1
|
Rojas-Solé C, Pinilla-González V, Lillo-Moya J, González-Fernández T, Saso L, Rodrigo R. Integrated approach to reducing polypharmacy in older people: exploring the role of oxidative stress and antioxidant potential therapy. Redox Rep 2024; 29:2289740. [PMID: 38108325 PMCID: PMC10732214 DOI: 10.1080/13510002.2023.2289740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Increased life expectancy, attributed to improved access to healthcare and drug development, has led to an increase in multimorbidity, a key contributor to polypharmacy. Polypharmacy is characterised by its association with a variety of adverse events in the older persons. The mechanisms involved in the development of age-related chronic diseases are largely unknown; however, altered redox homeostasis due to ageing is one of the main theories. In this context, the present review explores the development and interaction between different age-related diseases, mainly linked by oxidative stress. In addition, drug interactions in the treatment of various diseases are described, emphasising that the holistic management of older people and their pathologies should prevail over the individual treatment of each condition.
Collapse
Affiliation(s)
- Catalina Rojas-Solé
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Víctor Pinilla-González
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - José Lillo-Moya
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Tommy González-Fernández
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, Rome, Italy
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
2
|
Rouholamini FS, Aminaei M, Aminizadeh S. The effect of eight weeks of endurance training and MitoQ supplementation on antioxidant capacity and the expression of sestrin-2 and AMPK in cardiac tissue of aged rats. Exp Gerontol 2024; 196:112572. [PMID: 39233194 DOI: 10.1016/j.exger.2024.112572] [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: 05/12/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE The present study aimed to investigate the effects of endurance training (ET) in combination with MitoQ supplementation on antioxidant indices and the expression of sesterin-2 (SESN2) as an anti-aging factor and AMPK as an energy sensor in aged male Wistar rats. METHODS Twenty-eight aged Wistar rats (410 ± 15 g, 22 ± 1.5 months old) were randomly divided into four groups (n = 7): Control, ET (eight weeks endurance training on the treadmill), MitoQ (250 μ/L in drinking water), and ET + MitoQ. We measured the protein and gene expression of SESN2 and AMPK in the heart tissue by western blotting and real-time PCR, respectively. In addition, antioxidant indices, superoxide dismutase (SOD), and glutathione peroxidase (GPx) activity, and oxidant malondialdehyde (MDA) concentration in the cardiac tissue and serum were measured. RESULTS SESN2 and AMPK protein expression significantly increased in the MitoQ group compared to the control group (P = 0.002, P = 0.0003). MDA content in tissue and serum remained unchanged in all groups (P > 0.05). MitoQ supplementation significantly increased SOD and GPx enzyme activity in serum and cardiac tissue (P = 0.001). CONCLUSION Overall, ET and MitoQ alone and in combination have anti-aging effects and improve the expression of AMPK and SESN2. Additionally, ET and MitoQ lead to improved antioxidant capacity in aged rats by ameliorating the activity of antioxidant enzymes.
Collapse
Affiliation(s)
- Fatemeh Sadat Rouholamini
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohsen Aminaei
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Soheil Aminizadeh
- Physiology Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
3
|
Damanti S, Senini E, De Lorenzo R, Merolla A, Santoro S, Festorazzi C, Messina M, Vitali G, Sciorati C, Rovere-Querini P. Acute Sarcopenia: Mechanisms and Management. Nutrients 2024; 16:3428. [PMID: 39458423 PMCID: PMC11510680 DOI: 10.3390/nu16203428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Acute sarcopenia refers to the swift decline in muscle function and mass following acute events such as illness, surgery, trauma, or burns that presents significant challenges in hospitalized older adults. METHODS narrative review to describe the mechanisms and management of acute sarcopenia. RESULTS The prevalence of acute sarcopenia ranges from 28% to 69%, likely underdiagnosed due to the absence of muscle mass and function assessments in most clinical settings. Systemic inflammation, immune-endocrine dysregulation, and anabolic resistance are identified as key pathophysiological factors. Interventions include early mobilization, resistance exercise, neuromuscular electrical stimulation, and nutritional strategies such as protein supplementation, leucine, β-hydroxy-β-methyl-butyrate, omega-3 fatty acids, and creatine monohydrate. Pharmaceuticals show variable efficacy. CONCLUSIONS Future research should prioritize serial monitoring of muscle parameters, identification of predictive biomarkers, and the involvement of multidisciplinary teams from hospital admission to address sarcopenia. Early and targeted interventions are crucial to improve outcomes and prevent long-term disability associated with acute sarcopenia.
Collapse
Affiliation(s)
- Sarah Damanti
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Eleonora Senini
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Rebecca De Lorenzo
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Aurora Merolla
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Simona Santoro
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Costanza Festorazzi
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Marco Messina
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Giordano Vitali
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
| | - Clara Sciorati
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| | - Patrizia Rovere-Querini
- Internal Medicine Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.D.); (G.V.); (P.R.-Q.)
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, 20100 Milan, Italy; (E.S.); (R.D.L.); (A.M.); (S.S.); (C.F.); (M.M.)
| |
Collapse
|
4
|
Leote AC, Lopes F, Beyer A. Loss of coordination between basic cellular processes in human aging. NATURE AGING 2024; 4:1432-1445. [PMID: 39227753 PMCID: PMC11485205 DOI: 10.1038/s43587-024-00696-y] [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: 05/18/2023] [Accepted: 07/30/2024] [Indexed: 09/05/2024]
Abstract
Age-related loss of gene expression coordination has been reported for distinct cell types and may lead to impaired cellular function. Here we propose a method for quantifying age-related changes in transcriptional regulatory relationships between genes, based on a model learned from external data. We used this method to uncover age-related trends in gene-gene relationships across eight human tissues, which demonstrates that reduced co-expression may also result from coordinated transcriptional responses. Our analyses reveal similar numbers of strengthening and weakening gene-gene relationships with age, impacting both tissue-specific (for example, coagulation in blood) and ubiquitous biological functions. Regulatory relationships becoming weaker with age were established mostly between genes operating in distinct cellular processes. As opposed to that, regulatory relationships becoming stronger with age were established both within and between different cellular functions. Our work reveals that, although most transcriptional regulatory gene-gene relationships are maintained during aging, those with declining regulatory coupling result mostly from a loss of coordination between distinct cellular processes.
Collapse
Affiliation(s)
- Ana Carolina Leote
- Cologne Excellence Cluster on Cellular Stress Responses in Age-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Francisco Lopes
- Cologne Excellence Cluster on Cellular Stress Responses in Age-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department II of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Andreas Beyer
- Cologne Excellence Cluster on Cellular Stress Responses in Age-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
5
|
Costa Pereira JPD, Rüegg RAB, Costa EC, Fayh APT. Muscle quality and major adverse cardiovascular events in post-acute myocardial infarction: A prospective cohort study. Nutr Metab Cardiovasc Dis 2024; 34:2266-2272. [PMID: 38866608 DOI: 10.1016/j.numecd.2024.04.017] [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: 02/20/2024] [Revised: 03/23/2024] [Accepted: 04/24/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND & AIMS Functional muscle quality, as assessed through the muscle quality index (MQI), represents a contemporary method to measure the capacity to generate force. Despite its potential, the prognostic significance of MQI remains uncertain in various clinical conditions, particularly among patients following acute myocardial infarction (AMI). In light of this, our study sought to evaluate the prognostic relevance of MQI concerning major adverse cardiovascular events (MACE) in patients following AMI. METHODS AND RESULTS This is a secondary analysis of a prospective cohort study that included subjects aged ≥20 years from a Cardiovascular Unit Hospital. Functional muscle quality was estimated using MQI, defined as the ratio of handgrip strength (HGS) to muscle mass (MM) derived from bioelectrical impedance analysis. The outcomes included prolonged length of hospital stay, new adverse cardiovascular events (AMI, stroke and hospital readmission for unstable angina), and cardiovascular mortality. A composite score comprising all adverse events over the 1-year follow-up was calculated and defined as MACE. This study included 163 patients, with a median age of 61 years (IQ: 54-69 years), and the majority consisted of males (76.1%). Individual components of the functional muscle quality (HGS and MM) were not associated with any of the adverse outcomes. Only MQI was associated mortality over the 1-year follow-up. For each increase in MQI, the hazard of mortality decreases: adjusted HR: 0.08 (95% CI 0.01-0.84). CONCLUSION Functional muscle quality assessed by the MQI may be a valuable clinical predictor of 1-year cardiovascular mortality in patients hospitalized post-AMI.
