1
|
Keller MA, Nakamura M. Ketone Catabolism is Essential for Maintaining Normal Heart Function During Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643760. [PMID: 40161834 PMCID: PMC11952331 DOI: 10.1101/2025.03.18.643760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The heart utilizes various nutrient sources for energy production, primarily favoring fatty acid oxidation. While ketones can be fuel substrates, ketolysis has been shown to be dispensable for heart development and function in mice. However, the long-term consequences of ketolysis downregulation in the heart remain unknown. Here we demonstrate that ketone catabolism is essential for preserving cardiac function during aging. The cardiac expression of succinyl-CoA:3-ketoacid CoA transferase (SCOT), a rate-limiting enzyme in ketolysis, decreases with aging in female mice. SCOT cardiomyocyte-specific knockout (cKO) mice exhibit normal heart function at 10 weeks of age but progressively develop cardiac dysfunction and remodeling as they age, without overt hypertrophy in both sexes. Notably, ketone supplementation via a ketogenic diet partially rescues contractile dysfunction in SCOT cKO mice, suggesting ketone oxidation-independent mechanisms contribute to the development of cardiomyopathy caused by SCOT downregulation. These findings indicate that ketone catabolism is crucial for maintaining heart function during aging, and that ketones confer cardioprotection independently of ketone oxidation.
Collapse
|
2
|
Yu L, Liu S, Liu J, Li J, Zhang W, Lin L, Yang L, Zheng G. Smilaxchina L. polyphenols inhibit LPS-induced macrophage M1 polarization to alleviate inflammation through NF-κB signaling pathway in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119355. [PMID: 39800244 DOI: 10.1016/j.jep.2025.119355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As an important component of the cell wall of Gram-negative bacteria, lipopolysaccharide (LPS) is an important inducer of inflammation in humans. Smilax china L. is known for its diverse bioactive functions, particularly its anti-inflammatory effects. AIM OF THE STUDY This study aimed to investigate the bioactive function of Smilax china L. polyphenols (SCLP) on LPS-induced inflammation. MATERIALS AND METHODS Inflammation in RAW264.7 macrophages and mice were induced using LPS. The cytotoxicity of SCLP was investigated by MTT assay. Inflammatory factors were detected by ELISA and RT-PCR. The expression of NF-κB pathway-related proteins was analyzed by Western Blotting. RESULTS The results demonstrated that SCLP significantly reduced the levels of pro-inflammatory factors (TNF-α, IL-1β, and IL-6) and inhibited M1 polarization of macrophages in both RAW264.7 macrophages and mice (p < 0.05). Western Blotting analysis revealed that the levels of NF-κB signaling pathway-associated proteins (p-p65, p-IKB, p-IKK) were significantly reduced (p < 0.05). Notably, SCLP significantly downregulated the expression of pro-apoptotic proteins, while upregulating the expression of anti-apoptotic proteins in RAW264.7 macrophages (p < 0.05). Additionally, the levels of antioxidant enzymes were enhanced in mice, suggesting a potential reduction in the inflammatory response. CONCLUSIONS These findings indicated that SCLP might inhibit LPS-induced M1 polarization through the NF-κB signaling pathway, thereby reducing inflammation. Consequently, SCLP might serve as a promising bioactive substance for preventing inflammation-related injury.
Collapse
Affiliation(s)
- Longhui Yu
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Shanshan Liu
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jiluan Liu
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jingen Li
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Wenkai Zhang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Lezhen Lin
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Licong Yang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
| | - Guodong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
3
|
Perez-Serna AA, Guzman-Llorens D, Dos Santos RS, Marroqui L. Bcl-2 and Bcl-xL in Diabetes: Contributions to Endocrine Pancreas Viability and Function. Biomedicines 2025; 13:223. [PMID: 39857806 PMCID: PMC11760435 DOI: 10.3390/biomedicines13010223] [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: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes is a chronic metabolic disorder whose prevalence increases every year, affecting more than 530 million adults worldwide. Type 1 (T1D) and type 2 diabetes (T2D), the most common forms of diabetes, are characterized by the loss of functional pancreatic β-cells, mostly due to apoptosis. B-cell leukemia/lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL), two anti-apoptotic proteins belonging to the Bcl-2 family, are crucial for regulating the intrinsic pathway of apoptosis. However, over the years, they have been implicated in many other cellular processes, including intracellular Ca2+ homeostasis and the regulation of mitochondrial metabolism. Thus, understanding the biological processes in which these proteins are involved may be crucial to designing new therapeutic targets. This review summarizes the roles of Bcl-2 and Bcl-xL in apoptosis and metabolic homeostasis. It focuses on how the dysregulation of Bcl-2 and Bcl-xL affects pancreatic β-cell function and survival, and the consequences for diabetes development.
