuals and promote healthy cardiac aging.
Collapse
|
36
|
Li S, Yu Y, Wang B, Qiao S, Hu M, Wang H, Fu C, Dong B. Overexpression of G protein-coupled receptor 40 protects obesity-induced cardiomyopathy through the SIRT1/LKB1/AMPK pathway. Hum Gene Ther 2022; 33:598-613. [PMID: 35018806 DOI: 10.1089/hum.2021.176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Obesity has become a serious global public health problem, and cardiomyopathy caused by obesity has been paid more and more attention in recent years. As an important protein involved in glucose and lipid metabolism, G protein-coupled receptor 40 (GPR40) exerts cardioprotective effects in some disease models. The aim of this study was to explore whether GPR40 plays a protective role in obesity-induced cardiomyopathy. We established an obesity model by feeding rats with a high-fat diet, and H9c2 cells were stimulated with palmitic acid to mimic high-fat stimulation. Overexpression of GPR40 was achieved by infection with lentivirus or cDNA plasmids. Obesity-induced cardiac injury models exhibit cardiac dysfunction, myocardial hypertrophy and collagen accumulation, accompanied by increased inflammation, oxidative stress and apoptosis. However, GPR40 overexpression attenuated these alterations. Its anti-inflammatory effect may be through inhibiting the nuclear factor-κB pathway, and the anti-oxidative stress may be through activating the nuclear transcription factor erythroid 2-related factor 2 pathway. For the mechanism of GPR40 against obese cardiomyopathy, GPR40 overexpression not only activated the sirtuin 1 (SIRT1)- liver kinase B1 (LKB1)- AMP-activated protein kinase (AMPK) pathway, but also enhanced the binding of SIRT1 to LKB1. The anti-fibrotic, anti-inflammatory, anti-oxidative stress and anti-apoptotic effects of GPR40 overexpression were inhibited by SIRT1 small interfering RNA. In conclusion, GPR40 overexpression protects against obesity-induced cardiac injury in rats, possibly through the SIRT1- LKB1- AMPK pathway.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China, Jinan, China;
| | - Yalin Yu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China, Jinan, China;
| | - Boyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China, Jinan, China;
| | - Shiyuan Qiao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China, Jinan, China;
| | - Maomao Hu
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China, Jinan, China;
| | - Han Wang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China, Jinan, China;
| | - Changning Fu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China, Jinan, China.,Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China, Jinan, China;
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China, Jinan, Shandong, China;
| |
Collapse
|
37
|
Liu B, Deng B, Jiang X, Xu Y, Chen S, Cai M, Deng S, Ding W, Xu H, Zhang S, Tan ZB, Chen R, Zhang J. 10-gingerol, a natural AMPK agonist, suppresses neointimal hyperplasia and inhibits vascular smooth muscle cells proliferation. Food Funct 2022; 13:3234-3246. [DOI: 10.1039/d1fo03610f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background: Abnormal proliferation of vascular smooth muscle cells (VSMCs) in the intimal region is a key event in the development of neointimal hyperplasia. 10-G, a bioactive compound found in ginger,...
Collapse
|
38
|
Tang Z, Wei X, Li T, Wang W, Wu H, Dong H, Liu Y, Wei F, Shi L, Li X, Guo Z, Xiao X. Sestrin2-Mediated Autophagy Contributes to Drug Resistance via Endoplasmic Reticulum Stress in Human Osteosarcoma. Front Cell Dev Biol 2021; 9:722960. [PMID: 34646824 PMCID: PMC8502982 DOI: 10.3389/fcell.2021.722960] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/30/2021] [Indexed: 01/29/2023] Open
Abstract
One contributor to the high mortality of osteosarcoma is its reduced sensitivity to chemotherapy, but the mechanism involved is unclear. Improving the sensitivity of osteosarcoma to chemotherapy is urgently needed to improve patient survival. We found that chemotherapy triggered apoptosis of human osteosarcoma cells in vitro and in vivo; this was accompanied by increased Sestrin2 expression. Importantly, autophagy was also enhanced with increased Sestrin2 expression. Based on this observation, we explored the potential role of Sestrin2 in autophagy of osteosarcoma. We found that Sestrin2 inhibited osteosarcoma cell apoptosis by promoting autophagy via inhibition of endoplasmic reticulum stress, and this process is closely related to the PERK-eIF2α-CHOP pathway. In addition, our study showed that low Sestrin2 expression can effectively reduce autophagy of human osteosarcoma cells after chemotherapy, increase p-mTOR expression, decrease Bcl-2 expression, promote osteosarcoma cell apoptosis, and slow down tumour progression in NU/NU mice. Sestrin2 activates autophagy by inhibiting mTOR via the PERK-eIF2α-CHOP pathway and inhibits apoptosis via Bcl-2. Therefore, our results explain one underlying mechanism of increasing the sensitivity of osteosarcoma to chemotherapy and suggest that Sestrin2 is a promising gene target.