Collapse
Affiliation(s)
- Jarson Pedro da Costa Pereira
- Postgraduate Program in Nutrition and Public Health, Department of Nutrition, Federal University of Pernambuco, Recife, PE, Brazil
| | - Rodrigo Albert Baracho Rüegg
- Postgraduate Program in Health Science, Health Science Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Eduardo Caldas Costa
- ExCE Research Group, Department of Physical Education, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ana Paula Trussardi Fayh
- Postgraduate Program in Health Science, Health Science Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil; PesqClin Lab, Onofre Lopes University Hospital, Brazilian Company of Hospital Services (EBSERH), Federal University of Rio Grande do Norte, Natal, Brazil.
| |
Collapse
|
6
|
Chen S, He T, Chen J, Wen D, Wang C, Huang W, Yang Z, Yang M, Li M, Huang S, Huang Z, Zhu H. Betaine delays age-related muscle loss by mitigating Mss51-induced impairment in mitochondrial respiration via Yin Yang1. J Cachexia Sarcopenia Muscle 2024; 15:2104-2117. [PMID: 39187977 PMCID: PMC11446699 DOI: 10.1002/jcsm.13558] [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: 10/11/2023] [Revised: 05/18/2024] [Accepted: 07/05/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction is one of the hallmarks of aging and a leading contributor to sarcopenia. Nutrients are essential for improving mitochondrial function and skeletal muscle health during the aging process. Betaine is a nutrient with potential muscle-preserving properties. However, whether and how betaine could regulate the mitochondria function in aging muscle are poorly understood. We aimed to explore the molecular target and underlying mechanism of betaine in attenuating the age-related mitochondrial dysfunction in skeletal muscle. METHODS Young mice (YOU, 2 months), old mice (OLD, 15 months), and old mice with betaine treatment (BET, 15 months) were fed for 12 weeks. The effects of betaine on muscle mass, strength, function, and subcellular structure of muscle fibres were assessed. RNA sequencing (RNA-seq) was conducted to identify the molecular target of betaine. The impacts of betaine on mitochondrial-related molecules, superoxide accumulation, and oxidative respiration were examined using western blotting (WB), immunofluorescence (IF) and seahorse assay. The underlying mechanism of betaine regulation on the molecular target to maintain mitochondrial function was investigated by luciferase reporter assay, chromatin immunoprecipitation and electrophoretic mobility shift assay. Adenoassociated virus transfection, succinate dehydrogenase staining (SDH), and energy expenditure assessment were performed on 20-month-old mice for validating the mechanism in vivo. RESULTS Betaine intervention demonstrated anti-aging effects on the muscle mass (P = 0.017), strength (P = 0.010), and running distance (P = 0.013). Mitochondrial-related markers (ATP5a, Sdha, and Uqcrc2) were 1.1- to 1.5-fold higher in BET than OLD (all P ≤ 0.036) with less wasted mitochondrial vacuoles accumulating in sarcomere. Bioinformatic analysis from RNA-seq displayed pathways related to mitochondrial respiration activity was higher enriched in BET group (NES = -0.87, FDR = 0.10). The quantitative real time PCR (qRT-PCR) revealed betaine significantly reduced the expression of a novel mitochondrial regulator, Mss51 (-24.9%, P = 0.002). In C2C12 cells, betaine restored the Mss51-mediated suppression in mitochondrial respiration proteins (all P ≤ 0.041), attenuated oxygen consumption impairment, and superoxide accumulation (by 20.7%, P = 0.001). Mechanically, betaine attenuated aging-induced repression in Yy1 mRNA expression (BET vs. OLD: 2.06 vs. 1.02, P = 0.009). Yy1 transcriptionally suppressed Mss51 mRNA expression both in vitro and in vivo. This contributed to the preservation of mitochondrial respiration, improvement for energy expenditure (P = 0.008), and delay of muscle loss during aging process. CONCLUSIONS Altogether, betaine transcriptionally represses Mss51 via Yy1, improving age-related mitochondrial respiration in skeletal muscle. These findings suggest betaine holds promise as a dietary supplement to delay skeletal muscle degeneration and improve age-related mitochondrial diseases.
Collapse
Affiliation(s)
- Si Chen
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Tongtong He
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Jiedong Chen
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Dongsheng Wen
- Department of Hepatobiliary Oncology, State Key Laboratory of Oncology in South China, Sun Yat‐sen University Cancer CenterSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Chen Wang
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Wenge Huang
- Center of Experimental AnimalsSun Yat‐sen UniversityGuangzhouChina
| | - Zhijun Yang
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Mengtao Yang
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Mengchu Li
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Siyu Huang
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Zihui Huang
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| | - Huilian Zhu
- Department of Nutrition, School of Public HealthSun Yat‐sen UniversityGuangzhouChina
- School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and HealthSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
7
|
Lee JY, Shin SK, Han JW, Kwon EY, Bae HR. Cross-Species Studies Reveal That Dysregulated Mitochondrial Gene Expression and Electron Transport Complex I Activity Are Crucial for Sarcopenia. Int J Mol Sci 2024; 25:10302. [PMID: 39408631 PMCID: PMC11477305 DOI: 10.3390/ijms251910302] [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: 08/20/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
The significance of complex I of the electron transport chain (ETC) in the aging process is widely acknowledged; however, its specific impact on the development of sarcopenia in muscle remains poorly understood. This study elucidated the correlation between complex I inhibition and sarcopenia by conducting a comparative analysis of skeletal muscle gene expression in sarcopenia phenotypes from rats, mice, and humans. Our findings reveal a common mechanistic link across species, particularly highlighting the correlation between the suppression of complex I of ETC activity and dysregulated mitochondrial transcription and translation in sarcopenia phenotypes. Additionally, we observed macrophage dysfunction alongside abnormal metabolic processes within skeletal muscle tissues across all species, implicating their pathogenic role in the onset of sarcopenia. These discoveries underscore the importance of understanding the shared mechanisms associated with complex I of ETC in sarcopenia development. The identified correlations provide valuable insights into potential targets for therapeutic interventions aimed at mitigating the impact of sarcopenia, a condition with substantial implications for aging populations.
Collapse
Affiliation(s)
- Ji-Yoon Lee
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea (S.-K.S.)
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea (S.-K.S.)
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea
| | - Ji-Won Han
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea (S.-K.S.)
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea (S.-K.S.)
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea
- Center for Beautiful Aging, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea
| | - Heekyong R. Bae
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea (S.-K.S.)
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu 41566, Republic of Korea
| |
Collapse
|
8
|
Sahu Y, Jamadade P, Ch Maharana K, Singh S. Role of mitochondrial homeostasis in D-galactose-induced cardiovascular ageing from bench to bedside. Mitochondrion 2024; 78:101923. [PMID: 38925493 DOI: 10.1016/j.mito.2024.101923] [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: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Ageing is an inevitable phenomenon which affects the cellular to the organism level in the progression of the time. Oxidative stress and inflammation are now widely regarded as the key processes involved in the aging process, which may then cause significant harm to mitochondrial DNA, leading to apoptosis. Normal circulatory function is a significant predictor of disease-free life expectancy. Indeed, disorders affecting the cardiovascular system, which are becoming more common, are the primary cause of worldwide morbidity, disability, and mortality. Cardiovascular aging may precede or possibly underpin overall, age-related health decline. Numerous studies have foundmitochondrial mechanistc approachplays a vital role in the in the onset and development of aging. The D-galactose (D-gal)-induced aging model is well recognized and commonly used in the aging study. In this review we redeposit the association of the previous and current studies on mitochondrial homeostasis and its underlying mechanisms in D-galactose cardiovascular ageing. Further we focus the novel and the treatment strategies to combat the major complication leading to the cardiovascular ageing.
Collapse
Affiliation(s)
- Yogita Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Pratiksha Jamadade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Krushna Ch Maharana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India.
| |
Collapse
|
9
|
Chang ES, Song K, Song JY, Sung M, Lee MS, Oh JH, Kim JY, Park YH, Jung K, Choi YL. Real-time assessment of relative mitochondrial ATP synthesis response against inhibiting and stimulating substrates (MitoRAISE). Cancer Metab 2024; 12:25. [PMID: 39210390 PMCID: PMC11363686 DOI: 10.1186/s40170-024-00353-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Mitochondria are known to synthesize adenosine triphosphate (ATP) through oxidative phosphorylation. Understanding and accurately measuring mitochondrial ATP synthesis rate can provide insights into the functional status of mitochondria and how it contributes to overall cellular energy homeostasis. Traditional methods only estimate mitochondrial function by measuring ATP levels at a single point in time or through oxygen consumption rates. This study introduced the relative mitochondrial ATP synthesis response against inhibiting and stimulating substrates (MitoRAISE), designed to detect real-time changes in ATP levels as the cells respond to substrates. METHODS The sensitivity and specificity of the MitoRAISE assay were verified under various conditions, including the isolation of mitochondria, variations in cell numbers, cells exhibiting mitochondrial damage, and heterogeneous mixtures. Using peripheral blood mononuclear cells (PBMCs), we analyzed MitoRAISE data from 19 patients with breast cancer and 23 healthy women. RESULTS The parameters observed in the MitoRAISE data increased depending on the quantity of isolated mitochondria and cell count, whereas it remained unmeasured in mitochondrial-damaged cell lines. Basal ATP, rotenone response, malonate response, and mitochondrial DNA copy numbers were lower in PBMCs from patients with breast cancer than in those from healthy women. CONCLUSIONS The MitoRAISE assay has demonstrated its sensitivity and specificity by measuring relative ATP synthesis rates under various conditions. We propose MitoRAISE assay as a potential tool for monitoring changes in the mitochondrial metabolic status associated with various diseases.
Collapse
Affiliation(s)
- Eun Sol Chang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea
| | - Kyoung Song
- College of Pharmacy, Duksung Women's University, Seoul, South Korea
| | - Ji-Young Song
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea
| | - Minjung Sung
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea
| | - Mi-Sook Lee
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea
| | - Jung Han Oh
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea
| | - Ji-Yeon Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yeon Hee Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyungsoo Jung
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea.
| | - Yoon-La Choi
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea.
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-Ro 81, Gangnam-Go, Seoul, 06351, South Korea.
| |
Collapse
|
10
|
Zhai X, Wu W, Zeng S, Miao Y. Advance in the mechanism and clinical research of myalgia in long COVID. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:142-164. [PMID: 39310121 PMCID: PMC11411160 DOI: 10.62347/txvo6284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/18/2024] [Indexed: 09/25/2024]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, mortality rates of coronavirus disease 2019 (COVID-19) have significantly decreased. However, a variable proportion of patients exhibit persistent prolonged symptoms of COVID-19 infection (long COVID). This virus primarily attacks respiratory system, but numerous individuals complain persistent skeletal muscle pain or worsening pre-existing muscle pain post COVID-19, which severely affects the quality of life and recovery. Currently, there is limited research on the skeletal muscle pain in long COVID. In this brief review, we review potential pathological mechanisms of skeletal muscle pain in long COVID, and summarize the various auxiliary examinations and treatments for skeletal muscle pain in long COVID. We consider abnormal activation of inflammatory response, myopathy, and neurological damages as pivotal pathological mechanisms of skeletal muscle pain in long COVID. A comprehensive examination is significantly important in order to work out effective treatment plans and relieve skeletal muscle pain. So far, rehabilitation interventions for myalgia in long COVID contain but are not limited to drug, nutraceutical therapy, gut microbiome-targeted therapy, interventional therapy and strength training. Our study provides a potential mechanism reference for clinical researches, highlighting the importance of comprehensive approach and management of skeletal muscle pain in long COVID. The relief of skeletal muscle pain will accelerate rehabilitation process, improve activities of daily living and enhance the quality of life, promoting individuals return to society with profound significance.