Collapse
Affiliation(s)
- Atenea A. Perez-Serna
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Guzman-Llorens
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
| | - Reinaldo S. Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l’Almazara 11, 03203 Elche, Alicante, Spain
| | - Laura Marroqui
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Keller MA, Ivessa A, Liu T, Li H, Romanienko PJ, Nakamura M. KAT6A acetylation drives metabolic adaptation to mediate cellular growth and mitochondrial metabolism through AMPK interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633047. [PMID: 39829935 PMCID: PMC11741246 DOI: 10.1101/2025.01.14.633047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Diets influence metabolism and disease susceptibility, with lysine acetyltransferases (KATs) serving as key regulators through acetyl-CoA. We have previously demonstrated that a ketogenic diet alleviates cardiac pathology, though the underlying mechanisms remain largely unknown. Here we show that KAT6A acetylation is crucial for mitochondrial function and cell growth. Proteomic analysis revealed that KAT6A is acetylated at lysine (K)816 in the hearts of mice fed a ketogenic diet under hypertension, which enhances its interaction with AMPK regulatory subunits. RNA-sequencing analysis demonstrated that the KAT6A acetylation-mimetic mutant stimulates AMPK signaling in cardiomyocytes. Moreover, the acetylation-mimetic mutant mitigated phenylephrine-induced mitochondrial dysfunction and cardiomyocyte hypertrophy via AMPK activation. However, KAT6A-K816R acetylation-resistant knock-in mice unexpectedly exhibited smaller hearts with enhanced AMPK activity, conferring protection against neurohumoral stress-induced cardiac hypertrophy and remodeling. These findings indicate that KAT6A regulates metabolism and cellular growth by interacting with and modulating AMPK activity through K816-acetylation in a cell type-specific manner.
Collapse
|
5
|
Zhang T, Li L, Mo X, Xie S, Liu S, Zhao N, Zhang H, Chen S, Zeng X, Wang S, Deng W, Tang Q. Matairesinol blunts adverse cardiac remodeling and heart failure induced by pressure overload by regulating Prdx1 and PI3K/AKT/FOXO1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156054. [PMID: 39306883 DOI: 10.1016/j.phymed.2024.156054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Pathological cardiac remodeling is a critical process leading to heart failure, characterized primarily by inflammation and apoptosis. Matairesinol (Mat), a key chemical component of Podocarpus macrophyllus resin, exhibits a wide range of pharmacological activities, including anti-hydatid, antioxidant, antitumor, and anti-inflammatory effects. PURPOSE This study aims to investigate whether Matairesinol alleviate cardiac hypertrophy and remodeling caused by pressure overload and to elucidate its mechanism of action. METHODS An in vitro pressure loading model was established using neonatal rat cardiomyocytes treated with angiotensin Ⅱ, while an in vivo model was created using C57 mice subjected to transverse aortic constriction (TAC). To activate the PI3K/Akt/FoxO1 pathway, Ys-49 was employed. Moreover, small interfering RNA (siRNA) and short hairpin RNA (shRNA) were utilized to silence Prdx1 expression both in vitro and in vivo. Various techniques, including echocardiography, wheat germ agglutinin (WGA) staining, HE staining, PSR staining, and Masson trichrome staining, were used to assess cardiac function, cardiomyocyte cross-sectional area, and fibrosis levels in rats. Apoptosis in myocardial tissue and in vitro was detected by TUNEL assay, while reactive oxygen species (ROS) content in tissues and cells was measured using DHE staining. Furthermore, the affinity of Prdx1 with Mat and PI3K was analyzed using computer-simulated molecular docking. Western blotting and RT-PCR were utilized to evaluate Prdx1 levels and proteins related to apoptosis and oxidative stress, as well as the mRNA levels of cardiac hypertrophy and fibrosis-related indicators. RESULTS Mat significantly alleviated cardiac hypertrophy and fibrosis induced by TAC, preserved cardiac function, and markedly reduced cardiomyocyte apoptosis and oxidative damage. In vitro, mat attenuated ang Ⅱ - induced hypertrophy of nrvms and activation of neonatal rat fibroblasts. Notably, activation of the PI3K/Akt/FoxO1 pathway and downregulation of Prdx1 expression were observed in TAC mice; however, these effects were reversed by Mat treatment. Furthermore, Prdx1 knockdown activated the PI3K/Akt/FoxO1 pathway, leading to exacerbation of the disease. Molecular docking indicated that Molecular docking indicated that Mat upregulated Prdx1 expression by binding to it, thereby inhibiting the PI3K/Akt/FoxO1 pathway and protecting the heart by restoring Prdx1 expression levels. CONCLUSION Matairesinol alleviates pressure overload-induced cardiac remodeling both in vivo and in vitro by upregulating Prdx1 expression and inhibiting the PI3K/Akt/FoxO1 pathway. This study highlights the therapeutic potential of Matairesinol in the treatment of cardiac hypertrophy and remodeling, providing a promising avenue for future research and clinical application.