Collapse
Affiliation(s)
- Zhen Tang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinghui Wei
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Wei Wang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Hao Wu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hui Dong
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yichao Liu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Feilong Wei
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Lei Shi
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaokang Li
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Xin Xiao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
39
|
Zhang C, Xiang C, Tian X, Xue J, He G, Wu X, Mei Z, Li T. Roles of Nursing in the Management of Geriatric Cardiovascular Diseases. Front Med (Lausanne) 2021; 8:682218. [PMID: 34568358 PMCID: PMC8455815 DOI: 10.3389/fmed.2021.682218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/09/2021] [Indexed: 12/31/2022] Open
Abstract
The nursing field occupies the largest secion of the cardiovascular healthcare services. Despite this, the roles of nursing within the cardiovascular healthcare system has not been well displayed. The authors searched PubMed and Embase (between January 1, 1950, and June 17, 2021) and created a narrative review of recent publications regarding the role of nursing in the management of geriatric cardiovascular disease (CVD). Patients with geriatric CVD, which includes mainly myocardial ischemia and heart failure, were enrolled. Nursing can improve the outcomes of myocardial ischemia and heart failure. It plays a pivotal role in the recovery, rehabilitation, and outcomes of geriatric CVD, especially for chronic heart diseases. Taken together, this paper compiled is focused on the current status of cardiovascular nursing and may facilitate future treatment and rehabilitation in geriatric CVD.
Collapse
Affiliation(s)
- Chunzhi Zhang
- Department of Thoracic and Cardiovascular Surgery, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Congling Xiang
- Department of Cardiology, Xinchang Hospital Affiliated to Wenzhou Medical University, Xinchang, China
| | - Xin Tian
- Department of Cardiology, Traditional Chinese Medicine Hospital of Shaanxi Province, Xi'an, China
| | - Jun Xue
- Department of Vascular Surgery, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Gengxu He
- Department of Thoracic and Cardiovascular Surgery, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xueliang Wu
- Department of Vascular Surgery, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian Li
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
40
|
Clayton ZS, Hutton DA, Mahoney SA, Seals DR. Anthracycline chemotherapy-mediated vascular dysfunction as a model of accelerated vascular aging. ACTA ACUST UNITED AC 2021; 2:45-69. [PMID: 34212156 DOI: 10.1002/aac2.12033] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, and age is by far the greatest risk factor for developing CVD. Vascular dysfunction, including endothelial dysfunction and arterial stiffening, is responsible for much of the increase in CVD risk with aging. A key mechanism involved in vascular dysfunction with aging is oxidative stress, which reduces the bioavailability of nitric oxide (NO) and induces adverse changes to the extracellular matrix of the arterial wall (e.g., elastin fragmentation/degradation, collagen deposition) and an increase in advanced glycation end products, which form crosslinks in arterial wall structural proteins. Although vascular dysfunction and CVD are most prevalent in older adults, several conditions can "accelerate" these events at any age. One such factor is chemotherapy with anthracyclines, such as doxorubicin (DOXO), to combat common forms of cancer. Children, adolescents and young adults treated with these chemotherapeutic agents demonstrate impaired vascular function and an increased risk of future CVD development compared with healthy age-matched controls. Anthracycline treatment also worsens vascular dysfunction in mid-life (50-64 years of age) and older (65 and older) adults such that endothelial dysfunction and arterial stiffness are greater compared to age-matched controls. Collectively, these observations indicate that use of anthracycline chemotherapeutic agents induce a vascular aging-like phenotype and that the latter contributes to premature CVD in cancer survivors exposed to these agents. Here, we review the existing literature supporting these ideas, discuss potential mechanisms as well as interventions that may protect arteries from these adverse effects, identify research gaps and make recommendations for future research.