Collapse
Affiliation(s)
- Xiuyun Zhai
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
| | - Weijun Wu
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
| | - Siliang Zeng
- Department of Rehabilitation Therapy, School of Health, Shanghai Normal University Tianhua CollegeNo. 1661, North Shengxin Road, Shanghai 201815, China
| | - Yun Miao
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
- Department of Rehabilitation, School of International Medical Technology, Shanghai Sanda UniversityNo. 2727, Jinhai Road, Shanghai 201209, China
| |
Collapse
|
11
|
Sharma AR, Chatterjee S, Lee YH, Lee SS. Targeting Crosstalk of Signaling Pathways among Muscles-Bone-Adipose Tissue: A Promising Therapeutic Approach for Sarcopenia. Aging Dis 2024; 15:1619-1645. [PMID: 37815907 PMCID: PMC11272187 DOI: 10.14336/ad.2023.00903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/03/2023] [Indexed: 10/12/2023] Open
Abstract
The aging process is associated with the development of a wide range of degenerative disorders in mammals. These diseases are characterized by a progressive decline in function at multiple levels, including the molecular, cellular, tissue, and organismal. Furthermore, it is responsible for various healthcare costs in developing and developed countries. Sarcopenia is the deterioration in the quality and functionality of muscles, which is extremely concerning as it manages many functions in the human body. This article reviews the molecular crosstalk involved in sarcopenia and the specific roles of many mediator molecules in establishing cross-talk between muscles, bone, and fatty tissues, eventually leading to sarcopenia. Besides, the involvement of various etiological factors, such as neurology, endocrinology, lifestyle, etc., makes it exceedingly difficult for clinicians to develop a coherent hypothesis that may lead to the well-organized management system required to battle this debilitating disease. The several hallmarks contributing to the progression of the disease is a vital question that needs to be addressed to ensure an efficient treatment for sarcopenia patients. Also, the intricate molecular mechanism involved in developing this disease requires more studies. The direct relationship of cellular senescence with aging is one of the pivotal issues contributing to disease pathophysiology. Some patented treatment strategies have been discussed, including drugs undergoing clinical trials and emerging options like miRNA and protein-enclosed extracellular vesicles. A clear understanding of the secretome, including the signaling pathways involved between muscles, bone, and fatty tissues, is extremely beneficial for developing novel therapeutics for curing sarcopenia.
Collapse
Affiliation(s)
| | | | | | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| |
Collapse
|
12
|
Tang Y, Liu Z, Chen Q, Juaiti M, Yu Z, Liang B, Zha L. Association of sarcopenia with the long-term risk of atrial fibrillation: A prospective cohort study. Aging Cell 2024; 23:e14198. [PMID: 38739369 PMCID: PMC11320353 DOI: 10.1111/acel.14198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024] Open
Abstract
The relationship between sarcopenia and the long-term risk of atrial fibrillation (AF) remains unclear. This study recruited a large prospective Caucasian cohort from the UK Biobank. Participants were assessed at baseline with handgrip strength and muscle mass and were categorized into groups of non-sarcopenia, probable sarcopenia, and confirmed sarcopenia. Kaplan-Meier method and Cox proportional hazards model were used to explore the association between sarcopenia and the incidence of AF. The genetic predisposition of AF was assessed by polygenic risk score. Sensitivity analyses were performed to validate the results. A total of 384,433 participants with a median age of 58 years and 54.3% women were enrolled in this study. There were 24,007 cases of new-onset AF over a median follow-up of 12.56 years. The groups of non-sarcopenia, probable sarcopenia, and confirmed sarcopenia accounted for 22,290 (6.1%), 1665 (9.2%), and 52 (11.9%) cases, respectively. Compared with the non-sarcopenia group, participants with probable sarcopenia or confirmed sarcopenia had an 8% (95% CI, 1.03-1.14) or 61% (95% CI, 1.23-2.12) higher risk of AF incidence. The findings remained robust in multiple sensitivity analyses, such as subgroup analysis and further adjustment of genetic predisposition. Notably, the association between sarcopenia and a high AF risk was more pronounced in younger participants, women, and those with valvular heart disease. In conclusion, sarcopenia was associated with a high long-term risk of AF in Caucasians, supporting sarcopenia as a new independent risk factor of AF.
Collapse
Affiliation(s)
- Yiyang Tang
- Department of CardiologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Zhenghui Liu
- Department of NeurologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Qin Chen
- Department of CardiologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Mukamengjiang Juaiti
- Department of CardiologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Zaixin Yu
- Department of CardiologyXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Benhui Liang
- Department of CardiologyXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Lihuang Zha
- Department of CardiologyXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
13
|
Wang Y, Yang JS, Zhao M, Chen JQ, Xie HX, Yu HY, Liu NH, Yi ZJ, Liang HL, Xing L, Jiang HL. Mitochondrial endogenous substance transport-inspired nanomaterials for mitochondria-targeted gene delivery. Adv Drug Deliv Rev 2024; 211:115355. [PMID: 38849004 DOI: 10.1016/j.addr.2024.115355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Mitochondrial genome (mtDNA) independent of nuclear gene is a set of double-stranded circular DNA that encodes 13 proteins, 2 ribosomal RNAs and 22 mitochondrial transfer RNAs, all of which play vital roles in functions as well as behaviors of mitochondria. Mutations in mtDNA result in various mitochondrial disorders without available cures. However, the manipulation of mtDNA via the mitochondria-targeted gene delivery faces formidable barriers, particularly owing to the mitochondrial double membrane. Given the fact that there are various transport channels on the mitochondrial membrane used to transfer a variety of endogenous substances to maintain the normal functions of mitochondria, mitochondrial endogenous substance transport-inspired nanomaterials have been proposed for mitochondria-targeted gene delivery. In this review, we summarize mitochondria-targeted gene delivery systems based on different mitochondrial endogenous substance transport pathways. These are categorized into mitochondrial steroid hormones import pathways-inspired nanomaterials, protein import pathways-inspired nanomaterials and other mitochondria-targeted gene delivery nanomaterials. We also review the applications and challenges involved in current mitochondrial gene editing systems. This review delves into the approaches of mitochondria-targeted gene delivery, providing details on the design of mitochondria-targeted delivery systems and the limitations regarding the various technologies. Despite the progress in this field is currently slow, the ongoing exploration of mitochondrial endogenous substance transport and mitochondrial biological phenomena may act as a crucial breakthrough in the targeted delivery of gene into mitochondria and even the manipulation of mtDNA.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Song Yang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Min Zhao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Qi Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hai-Xin Xie
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hao-Yuan Yu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Na-Hui Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zi-Juan Yi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hui-Lin Liang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
14
|
Noguchi I, Maeda H, Kobayashi K, Nagasaki T, Kato H, Yanagisawa H, Wada N, Kanazawa G, Kaji T, Sakai H, Fujimaki S, Ono Y, Taguchi K, Chuang VTG, Saruwatari J, Otagiri M, Watanabe H, Maruyama T. Carbon monoxide-loaded cell therapy as an exercise mimetic for sarcopenia treatment. Free Radic Biol Med 2024; 220:67-77. [PMID: 38657755 DOI: 10.1016/j.freeradbiomed.2024.04.231] [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: 03/04/2024] [Revised: 04/17/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
Sarcopenia is characterized by loss of muscle strength and muscle mass with aging. The growing number of sarcopenia patients as a result of the aging population has no viable treatment. Exercise maintains muscle strength and mass by increasing peroxisome growth factor activating receptor γ-conjugating factor-1α (PGC-1α) and Akt signaling in skeletal muscle. The present study focused on the carbon monoxide (CO), endogenous activator of PGC-1α and Akt, and investigated the therapeutic potential of CO-loaded red blood cells (CO-RBCs), which is bioinspired from in vivo CO delivery system, as an exercise mimetic for the treatment of sarcopenia. Treatment of C2C12 myoblasts with the CO-donor increased the protein levels of PGC-1α which enhanced mitochondrial biogenesis and energy production. The CO-donor treatment also activated Akt, indicating that CO promotes muscle synthesis. CO levels were significantly elevated in the skeletal muscle of normal mice after intravenous administration of CO-RBCs. Furthermore, CO-RBCs restored the mRNA expression levels of PGC-1α in the skeletal muscle of two experimental sarcopenia mouse models, denervated (Den) and hindlimb unloading (HU) models. CO-RBCs also restored muscle mass in Den mice by activating Akt signaling and suppressing the muscle atrophy factors myostatin and atrogin-1, and oxidative stress. Treadmill tests further showed that the reduced running distance in HU mice was significantly restored by CO-RBC administration. These findings suggest that CO-RBCs have potential as an exercise mimetic for sarcopenia treatment.