Collapse
Key Words
- ANP, atrial natriuretic peptide
- Abbreviations: MAT, matairesinol
- BNP, B-type natriuretic peptide
- Cardiac fibrosis
- Cardiac hypertrophy
- Cardiac remodeling
- LV, left ventricular
- LVEDd, left ventricular end-diastolic dimension
- LVEF, left ventricular ejection fraction
- Matairesinol
- NRCFS, neonatal rat cardiac fibroblasts
- PRDX 1
- PRDX1, peroxiredoxin 1
- ROS, reactive oxygen species
- Sh-RNA, short-hairpin RNA
- Si-RNA, small interfering RNA
- TAC, transverse aortic contraction
- β-MHC, Β-myosin heavy chain
Collapse
Affiliation(s)
- Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Xiaotong Mo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Shiqiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Xiaofeng Zeng
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Shasha Wang
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
6
|
Tang H, Kong Q, Zhang Z, Wu W, Yuan L, Liu X. Regulation of transcription factor function by purinergic signalling in cardiovascular diseases. Purinergic Signal 2024:10.1007/s11302-024-10045-8. [PMID: 39215950 DOI: 10.1007/s11302-024-10045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs), including hypertension, atherosclerosis, myocardial ischemia, and myocardial infarction, constitute the primary cause of mortality worldwide. Transcription factors play critical roles in the development of CVDs and contribute to the pathophysiology of these diseases by coordinating the transcription of many genes involved in inflammation, oxidative stress, angiogenesis, and glycolytic metabolism. One important regulator of hemostasis in both healthy and pathological settings has been identified as a purinergic signalling pathway. Research has demonstrated that several signalling networks implicated in the pathophysiology of CVDs are formed by transcription factors that are regulated by purinergic substances. Here, we briefly summarize the roles and mechanisms of the transcription factors regulated by purinergic pathways in various types of CVD. This information will be essential for discovering novel approaches for CVD treatment and prevention.
Collapse
Affiliation(s)
- Hao Tang
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qihang Kong
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhewei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenchao Wu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Lixing Yuan
- Public Laboratory of West China Second University Hospital and Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, 610041, China.
| | - Xiaojing Liu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Shi X, Dang X, Huang Z, Lu Y, Tong H, Liang F, Zhuang F, Li Y, Cai Z, Huo H, Jiang Z, Pan C, Wang X, Gu C, He B. SUMOylation of TEAD1 Modulates the Mechanism of Pathological Cardiac Hypertrophy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305677. [PMID: 38225750 PMCID: PMC10966521 DOI: 10.1002/advs.202305677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/14/2023] [Indexed: 01/17/2024]
Abstract
Pathological cardiac hypertrophy is the leading cause of heart failure and has an extremely complicated pathogenesis. TEA domain transcription factor 1 (TEAD1) is recognized as an important transcription factor that plays a key regulatory role in cardiovascular disease. This study aimed to explore the role of TEAD1 in cardiac hypertrophy and to clarify the regulatory role of small ubiquitin-like modifier (SUMO)-mediated modifications. First, the expression level of TEAD1 in patients with heart failure, mice, and cardiomyocytes is investigated. It is discovered that TEAD1 is modified by SUMO1 during cardiac hypertrophy and that the process of deSUMOylation is regulated by SUMO-specific protease 1 (SENP1). Lysine 173 is an essential site for TEAD1 SUMOylation, which affects the protein stability, nuclear localization, and DNA-binding ability of TEAD1 and enhances the interaction between TEAD1 and its transcriptional co-activator yes-associated protein 1 in the Hippo pathway. Finally, adeno-associated virus serotype 9 is used to construct TEAD1 wild-type and KR mutant mice and demonstrated that the deSUMOylation of TEAD1 markedly exacerbated cardiomyocyte enlargement in vitro and in a mouse model of cardiac hypertrophy. The results provide novel evidence that the SUMOylation of TEAD1 is a promising therapeutic strategy for hypertrophy-related heart failure.
Collapse
Affiliation(s)
- Xin Shi
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Xuening Dang
- Department of Cardiovascular SurgeryShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhenyu Huang
- Department of Central LaboratoryShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Yanqiao Lu
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Huan Tong
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Feng Liang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Fei Zhuang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Yi Li
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhaohua Cai
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Huanhuan Huo
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhaolei Jiang
- Department of Cardiothoracic SurgeryXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200030China
| | - Changqing Pan
- General Surgery DepartmentShanghai Chest HospitalSchool of Medicine Shanghai Jiao Tong UniversityShanghai200030China
| | - Xia Wang
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Chang Gu
- Department of Cardiothoracic SurgeryXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200030China
- Department of Thoracic SurgeryShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Ben He
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| |
Collapse
|