Collapse
|
41
|
Yu M, Zhang H, Wang B, Zhang Y, Zheng X, Shao B, Zhuge Q, Jin K. Key Signaling Pathways in Aging and Potential Interventions for Healthy Aging. Cells 2021; 10:cells10030660. [PMID: 33809718 PMCID: PMC8002281 DOI: 10.3390/cells10030660] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is a fundamental biological process accompanied by a general decline in tissue function. Indeed, as the lifespan increases, age-related dysfunction, such as cognitive impairment or dementia, will become a growing public health issue. Aging is also a great risk factor for many age-related diseases. Nowadays, people want not only to live longer but also healthier. Therefore, there is a critical need in understanding the underlying cellular and molecular mechanisms regulating aging that will allow us to modify the aging process for healthy aging and alleviate age-related disease. Here, we reviewed the recent breakthroughs in the mechanistic understanding of biological aging, focusing on the adenosine monophosphate-activated kinase (AMPK), Sirtuin 1 (SIRT1) and mammalian target of rapamycin (mTOR) pathways, which are currently considered critical for aging. We also discussed how these proteins and pathways may potentially interact with each other to regulate aging. We further described how the knowledge of these pathways may lead to new interventions for antiaging and against age-related disease.
Collapse
Affiliation(s)
- Mengdi Yu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
| | - Hongxia Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Brian Wang
- Pathnova Laboratories Pte. Ltd. 1 Research Link, Singapore 117604, Singapore;
| | - Yinuo Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
| | - Xiaoying Zheng
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
| | - Bei Shao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China;
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
- Correspondence: (Q.Z.); (K.J.); Tel.: +86-577-55579339 (Q.Z.); +1-81-7735-2579 (K.J.)
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: (Q.Z.); (K.J.); Tel.: +86-577-55579339 (Q.Z.); +1-81-7735-2579 (K.J.)
| |
Collapse
|
42
|
Viswanathan V, Puvvula A, Jamthikar AD, Saba L, Johri AM, Kotsis V, Khanna NN, Dhanjil SK, Majhail M, Misra DP, Agarwal V, Kitas GD, Sharma AM, Kolluri R, Naidu S, Suri JS. Bidirectional link between diabetes mellitus and coronavirus disease 2019 leading to cardiovascular disease: A narrative review. World J Diabetes 2021; 12:215-237. [PMID: 33758644 PMCID: PMC7958478 DOI: 10.4239/wjd.v12.i3.215] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/20/2020] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic where several comorbidities have been shown to have a significant effect on mortality. Patients with diabetes mellitus (DM) have a higher mortality rate than non-DM patients if they get COVID-19. Recent studies have indicated that patients with a history of diabetes can increase the risk of severe acute respiratory syndrome coronavirus 2 infection. Additionally, patients without any history of diabetes can acquire new-onset DM when infected with COVID-19. Thus, there is a need to explore the bidirectional link between these two conditions, confirming the vicious loop between "DM/COVID-19". This narrative review presents (1) the bidirectional association between the DM and COVID-19, (2) the manifestations of the DM/COVID-19 loop leading to cardiovascular disease, (3) an understanding of primary and secondary factors that influence mortality due to the DM/COVID-19 loop, (4) the role of vitamin-D in DM patients during COVID-19, and finally, (5) the monitoring tools for tracking atherosclerosis burden in DM patients during COVID-19 and "COVID-triggered DM" patients. We conclude that the bidirectional nature of DM/COVID-19 causes acceleration towards cardiovascular events. Due to this alarming condition, early monitoring of atherosclerotic burden is required in "Diabetes patients during COVID-19" or "new-onset Diabetes triggered by COVID-19 in Non-Diabetes patients".