Collapse
Affiliation(s)
- Isamu Noguchi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Kazuki Kobayashi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Taisei Nagasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Hiromasa Kato
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Hiroki Yanagisawa
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Naoki Wada
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Gai Kanazawa
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Tsubasa Kaji
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Hiromi Sakai
- Department of Chemistry, Nara Medical University, Nara, Japan.
| | - Shin Fujimaki
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan.
| | - Victor Tuan Giam Chuang
- Pharmacy Discipline, Curtin Medical School, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, 6845, Western Australia, Australia.
| | - Junji Saruwatari
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences and DDS Research Institute, Sojo University, Kumamoto, Japan.
| | - Hiroshi Watanabe
- Department of Clinical Pharmacy and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
15
|
Wang Q, Lan X, Ke H, Xu S, Huang C, Wang J, Wang X, Huang T, Wu X, Chen M, Guo Y, Zeng L, Tian XL, Xiang Y. Histone β-hydroxybutyrylation is critical in reversal of sarcopenia. Aging Cell 2024:e14284. [PMID: 39076122 DOI: 10.1111/acel.14284] [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: 03/25/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
Sarcopenia, a leading cause for global disability and mortality, is an age-related muscular disorder, characterized by accelerated muscle mass loss and functional decline. It is known that caloric restriction (CR), ketogenic diet or endurance exercise lessen sarcopenia and elevate circulating β-hydroxybutyrate (β-HB) levels. Whether the elevated β-HB is essential to the reversal of sarcopenia, however, remains unclear. Here we show in both Caenorhabditis elegans and mouse models that an increase of β-HB reverse myofiber atrophy and improves motor functions at advanced ages. β-HB-induced histone lysine β-hydroxybutyrylation (Kbhb) is indispensable for the reversal of sarcopenia. Histone Kbhb enhances transcription of genes associated with mitochondrial pathways, including oxidative phosphorylation, ATP metabolic process and aerobic respiration. This ultimately leads to improve mitochondrial integrity and enhance mitochondrial respiration. The histone Kbhb are validated in mouse model with CR. Thus, we demonstrate that β-HB induces histone Kbhb, increases mitochondrial function, and reverses sarcopenia.
Collapse
Affiliation(s)
- Qiquan Wang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Xinqiang Lan
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Hao Ke
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Siman Xu
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Chunping Huang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Jiali Wang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Xiang Wang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Tiane Huang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Xia Wu
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Mengxin Chen
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Yingqi Guo
- Institutional Center for Shared Technologies and Facilities of the Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Lin Zeng
- Institutional Center for Shared Technologies and Facilities of the Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| | - Yang Xiang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Aging and Diseases, Nanchang, China
| |
Collapse
|
16
|
Traa A, Tamez González AA, Van Raamsdonk JM. Developmental disruption of the mitochondrial fission gene drp-1 extends the longevity of daf-2 insulin/IGF-1 receptor mutant. GeroScience 2024:10.1007/s11357-024-01276-z. [PMID: 39028454 DOI: 10.1007/s11357-024-01276-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
The dynamic nature of the mitochondrial network is regulated by mitochondrial fission and fusion, allowing for re-organization of mitochondria to adapt to the cell's ever-changing needs. As organisms age, mitochondrial fission and fusion become dysregulated and mitochondrial networks become increasingly fragmented. Modulation of mitochondrial dynamics has been shown to affect longevity in fungi, yeast, Drosophila and C. elegans. Disruption of the mitochondrial fission gene drp-1 drastically increases the already long lifespan of daf-2 insulin/IGF-1 signaling (IIS) mutants. In this work, we determined the conditions required for drp-1 disruption to extend daf-2 longevity and explored the molecular mechanisms involved. We found that knockdown of drp-1 during development is sufficient to extend daf-2 lifespan, while tissue-specific knockdown of drp-1 in neurons, intestine or muscle failed to increase daf-2 longevity. Disruption of other genes involved in mitochondrial fission also increased daf-2 lifespan as did treatment with RNA interference clones that decrease mitochondrial fragmentation. In exploring potential mechanisms involved, we found that deletion of drp-1 increases resistance to chronic stresses. In addition, we found that disruption of drp-1 increased mitochondrial and peroxisomal connectedness in daf-2 worms, increased oxidative phosphorylation and ATP levels, and increased mitophagy in daf-2 worms, but did not affect their ROS levels, food consumption or mitochondrial membrane potential. Disruption of mitophagy through RNA interference targeting pink-1 decreased the lifespan of daf-2;drp-1 worms suggesting that increased mitophagy contributes to their extended lifespan. Overall, this work defined the conditions under which drp-1 disruption increases daf-2 lifespan and has identified multiple changes in daf-2;drp-1 mutants that may contribute to their lifespan extension.
Collapse
Affiliation(s)
- Annika Traa
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Metabolic Disorders and Complications Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Aura A Tamez González
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Metabolic Disorders and Complications Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jeremy M Van Raamsdonk
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
- Metabolic Disorders and Complications Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Piergentili R, Sechi S. Non-Coding RNAs of Mitochondrial Origin: Roles in Cell Division and Implications in Cancer. Int J Mol Sci 2024; 25:7498. [PMID: 39000605 PMCID: PMC11242419 DOI: 10.3390/ijms25137498] [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: 06/05/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are a heterogeneous group, in terms of structure and sequence length, consisting of RNA molecules that do not code for proteins. These ncRNAs have a central role in the regulation of gene expression and are virtually involved in every process analyzed, ensuring cellular homeostasis. Although, over the years, much research has focused on the characterization of non-coding transcripts of nuclear origin, improved bioinformatic tools and next-generation sequencing (NGS) platforms have allowed the identification of hundreds of ncRNAs transcribed from the mitochondrial genome (mt-ncRNA), including long non-coding RNA (lncRNA), circular RNA (circRNA), and microRNA (miR). Mt-ncRNAs have been described in diverse cellular processes such as mitochondrial proteome homeostasis and retrograde signaling; however, the function of the majority of mt-ncRNAs remains unknown. This review focuses on a subgroup of human mt-ncRNAs whose dysfunction is associated with both failures in cell cycle regulation, leading to defects in cell growth, cell proliferation, and apoptosis, and the development of tumor hallmarks, such as cell migration and metastasis formation, thus contributing to carcinogenesis and tumor development. Here we provide an overview of the mt-ncRNAs/cancer relationship that could help the future development of new biomedical applications in the field of oncology.
Collapse
Affiliation(s)
| | - Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| |
Collapse
|
18
|
Su D, Song Y, Li D, Yang S, Zhan S, Zhong T, Guo J, Cao J, Li L, Zhang H, Wang L. Cold exposure affects glucose metabolism, lipid droplet deposition and mitophagy in skeletal muscle of newborn goats. Domest Anim Endocrinol 2024; 88:106847. [PMID: 38479188 DOI: 10.1016/j.domaniend.2024.106847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 06/07/2024]
Abstract
Cold exposure is a common stressor for newborn goats. Skeletal muscle plays an important role in maintaining whole-body homeostasis of glucose and lipid metabolism. However, the molecular mechanisms underlying regulation of skeletal muscle of newborn goats by cold exposure remains unclear. In this study, we found a significant increase (P < 0.01) in serum glucagon levels after 24 h of cold exposure (COLD, 6°C), while glucose and insulin concentrations were significantly decreased (P < 0.01) compared to room temperature (RT, 25°C). Additionally, we found that cold exposure reduced glycogen content (P < 0.01) in skeletal muscle. Pathway enrichment analysis revealed that cold exposure activated skeletal muscle glucose metabolism pathways (including insulin resistance and the insulin signaling pathway) and mitophagy-related pathways. Cold exposure up-regulated the expression of genes involved in fatty acid and triglyceride synthesis, promoting skeletal muscle lipid deposition. Notably, cold exposure induced mitophagy in skeletal muscle.
Collapse
Affiliation(s)
- Duo Su
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yulong Song
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Die Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shizhong Yang
- Institute of Liangshan Agricultural Science Research, Xichang 615042, China
| | - Siyuan Zhan
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Zhong
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
19
|
Wang F, Huynh PM, An YA. Protocol for Seahorse analysis of ex vivo mouse brown and white adipose tissues. STAR Protoc 2024; 5:103042. [PMID: 38850537 PMCID: PMC11215091 DOI: 10.1016/j.xpro.2024.103042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/12/2024] [Accepted: 04/12/2024] [Indexed: 06/10/2024] Open
Abstract
The mitochondrial stress test is a gold-standard approach for assessing adipose tissue physiological functions and pathological changes. Here, we present a protocol for conducting Seahorse assays using ex vivo mouse brown and white adipose depots. We describe steps for rehydrating the cartridge, preparing freshly harvested fat depots, placing them onto an islet capture plate, and incubating them in a non-CO2 incubator. We then detail procedures for adding mitochondrial stressor solutions and conducting the mitochondrial stress test using the Seahorse XFe24 Analyzer. For complete details on the use and execution of this protocol, please refer to An et al.1.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX 77030, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX 77030, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Wu K, Shieh JS, Qin L, Guo JJ. Mitochondrial mechanisms in the pathogenesis of chronic inflammatory musculoskeletal disorders. Cell Biosci 2024; 14:76. [PMID: 38849951 PMCID: PMC11162051 DOI: 10.1186/s13578-024-01259-9] [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: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Chronic inflammatory musculoskeletal disorders characterized by prolonged muscle inflammation, resulting in enduring pain and diminished functionality, pose significant challenges for the patients. Emerging scientific evidence points to mitochondrial malfunction as a pivotal factor contributing to these ailments. Mitochondria play a critical role in powering skeletal muscle activity, but in the context of persistent inflammation, disruptions in their quantity, configuration, and performance have been well-documented. Various disturbances, encompassing alterations in mitochondrial dynamics (such as fission and fusion), calcium regulation, oxidative stress, biogenesis, and the process of mitophagy, are believed to play a central role in the progression of these disorders. Additionally, unfolded protein responses and the accumulation of fatty acids within muscle cells may adversely affect the internal milieu, impairing the equilibrium of mitochondrial functioning. The structural discrepancies between different mitochondrial subsets namely, intramyofibrillar and subsarcolemmal mitochondria likely impact their metabolic capabilities and susceptibility to inflammatory influences. The release of signals from damaged mitochondria is known to incite inflammatory responses. Intriguingly, migrasomes and extracellular vesicles serve as vehicles for intercellular transfer of mitochondria, aiding in the removal of impaired mitochondria and regulation of inflammation. Viral infections have been implicated in inducing stress on mitochondria. Prolonged dysfunction of these vital organelles sustains oxidative harm, metabolic irregularities, and heightened cytokine release, impeding the body's ability to repair tissues. This review provides a comprehensive analysis of advancements in understanding changes in the intracellular environment, mitochondrial architecture and distribution, biogenesis, dynamics, autophagy, oxidative stress, cytokines associated with mitochondria, vesicular structures, and associated membranes in the context of chronic inflammatory musculoskeletal disorders. Strategies targeting key elements regulating mitochondrial quality exhibit promise in the restoration of mitochondrial function, alleviation of inflammation, and enhancement of overall outcomes.
Collapse
Affiliation(s)
- Kailun Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of Soochow University/Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People's Republic of China
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Ju-Sheng Shieh
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 11490, Taiwan
| | - Ling Qin
- Musculoskeletal Research Laboratory of the Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Jiong Jiong Guo
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.
- MOE China-Europe Sports Medicine Belt and Road Joint Laboratory, Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
21
|
Yang Z, Wei J, Liu H, Zhang H, Liu R, Tang N, Yang X. Changes in muscle strength and risk of cardiovascular disease among middle-aged and older adults in China: Evidence from a prospective cohort study. Chin Med J (Engl) 2024; 137:1343-1350. [PMID: 38407330 PMCID: PMC11191030 DOI: 10.1097/cm9.0000000000002968] [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: 10/22/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Evidence indicates that low muscle strength is associated with an increased cardiovascular diseases (CVDs) risk. However, the association between muscle strength changes based on repeated measurements and CVD incidence remains unclear. METHODS The study used data from the China Health and Retirement Longitudinal Study in 2011 (Wave 1), 2013 (Wave 2), 2015 (Wave 3), and 2018 (Wave 4). Low muscle strength was defined as handgrip strength <28 kg for men or <18 kg for women, or chair-rising time ≥12 s. Based on changes in muscle strength from Waves 1 to 2, participants were categorized into four groups of Normal-Normal, Low-Normal, Normal-Low, and Low-Low. CVD events, including heart disease and stroke, were recorded using a self-reported questionnaire during Waves 3 and 4 visits. Cox proportional hazards models were used to investigate the association between muscle strength changes and CVD incidence after multivariable adjustments. Hazard ratios (HRs) and 95% confidence intervals (95% CIs) were estimated with the Normal-Normal group as the reference. RESULTS A total of 1164 CVD cases were identified among 6608 participants. Compared to participants with sustained normal muscle strength, the CVD risks increased progressively across groups of the Low-Normal (HR = 1.20, 95% CI: 1.01-1.43), the Normal-Low (HR = 1.35, 95% CI: 1.14-1.60), and the Low-Low (HR = 1.76, 95% CI: 1.49-2.07). Similar patterns were observed for the significant associations between muscle strength status and the incidence risks of heart disease and stroke. Subgroup analyses showed that the significant associations between CVD and muscle strength changes were consistent across age, sex, and body mass index (BMI) categories. CONCLUSIONS The study found that muscle strength changes were associated with CVD risk. This suggests that continuous tracking of muscle status may be helpful in screening cardiovascular risk.
Collapse
Affiliation(s)
- Ze Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Jiemin Wei
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Hongbo Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Honglu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Ruifang Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Naijun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Xueli Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| |
Collapse
|
22
|
Celik D, Campisi M, Cannella L, Pavanello S. The effect of low birth weight as an intrauterine exposure on the early onset of sarcopenia through possible molecular pathways. J Cachexia Sarcopenia Muscle 2024; 15:770-780. [PMID: 38553412 PMCID: PMC11154781 DOI: 10.1002/jcsm.13455] [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: 08/31/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 06/07/2024] Open
Abstract
Sarcopenia, a musculoskeletal disease characterized by the progressive loss of skeletal muscle mass, strength, and physical performance, presents significant challenges to global public health due to its adverse effects on mobility, morbidity, mortality, and healthcare costs. This comprehensive review explores the intricate connections between sarcopenia and low birth weight (LBW), emphasizing the developmental origins of health and disease (DOHaD) hypothesis, inflammatory processes (inflammaging), mitochondrial dysfunction, circadian rhythm disruptions, epigenetic mechanisms, and genetic variations revealed through genome-wide studies (GWAS). A systematic search strategy was developed using PubMed to identify relevant English-language publications on sarcopenia, LBW, DOHaD, inflammaging, mitochondrial dysfunction, circadian disruption, epigenetic mechanisms, and GWAS. The publications consist of 46.2% reviews, 21.2% cohort studies, 4.8% systematic reviews, 1.9% cross-sectional studies, 13.4% animal studies, 4.8% genome-wide studies, 5.8% epigenome-wide studies, and 1.9% book chapters. The review identified key factors contributing to sarcopenia development, including the DOHaD hypothesis, LBW impact on muscle mass, inflammaging, mitochondrial dysfunction, the influence of clock genes, the role of epigenetic mechanisms, and genetic variations revealed through GWAS. The DOHaD theory suggests that LBW induces epigenetic alterations during foetal development, impacting long-term health outcomes, including the early onset of sarcopenia. LBW correlates with reduced muscle mass, grip strength, and lean body mass in adulthood, increasing the risk of sarcopenia. Chronic inflammation (inflammaging) and mitochondrial dysfunction contribute to sarcopenia, with LBW linked to increased oxidative stress and dysfunction. Disrupted circadian rhythms, regulated by genes such as BMAL1 and CLOCK, are associated with both LBW and sarcopenia, impacting lipid metabolism, muscle mass, and the ageing process. Early-life exposures, including LBW, induce epigenetic modifications like DNA methylation (DNAm) and histone changes, playing a pivotal role in sarcopenia development. Genome-wide studies have identified candidate genes and variants associated with lean body mass, muscle weakness, and sarcopenia, providing insights into genetic factors contributing to the disorder. LBW emerges as a potential early predictor of sarcopenia development, reflecting the impact of intrauterine exposures on long-term health outcomes. Understanding the complex interplay between LBW with inflammaging, mitochondrial dysfunction, circadian disruption, and epigenetic factors is essential for elucidating the pathogenesis of sarcopenia and developing targeted interventions. Future research on GWAS and the underlying mechanisms of LBW-associated sarcopenia is warranted to inform preventive strategies and improve public health outcomes.
Collapse
Affiliation(s)
- Dilek Celik
- Department of Pharmceutical and Pharmacological SciencesUniversity of PaduaPaduaItaly
| | - Manuela Campisi
- Department of Cardiac Thoracic Vascular Sciences and Public HealthUniversity of PaduaPaduaItaly
| | - Luana Cannella
- Department of Cardiac Thoracic Vascular Sciences and Public HealthUniversity of PaduaPaduaItaly
| | - Sofia Pavanello
- Department of Cardiac Thoracic Vascular Sciences and Public HealthUniversity of PaduaPaduaItaly
- University Hospital of PadovaPaduaItaly
| |
Collapse
|
23
|
Zhang TR, Chiang CH, Hsu TC, Wang CY, Chen CY. Age and dietary restriction modulate mitochondrial quality in quadriceps femoris muscle of male mice. Biogerontology 2024; 25:447-459. [PMID: 38183523 DOI: 10.1007/s10522-023-10086-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
Dietary restriction (DR) is a potential intervention for ameliorating ageing-related damages. Mitochondrial quality control is the key mechanism for regulating cellular functions in skeletal muscle. This study aimed to explore the effect of age and DR on the homeostasis of mitochondrial quality control in skeletal muscle. To study the effect of age on mitochondrial homeostasis, young (3 months old) male C57BL/6J mice were fed ad libitum (AL) until 7 (Young), 14 (Middle), and 19 months (Aged) of age. For the DR intervention, 60% of AL intake was given to the mice at 3 months of age until they reached 19 months of age (16 months). The quadriceps femoris muscle was collected for further analysis. Significant changes in the skeletal muscle were noticed during the transition between middle age and the elderly stages. An accumulation of collagen was observed in the muscle after middle age. Compared with the Middle muscle, Aged muscle displayed a greater expression of VDAC, and lower expressions of mitochondrial dynamic proteins and OXPHOS proteins. The DR intervention attenuated collagen content and elongated the sarcomere length in the skeletal muscle during ageing. In addition, DR adjusted the abnormalities in mitochondrial morphology in the Aged muscle. DR downregulated VDAC expression, but upregulated OPA1 and DRP1 expressions. Taken together, greater pathological changes were noticed in the skeletal muscle during ageing, especially in the transition between middle age and the elderly, whereas early-onset DR attenuated the muscular ageing via normalising partial functions of mitochondria.