Collapse
Affiliation(s)
- Vijay Viswanathan
- M Viswanathan Hospital for Diabetes, M Viswanathan Diabetes Research Centre, Chennai 600013, India
| | - Anudeep Puvvula
- Annu’s Hospitals for Skin and Diabetes, Nellore 524101, Andhra Pradesh, India
| | - Ankush D Jamthikar
- Department of Electronics and Communications, Visvesvaraya National Institute of Technology, Nagpur 440010, Maharashtra, India
| | - Luca Saba
- Department of Radiology, University of Cagliari, Monserrato 09045, Cagliari, Italy
| | - Amer M Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Vasilios Kotsis
- 3rd Department of Internal Medicine, Hypertension Center, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki 541-24, Greece
| | - Narendra N Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110020, India
| | - Surinder K Dhanjil
- Stroke Diagnosis and Monitoring Division, AtheroPoint™ LLC, CA 95661, United States
| | - Misha Majhail
- Stroke Diagnosis and Monitoring Division, AtheroPoint™, Roseville, CA 95661, United States
| | - Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Vikas Agarwal
- Departments of Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - George D Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, United Kingdom
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, United Kingdom
| | - Aditya M Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Raghu Kolluri
- OhioHealth Heart and Vascular, Ohio, OH 43082, United States
| | - Subbaram Naidu
- Electrical Engineering Department, University of Minnesota, Duluth, MN 55812, United States
| | - Jasjit S Suri
- Stroke Diagnosis and Monitoring Division, AtheroPoint™, Roseville, CA 95661, United States
| |
Collapse
|
43
|
Lu Y, Zhu S, Wang X, Liu J, Li Y, Wang W, Wang S, Wang F. ShengMai-San Attenuates Cardiac Remodeling in Diabetic Rats by Inhibiting NOX-Mediated Oxidative Stress. Diabetes Metab Syndr Obes 2021; 14:647-657. [PMID: 33603429 PMCID: PMC7884944 DOI: 10.2147/dmso.s287582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/26/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE ShengMai-San (SMS) is traditionally used to treat ischemic cardiovascular and cerebrovascular diseases. Recently, several studies have reported the cardioprotective effects of SMS in diabetic animals. However, the potential mechanisms have not yet been fully elucidated. In this study, we investigated whether SMS exerts a beneficial effect in diabetic cardiomyopathy (DCM) by alleviating NADPH oxidase (NOX)-mediated oxidative stress. METHODS SD rats were randomly divided into a negative control group (NC), diabetes mellitus group (DM) and SMS-treated group (SMS). The myocardial structure alterations, apoptosis and biomarkers of oxidative stress were observed. Moreover, to explore the protective mechanism of SMS, the activation of AMPKα, expression and translocation of NOX-related proteins were assessed. RESULTS Diabetes led to excessive collagen content, fibrosis, and apoptosis in the myocardium. Oxidative stress in diabetic hearts was indicated by low levels of T-AOC, high levels of 8-iso-PGF2α and 8-OHdG, inactivation of AMPKα, elevated expression of NOX2 and NOX4 and translocation of NOX isoforms p47phox and p67phox. Treatment with SMS for 10 weeks resulted in the alleviation of diabetes-associated myocardial structure abnormalities and apoptosis. Additionally, SMS attenuated the accumulation of oxidative stress markers in myocardial tissue. Further investigation showed that SMS was able to reverse the levels of oxidative stress-associated proteins NOX2 and NOX4 in the DM rats. Moreover, SMS treatment blunted the translocation of NADPH oxidase isoforms p47phox and p67phox as well. Furthermore, SMS promoted the activation of AMPK in the cardiac tissue of diabetic rats. CONCLUSION These findings indicate that SMS exhibits therapeutic properties against diabetic cardiomyopathy by attenuating myocardial oxidative damage via activation of AMPKα and inhibition of NOX signaling.