Collapse
Affiliation(s)
- Ting-Rui Zhang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chun-Hsien Chiang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Tzu-Chieh Hsu
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chih-Yun Wang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan.
| |
Collapse
|
24
|
Xu H, Xiang QY, Zhan JK, Wang Y, Wang YJ, Li S, Liu YS. Association between macro- and microvascular damage and sarcopenia index in individuals with type 2 diabetes mellitus. Appl Physiol Nutr Metab 2024; 49:762-772. [PMID: 38346295 DOI: 10.1139/apnm-2023-0476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Sarcopenia was recently reported to be relevant to an increased macro-and microvascular disease risk. Sarcopenia index (SI) has been identified as a surrogate marker for sarcopenia. The aim of the present study was to investigate the association between macro- and microvascular disease and SI in patients with type 2 diabetes mellitus (T2DM). A total of 783 patients with T2DM were enrolled in this cross-sectional study. The SI was calculated by (serum creatinine [mg/dL]/cystatin C [mg/L]) × 100. The subjects were divided into three groups according to SI tertiles: T1 (41.27-81.37), T2 (81.38- 99.55), and T3 (99.56-192.31). Parameters of macro- and microvascular complications, including diabetic retinopathy (DR), micro- and macroalbuminuria (MAU), diabetic peripheral neuropathy (DPN), and lower extremity peripheral artery disease (LEAD) were evaluated. Multivariate logistic regression analysis revealed that when taking the top tertile of SI as a reference, an increasing trend of the prevalence of DR, MAU, DPN, and LEAD were presented (all P for trend < 0.05), where the OR (95% CI) for DR prevalence was 1.967 (1.252-3.090) in T2, 2.195 (1.278-3.769) in T1, for MAU was 1.805 (1.149-2.837) in T2, 2.537 (1.490-4.320) in T1, for DPN was 2.244 (1.485-3.391) in T2, 3.172 (1.884-5.341) in T1, and for LEAD was 2.017 (1.002-4.057) in T2, 2.405 (1.107-5.225) in T1 (all P < 0.05). Patients with lower SI were more inclined to have an increased risk of macro- and microvascular damage in T2DM population, which may be related to sarcopenia.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, Peking University First Hospital, Beijing, 100034, China
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
| | - Qun-Yan Xiang
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jun-Kun Zhan
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yi Wang
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yan-Jiao Wang
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shuang Li
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - You-Shuo Liu
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, 410011, China
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|
25
|
Neal CL, Kronert WA, Camillo JRT, Suggs JA, Huxford T, Bernstein SI. Aging-affiliated post-translational modifications of skeletal muscle myosin affect biochemical properties, myofibril structure, muscle function, and proteostasis. Aging Cell 2024; 23:e14134. [PMID: 38506610 PMCID: PMC11296117 DOI: 10.1111/acel.14134] [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: 09/20/2023] [Revised: 12/18/2023] [Accepted: 02/12/2024] [Indexed: 03/21/2024] Open
Abstract
The molecular motor myosin is post-translationally modified in its globular head, its S2 hinge, and its thick filament domain during human skeletal muscle aging. To determine the importance of such modifications, we performed an integrative analysis of transgenic Drosophila melanogaster expressing myosin containing post-translational modification mimic mutations. We determined effects on muscle function, myofibril structure, and myosin biochemistry. Modifications in the homozygous state decreased jump muscle function by a third at 3 weeks of age and reduced indirect flight muscle function to negligible levels in young flies, with severe effects on flight muscle myofibril assembly and/or maintenance. Expression of mimic mutations in the heterozygous state or in a wild-type background yielded significant, but less severe, age-dependent effects upon flight muscle structure and function. Modification of the residue in the globular head disabled ATPase activity and in vitro actin filament motility, whereas the S2 hinge mutation reduced actin-activated ATPase activity by 30%. The rod modification diminished filament formation in vitro. The latter mutation also reduced proteostasis, as demonstrated by enhanced accumulation of polyubiquitinated proteins. Overall, we find that mutation of amino acids at sites that are chemically modified during human skeletal muscle aging can disrupt myosin ATPase, myosin filament formation, and/or proteostasis, providing a mechanistic basis for the observed muscle defects. We conclude that age-specific post-translational modifications present in human skeletal muscle are likely to act in a dominant fashion to affect muscle structure and function and may therefore be implicated in degeneration and dysfunction associated with sarcopenia.
Collapse
Affiliation(s)
- Clara L. Neal
- Department of Biology, Molecular Biology Institute, Heart InstituteSan Diego State UniversitySan DiegoCaliforniaUSA
| | - William A. Kronert
- Department of Biology, Molecular Biology Institute, Heart InstituteSan Diego State UniversitySan DiegoCaliforniaUSA
| | - Jared Rafael T. Camillo
- Department of Biology, Molecular Biology Institute, Heart InstituteSan Diego State UniversitySan DiegoCaliforniaUSA
| | - Jennifer A. Suggs
- Department of Biology, Molecular Biology Institute, Heart InstituteSan Diego State UniversitySan DiegoCaliforniaUSA
| | - Tom Huxford
- Department of Chemistry and BiochemistrySan Diego State UniversitySan DiegoCaliforniaUSA
| | - Sanford I. Bernstein
- Department of Biology, Molecular Biology Institute, Heart InstituteSan Diego State UniversitySan DiegoCaliforniaUSA
| |
Collapse
|
26
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
27
|
Jitngamsujarit S, Salang L, Saengboonmee C, Sorin S, Thithuan K, Pongsritasana T, Sukkasame S. Advancing Age May Decrease Mitochondrial Activity in Cumulus Cells. J Clin Med 2024; 13:2800. [PMID: 38792342 PMCID: PMC11122456 DOI: 10.3390/jcm13102800] [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: 04/18/2024] [Revised: 05/04/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Background: The goal of this study was to compare mitochondrial activity in cumulus cells (CCs) between young and advancing-aged women, the factors that affect mitochondrial activity, and their association with blastocyst quality. Materials and methods: This prospective study included 80 infertile women who underwent ICSI between May and October 2023. Participants were divided into two groups: older and younger than 38. The oocyte mitochondrial activity from CCs was evaluated using MitoTracker, and the mean fluorescence intensity (MFI) was also evaluated. Results: The univariate and multivariate analyses revealed a significant difference in the MFI between the woman ≥ 38 age group and the lower age group (162.68 ± 79.87 vs. 228.39 ± 121.38; p-value = 0.005; 95%CI 19.97, 111.45). The factors that affected the MFI were women ≥ 38 years of age (p-value = 0.005; 95%CI -111.45, -19.91), total gonadotropin dosages (p-value = 0.006; 95%CI -0.08, 0.01), and gonadotropin-releasing hormone agonist (GnRHa) triggering (p-value = 0.006; 95%CI 36.46, 210.06). However, only women aged ≥38 years remained statistically significant after a multivariable regression analysis (p-value = 0.014; 95%CI -121.00, -14.30). In addition, only male age (mean age ± SD = 38.26 ± 5.13) was associated with high blastocyst quality in univariate and mixed multivariate analyses (OR 0.91; 95%CI 0.56, 3.04). The chemical pregnancy rate was not significantly different between the two age groups (34.5% vs. 56.7%; p-value = 0.162; 95%CI 0.2, 1.30). Conclusion: Advancing age decreased mitochondrial activity in CCs but did not affect blastocyst quality. By contrast, male age may be a predictor of high-grade blastocyst quality.
Collapse
Affiliation(s)
- Suwichaya Jitngamsujarit
- Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (S.J.); (T.P.); (S.S.)
| | - Lingling Salang
- Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (S.J.); (T.P.); (S.S.)
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (C.S.); (S.S.); (K.T.)
| | - Supannika Sorin
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (C.S.); (S.S.); (K.T.)
| | - Kanyarat Thithuan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (C.S.); (S.S.); (K.T.)
| | - Thanida Pongsritasana
- Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (S.J.); (T.P.); (S.S.)
| | - Sineenart Sukkasame
- Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (S.J.); (T.P.); (S.S.)
| |
Collapse
|
28
|
Wong JJ, Ho JS, Teo LLY, Wee HN, Chua KV, Ching J, Gao F, Tan SY, Tan RS, Kovalik JP, Koh AS. Effects of short-term moderate intensity exercise on the serum metabolome in older adults: a pilot randomized controlled trial. COMMUNICATIONS MEDICINE 2024; 4:80. [PMID: 38704414 PMCID: PMC11069586 DOI: 10.1038/s43856-024-00507-w] [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: 05/09/2023] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND We previously reported changes in the serum metabolome associated with impaired myocardial relaxation in an asymptomatic older community cohort. In this prospective parallel-group randomized control pilot trial, we subjected community adults without cardiovascular disease to exercise intervention and evaluated the effects on serum metabolomics. METHODS Between February 2019 to November 2019, thirty (83% females) middle-aged adults (53 ± 4 years) were randomized with sex stratification to either twelve weeks of moderate-intensity exercise training (Intervention) (n = 15) or Control (n = 15). The Intervention group underwent once-weekly aerobic and strength training sessions for 60 min each in a dedicated cardiac exercise laboratory for twelve weeks (ClinicalTrials.gov: NCT03617653). Serial measurements were taken pre- and post-intervention, including serum sampling for metabolomic analyses. RESULTS Twenty-nine adults completed the study (Intervention n = 14; Control n = 15). Long-chain acylcarnitine C20:2-OH/C18:2-DC was reduced in the Intervention group by a magnitude of 0.714 but increased in the Control group by a magnitude of 1.742 (mean difference -1.028 age-adjusted p = 0.004). Among Controls, alanine correlated with left ventricular mass index (r = 0.529, age-adjusted p = 0.018) while aspartate correlated with Lateral e' (r = -764, age-adjusted p = 0.016). C20:3 correlated with E/e' ratio fold-change in the Intervention group (r = -0.653, age-adjusted p = 0.004). Among Controls, C20:2/C18:2 (r = 0.795, age-adjusted p = 0.005) and C20:2-OH/C18:2-DC fold-change (r = 0.742, age-adjusted p = 0.030) correlated with change in E/A ratio. CONCLUSIONS Corresponding relationships between serum metabolites and cardiac function in response to exercise intervention provided pilot observations. Future investigations into cellular fuel oxidation or central carbon metabolism pathways that jointly impact the heart and related metabolic systems may be critical in preventive trials.
Collapse
Affiliation(s)
- Jie Jun Wong
- National Heart Centre Singapore, Singapore, Singapore
| | - Jien Sze Ho
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Louis L Y Teo
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | | | | | | | - Fei Gao
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Swee Yaw Tan
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Ru-San Tan
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Jean-Paul Kovalik
- Duke-NUS Medical School, Singapore, Singapore
- Singapore General Hospital, Singapore, Singapore
| | - Angela S Koh
- National Heart Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
29
|
Bradley CE, Fletcher E, Wilkinson T, Ring A, Ferrer L, Miserlis D, Pacher P, Koutakis P. Mitochondrial fatty acid beta-oxidation: a possible therapeutic target for skeletal muscle lipotoxicity in peripheral artery disease myopathy. EXCLI JOURNAL 2024; 23:523-533. [PMID: 38741727 PMCID: PMC11089102 DOI: 10.17179/excli2024-7004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
Peripheral artery disease (PAD) is an atherosclerotic disease impacting over 200 million individuals and the prevalence increases with age. PAD occurs when plaque builds up within the peripheral arteries, leading to reduced blood flow and oxygen supply to the outer extremities. Individuals who experience PAD suffer from ischemia, which is typically accompanied by significant damage to skeletal muscles. Additionally, this tissue damage affects mitochondria, causing them to become dysregulated and dysfunctional, resulting in decreased metabolic rates. As there is no known cure for PAD, researchers are exploring potential therapeutic targets by examining coexisting cardiovascular conditions and metabolic risk factors, such as the aging process. Among these comorbidities, type-two diabetes mellitus and obesity are particularly common in PAD cases. These conditions, along with aging itself, are associated with an elevated accumulation of ectopic lipids within skeletal muscles, similar to what is observed in PAD. Researchers have attempted to reduce excess lipid accumulation by increasing the rate of fatty acid beta oxidation. Manipulating acetyl coenzyme A carboxylase 2, a key regulatory protein of fatty acid beta oxidation, has been the primary focus of such research. When acetyl coenzyme A carboxylase 2 is inhibited, it interrupts the conversion of acetyl-CoA into malonyl-CoA, resulting in an increase in the rate of fatty acid beta oxidation. By utilizing samples from PAD patients and applying the pharmacological strategies developed for acetyl coenzyme A carboxylase 2 in diabetes and obesity to PAD, a potential new therapeutic avenue may emerge, offering hope for improved quality of life for individuals suffering from PAD.
Collapse
Affiliation(s)
- Cassandra E. Bradley
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Emma Fletcher
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Trevor Wilkinson
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Andrew Ring
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Lucas Ferrer
- Department of Surgery, University of Texas at Austin Dell Medical School, 1601 Trinity St, Room 6708A, Austin, TX 78712, USA
| | - Dimitrios Miserlis
- Department of Surgery, University of Texas at Austin Dell Medical School, 1601 Trinity St, Room 6708A, Austin, TX 78712, USA
| | - Pal Pacher
- National Institutes of Health, Bethesda, MD, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| |
Collapse
|
30
|
Vlasova AD, Bukhalovich SM, Bagaeva DF, Polyakova AP, Ilyinsky NS, Nesterov SV, Tsybrov FM, Bogorodskiy AO, Zinovev EV, Mikhailov AE, Vlasov AV, Kuklin AI, Borshchevskiy VI, Bamberg E, Uversky VN, Gordeliy VI. Intracellular microbial rhodopsin-based optogenetics to control metabolism and cell signaling. Chem Soc Rev 2024; 53:3327-3349. [PMID: 38391026 DOI: 10.1039/d3cs00699a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Microbial rhodopsin (MRs) ion channels and pumps have become invaluable optogenetic tools for neuroscience as well as biomedical applications. Recently, MR-optogenetics expanded towards subcellular organelles opening principally new opportunities in optogenetic control of intracellular metabolism and signaling via precise manipulations of organelle ion gradients using light. This new optogenetic field expands the opportunities for basic and medical studies of cancer, cardiovascular, and metabolic disorders, providing more detailed and accurate control of cell physiology. This review summarizes recent advances in studies of the cellular metabolic processes and signaling mediated by optogenetic tools targeting mitochondria, endoplasmic reticulum (ER), lysosomes, and synaptic vesicles. Finally, we discuss perspectives of such an optogenetic approach in both fundamental and applied research.
Collapse
Affiliation(s)
- Anastasiia D Vlasova
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Siarhei M Bukhalovich
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Diana F Bagaeva
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksandra P Polyakova
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Nikolay S Ilyinsky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Semen V Nesterov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Fedor M Tsybrov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Andrey O Bogorodskiy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Egor V Zinovev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Anatolii E Mikhailov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Alexey V Vlasov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Dubna, Russia
| | - Alexander I Kuklin
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Dubna, Russia
| | - Valentin I Borshchevskiy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Dubna, Russia
| | - Ernst Bamberg
- Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Valentin I Gordeliy
- Institut de Biologie Structurale Jean-Pierre Ebel, Université Grenoble Alpes-Commissariat à l'Energie Atomique et aux Energies Alternatives-CNRS, 38027 Grenoble, France.
| |
Collapse
|
31
|
Wang H, Yuan T, Wang Y, Liu C, Li D, Li Z, Sun S. Osteoclasts and osteoarthritis: Novel intervention targets and therapeutic potentials during aging. Aging Cell 2024; 23:e14092. [PMID: 38287696 PMCID: PMC11019147 DOI: 10.1111/acel.14092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Osteoarthritis (OA), a chronic degenerative joint disease, is highly prevalent among the aging population, and often leads to joint pain, disability, and a diminished quality of life. Although considerable research has been conducted, the precise molecular mechanisms propelling OA pathogenesis continue to be elusive, thereby impeding the development of effective therapeutics. Notably, recent studies have revealed subchondral bone lesions precede cartilage degeneration in the early stage of OA. This development is marked by escalated osteoclast-mediated bone resorption, subsequent imbalances in bone metabolism, accelerated bone turnover, and a decrease in bone volume, thereby contributing significantly to the pathological changes. While the role of aging hallmarks in OA has been extensively elucidated from the perspective of chondrocytes, their connection with osteoclasts is not yet fully understood. There is compelling evidence to suggest that age-related abnormalities such as epigenetic alterations, proteostasis network disruption, cellular senescence, and mitochondrial dysfunction, can stimulate osteoclast activity. This review intends to systematically discuss how aging hallmarks contribute to OA pathogenesis, placing particular emphasis on the age-induced shifts in osteoclast activity. It also aims to stimulate future studies probing into the pathological mechanisms and therapeutic approaches targeting osteoclasts in OA during aging.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Yi Wang
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Changxing Liu
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Dengju Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Ziqing Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
32
|
Somers T, Siddiqi S, Maas RGC, Sluijter JPG, Buikema JW, van den Broek PHH, Meuwissen TJ, Morshuis WJ, Russel FGM, Schirris TJJ. Statins affect human iPSC-derived cardiomyocytes by interfering with mitochondrial function and intracellular acidification. Basic Res Cardiol 2024; 119:309-327. [PMID: 38305903 DOI: 10.1007/s00395-023-01025-x] [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: 02/06/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Statins are effective drugs in reducing cardiovascular morbidity and mortality by inhibiting cholesterol synthesis. These effects are primarily beneficial for the patient's vascular system. A significant number of statin users suffer from muscle complaints probably due to mitochondrial dysfunction, a mechanism that has recently been elucidated. This has raised our interest in exploring the effects of statins on cardiac muscle cells in an era where the elderly and patients with poorer functioning hearts and less metabolic spare capacity start dominating our patient population. Here, we investigated the effects of statins on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-derived CMs). hiPSC-derived CMs were exposed to simvastatin, atorvastatin, rosuvastatin, and cerivastatin at increasing concentrations. Metabolic assays and fluorescent microscopy were employed to evaluate cellular viability, metabolic capacity, respiration, intracellular acidity, and mitochondrial membrane potential and morphology. Over a concentration range of 0.3-100 µM, simvastatin lactone and atorvastatin acid showed a significant reduction in cellular viability by 42-64%. Simvastatin lactone was the most potent inhibitor of basal and maximal respiration by 56% and 73%, respectively, whereas simvastatin acid and cerivastatin acid only reduced maximal respiration by 50% and 42%, respectively. Simvastatin acid and lactone and atorvastatin acid significantly decreased mitochondrial membrane potential by 20%, 6% and 3%, respectively. The more hydrophilic atorvastatin acid did not seem to affect cardiomyocyte metabolism. This calls for further research on the translatability to the clinical setting, in which a more conscientious approach to statin prescribing might be considered, especially regarding the current shift in population toward older patients with poor cardiac function.
Collapse
Affiliation(s)
- Tim Somers
- Department of Cardiothoracic Surgery, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Sailay Siddiqi
- Department of Cardiothoracic Surgery, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Renee G C Maas
- Department of Cardiology, Experimental Cardiology Laboratory, Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Jan W Buikema
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam Heart Center, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HZ, Amsterdam, The Netherlands
| | - Petra H H van den Broek
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Tanne J Meuwissen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Wim J Morshuis
- Department of Cardiothoracic Surgery, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Frans G M Russel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
| | - Tom J J Schirris
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
33
|
Yin A, Fu W, Elengickal A, Kim J, Liu Y, Bigot A, Mamchaoui K, Call JA, Yin H. Chronic hypoxia impairs skeletal muscle repair via HIF-2α stabilization. J Cachexia Sarcopenia Muscle 2024; 15:631-645. [PMID: 38333911 PMCID: PMC10995261 DOI: 10.1002/jcsm.13436] [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: 08/14/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Chronic hypoxia and skeletal muscle atrophy commonly coexist in patients with COPD and CHF, yet the underlying physio-pathological mechanisms remain elusive. Muscle regeneration, driven by muscle stem cells (MuSCs), holds therapeutic potential for mitigating muscle atrophy. This study endeavours to investigate the influence of chronic hypoxia on muscle regeneration, unravel key molecular mechanisms, and explore potential therapeutic interventions. METHODS Experimental mice were exposed to prolonged normobaric hypoxic air (15% pO2, 1 atm, 2 weeks) to establish a chronic hypoxia model. The impact of chronic hypoxia on body composition, muscle mass, muscle strength, and the expression levels of hypoxia-inducible factors HIF-1α and HIF-2α in MuSC was examined. The influence of chronic hypoxia on muscle regeneration, MuSC proliferation, and the recovery of muscle mass and strength following cardiotoxin-induced injury were assessed. The muscle regeneration capacities under chronic hypoxia were compared between wildtype mice, MuSC-specific HIF-2α knockout mice, and mice treated with HIF-2α inhibitor PT2385, and angiotensin converting enzyme (ACE) inhibitor lisinopril. Transcriptomic analysis was performed to identify hypoxia- and HIF-2α-dependent molecular mechanisms. Statistical significance was determined using analysis of variance (ANOVA) and Mann-Whitney U tests. RESULTS Chronic hypoxia led to limb muscle atrophy (EDL: 17.7%, P < 0.001; Soleus: 11.5% reduction in weight, P < 0.001) and weakness (10.0% reduction in peak-isometric torque, P < 0.001), along with impaired muscle regeneration characterized by diminished myofibre cross-sectional areas, increased fibrosis (P < 0.001), and incomplete strength recovery (92.3% of pre-injury levels, P < 0.05). HIF-2α stabilization in MuSC under chronic hypoxia hindered MuSC proliferation (26.1% reduction of MuSC at 10 dpi, P < 0.01). HIF-2α ablation in MuSC mitigated the adverse effects of chronic hypoxia on muscle regeneration and MuSC proliferation (30.9% increase in MuSC numbers at 10 dpi, P < 0.01), while HIF-1α ablation did not have the same effect. HIF-2α stabilization under chronic hypoxia led to elevated local ACE, a novel direct target of HIF-2α. Notably, pharmacological interventions with PT2385 or lisinopril enhanced muscle regeneration under chronic hypoxia (PT2385: 81.3% increase, P < 0.001; lisinopril: 34.6% increase in MuSC numbers at 10 dpi, P < 0.05), suggesting their therapeutic potential for alleviating chronic hypoxia-associated muscle atrophy. CONCLUSIONS Chronic hypoxia detrimentally affects skeletal muscle regeneration by stabilizing HIF-2α in MuSC and thereby diminishing MuSC proliferation. HIF-2α increases local ACE levels in skeletal muscle, contributing to hypoxia-induced regenerative deficits. Administration of HIF-2α or ACE inhibitors may prove beneficial to ameliorate chronic hypoxia-associated muscle atrophy and weakness by improving muscle regeneration under chronic hypoxia.