Collapse
Affiliation(s)
- Yanting Lu
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Shu Zhu
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Xiaoyan Wang
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Juhai Liu
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Yingying Li
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Wei Wang
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Shijun Wang
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Furong Wang
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| |
Collapse
|
44
|
Li T, Yin Y, Mu N, Wang Y, Liu M, Chen M, Jiang W, Yu L, Li Y, Ma H. Metformin-Enhanced Cardiac AMP-Activated Protein Kinase/Atrogin-1 Pathways Inhibit Charged Multivesicular Body Protein 2B Accumulation in Ischemia-Reperfusion Injury. Front Cell Dev Biol 2021; 8:621509. [PMID: 33614629 PMCID: PMC7892907 DOI: 10.3389/fcell.2020.621509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Cardiac autophagic flux is impaired during myocardial ischemia/reperfusion (MI/R). Impaired autophagic flux may exacerbate MI/R injury. Charged multivesicular body protein 2B (CHMP2B) is a subunit of the endosomal sorting complex required for transport (ESCRT-III) complex that is required for autophagy. However, the reverse role of CHMP2B accumulation in autophagy and MI/R injury has not been established. The objective of this article is to elucidate the roles of AMP-activated protein kinase (AMPK)/atrogin-1 pathways in inhibiting CHMP2B accumulation in ischemia-reperfusion injury. Methods: Male C57BL/6 mice (3-4 months) and H9c2 cardiomyocytes were used to evaluate MI/R and hypoxia/reoxygenation (H/R) injury in vivo and in vitro, respectively. MI/R was built by a left lateral thoracotomy and occluded the left anterior descending artery. H9c2 cells were firstly treated in 95% N2 and 5% CO2 for 15 h and reoxygenation for 1 h. Metformin (100 mg/kg/d) and CHMP2B (Ad-CHMP2B) transfected adenoviruses were administered to the mice. The H9c2 cells were treated with metformin (2.5 mM), MG-132 (10 μM), bafilomycin A1 (10 nM), and compound C (20 μM). Results: Autophagic flux was found to be inhibited in H/R-treated cardiomyocytes and MI/R mice, with elevated cardiac CHMP2B accumulation. Upregulated CHMP2B levels in the in vivo and in vitro experiments were shown to inhibit autophagic flux leading to the deterioration of H/R-cardiomyocytes and MI/R injury. This finding implies that CHMP2B accumulation increases the risk of myocardial ischemia. Metformin suppressed CHMP2B accumulation and ameliorated H/R-induced autophagic dysfunction by activating AMPK. Activated AMPK upregulated the messenger RNA expression and protein levels of atrogin-1, a muscle-specific ubiquitin ligase, in the myocardium. Atrogin-1 significantly enhanced the interaction between atrogin-1 and CHMP2B, therefore, promoting CHMP2B degradation in the MI/R myocardium. Finally, this study revealed that metformin-inhibited CHMP2B accumulation induced autophagic impairment and ischemic susceptibility in vivo through the AMPK-regulated CHMP2B degradation by atrogin-1. Conclusion: Impaired CHMP2B clearance in vitro and in vivo inhibits autophagic flux and weakens the myocardial ischemic tolerance. Metformin treatment degrades CHMP2B through the AMPK-atrogin-1-dependent pathway to maintain the homeostasis of autophagic flux. This is a novel mechanism that enriches the understanding of cardioprotection.
Collapse
Affiliation(s)
- Tian Li
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Mai Chen
- Department of Cardiovascular Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Li
- Department of Cardiovascular Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
45
|
Xu L, Qiao Y, Zheng Q. Identification of an autophagy-related gene expression signature for colorectal cancer. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1872716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Lijun Xu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yuqi Qiao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qing Zheng
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
46
|
Silwal P, Paik S, Kim JK, Yoshimori T, Jo EK. Regulatory Mechanisms of Autophagy-Targeted Antimicrobial Therapeutics Against Mycobacterial Infection. Front Cell Infect Microbiol 2021; 11:633360. [PMID: 33828998 PMCID: PMC8019938 DOI: 10.3389/fcimb.2021.633360] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/08/2021] [Indexed: 01/25/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular pathogen causing human tuberculosis, an infectious disease that still remains as a global health problem. Autophagy, a lysosomal degradative process, has emerged as a critical pathway to restrict intracellular Mtb growth through enhancement of phagosomal maturation. Indeed, several autophagy-modulating agents show promise as host-directed therapeutics for Mtb infection. In this Review, we discuss recent progress in our understanding the molecular mechanisms underlying the action of autophagy-modulating agents to overcome the immune escape strategies mediated by Mtb. The factors and pathways that govern such mechanisms include adenosine 5'-monophosphate-activated protein kinase, Akt/mammalian TOR kinase, Wnt signaling, transcription factor EB, cathelicidins, inflammation, endoplasmic reticulum stress, and autophagy-related genes. A further understanding of these mechanisms will facilitate the development of host-directed therapies against tuberculosis as well as infections with other intracellular bacteria targeted by autophagic degradation.
Collapse
Affiliation(s)
- Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
- *Correspondence: Eun-Kyeong Jo,
| |
Collapse
|