Collapse
Affiliation(s)
- Amelia Yin
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Wenyan Fu
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Anthony Elengickal
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Joonhee Kim
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Yang Liu
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de MyologieCentre de Recherche en MyologieParisFrance
| | - Kamal Mamchaoui
- Sorbonne Université, Inserm, Institut de MyologieCentre de Recherche en MyologieParisFrance
| | - Jarrod A. Call
- Department of Physiology and PharmacologyThe University of GeorgiaAthensGAUSA
| | - Hang Yin
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| |
Collapse
|
34
|
Nie L, He K, Qiu C, Li Q, Xiong B, Gao C, Zhang X, Jing M, Wu W, Liu J, Zhang G, Zhang Z, Yang X, Sun Y, Wang Y. Tetramethylpyrazine Nitrone alleviates D-galactose-induced murine skeletal muscle aging and motor deficits by activating the AMPK signaling pathway. Biomed Pharmacother 2024; 173:116415. [PMID: 38479182 DOI: 10.1016/j.biopha.2024.116415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Tetramethylpyrazine nitrone (TBN), a novel derivative of tetramethylpyrazine (TMP) designed and synthesized by our group, possesses multi-functional mechanisms of action and displays broad protective effects in vitro and in animal models of age-related brain disorders such as stroke, Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Parkinson's disease (PD). In the present report, we investigated the effects of TBN on aging, specifically on muscle aging and the associated decline of motor functions. Using a D-galactose-induced aging mouse model, we found that TBN could reverse the levels of several senescence and aging markers including p16, p21, ceramides, and telomere length and increase the wet-weight ratio of gastrocnemius muscle tissue, demonstrating its efficacy in ameliorating muscle aging. Additionally, the pharmacological effects of TBN on motor deficits (gait analysis, pole-climbing test and grip strength test), muscle fibrosis (hematoxylin & eosin (HE), Masson staining, and αSMA staining), inflammatory response (IL-1β, IL-6, and TNF-α), and mitochondrial function (ATP, mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) were also confirmed in the D-galactose-induced aging models. Further experiments demonstrated that TBN alleviated muscle aging and improved the decline of age-related motor deficits through an AMPK-dependent mechanism. These findings highlight the significance of TBN as a potential anti-aging agent to combat the occurrence and development of aging and age-related diseases.
Collapse
Affiliation(s)
- Lulin Nie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China; Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Kaiwu He
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Chaoming Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China; Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Qing Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Bocheng Xiong
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Chuanyue Gao
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Xiufen Zhang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Mei Jing
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Wei Wu
- Department of Hematology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Jianjun Liu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Gaoxiao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
| | - Yewei Sun
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China.
| | - Yuqiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou 510632, China.
| |
Collapse
|
35
|
Chen Z, Zhang C, Zhu Y, Gao D, Mao M, Zuo Z. Sacubitril/valsartan can improve the cardiac function in heart failure patients with a history of cancer: An observational study. Medicine (Baltimore) 2024; 103:e37613. [PMID: 38517992 PMCID: PMC10957021 DOI: 10.1097/md.0000000000037613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 02/23/2024] [Indexed: 03/24/2024] Open
Abstract
Sacubitril/Valsartan, the combination of angiotensin receptor inhibitor and neprilysin inhibitor, is now becoming the class 1 recommendation for HFrEF. Some studies have shown the positive effect of Sacubitril/Valsartan on HFrEF cancer patients, while there is devoid of evidence about the effect of this drug in aged cancer patients with HFmrEF and HFpEF. By searching the patients with a diagnosis of both cancer and Heart failure (HF) over 65, the patients who had received treatment with Sacubitril/Valsartan were selected as the candidates for Sacubitril/Valsartan group, and the patients who had received conventional HF therapy without Sacubitril/Valsartan were chosen as the control group. Data were collected for up to 9 months. We filtered 38 patients and 50 patients valid for Sacubitril/Valsartan group and control group, respectively. After initiation of heart failure management, our study found a better cardiac condition in Sacubitril/Valsartan group, having better LVEF, LVFS, NT-proBNP in 3rd, 6th, 9th month (P < .05) and better NYHA function classification after the treatment. We also observed fewer cases of deterioration on LAD (P = .029) and LVEDD (P = .023) in Sacubitril/Valsartan group. In subgroup analysis, our study showed that all 3 kinds of HF patients had better LVEF, LVFS, and NT-proBNP in Sacubitril/Valsartan group (P < .05). Our study further indicated that Sacubitril/Valsartan can improve cardiac function and benefit cardiac remolding in aged cancer patients of all 3 kinds of HF. This is the first study to provide new evidence for the use of Sacubitril/Valsartan in aged cancer patients of 3 kinds of HF.
Collapse
Affiliation(s)
- Zhulu Chen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxi Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Diansa Gao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Mao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhong Zuo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
36
|
Yuan W, Lin H, Sun Y, Liu L, Yan M, Song Y, Zhang X, Lu X, Xu Y, He Q, Ouyang K, Zhang C, Pan Y, Huang Y, Li Y, Lu X, Liu J. Myocardin reverses insulin resistance and ameliorates cardiomyopathy by increasing IRS-1 expression in a murine model of lipodystrophy caused by adipose deficiency of vacuolar H +-ATPase V0d1 subunit. Theranostics 2024; 14:2246-2264. [PMID: 38505620 PMCID: PMC10945344 DOI: 10.7150/thno.93192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/02/2024] [Indexed: 03/21/2024] Open
Abstract
Aim: Adipose tissue (AT) dysfunction that occurs in both obesity and lipodystrophy is associated with the development of cardiomyopathy. However, it is unclear how dysfunctional AT induces cardiomyopathy due to limited animal models available. We have identified vacuolar H+-ATPase subunit Vod1, encoded by Atp6v0d1, as a master regulator of adipogenesis, and adipose-specific deletion of Atp6v0d1 (Atp6v0d1AKO) in mice caused generalized lipodystrophy and spontaneous cardiomyopathy. Using this unique animal model, we explore the mechanism(s) underlying lipodystrophy-related cardiomyopathy. Methods and Results: Atp6v0d1AKO mice developed cardiac hypertrophy at 12 weeks, and progressed to heart failure at 28 weeks. The Atp6v0d1AKO mouse hearts exhibited excessive lipid accumulation and altered lipid and glucose metabolism, which are typical for obesity- and diabetes-related cardiomyopathy. The Atp6v0d1AKO mice developed cardiac insulin resistance evidenced by decreased IRS-1/2 expression in hearts. Meanwhile, the expression of forkhead box O1 (FoxO1), a transcription factor which plays critical roles in regulating cardiac lipid and glucose metabolism, was increased. RNA-seq data and molecular biological assays demonstrated reduced expression of myocardin, a transcription coactivator, in Atp6v0d1AKO mouse hearts. RNA interference (RNAi), luciferase reporter and ChIP-qPCR assays revealed the critical role of myocardin in regulating IRS-1 transcription through the CArG-like element in IRS-1 promoter. Reducing IRS-1 expression with RNAi increased FoxO1 expression, while increasing IRS-1 expression reversed myocardin downregulation-induced FoxO1 upregulation in cardiomyocytes. In vivo, restoring myocardin expression specifically in Atp6v0d1AKO cardiomyocytes increased IRS-1, but decreased FoxO1 expression. As a result, the abnormal expressions of metabolic genes in Atp6v0d1AKO hearts were reversed, and cardiac dysfunctions were ameliorated. Myocardin expression was also reduced in high fat diet-induced diabetic cardiomyopathy and palmitic acid-treated cardiomyocytes. Moreover, increasing systemic insulin resistance with rosiglitazone restored cardiac myocardin expression and improved cardiac functions in Atp6v0d1AKO mice. Conclusion: Atp6v0d1AKO mice are a novel animal model for studying lipodystrophy- or metabolic dysfunction-related cardiomyopathy. Moreover, myocardin serves as a key regulator of cardiac insulin sensitivity and metabolic homeostasis, highlighting myocardin as a potential therapeutic target for treating lipodystrophy- and diabetes-related cardiomyopathy.
Collapse
Affiliation(s)
- Wenlin Yuan
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Hui Lin
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yuan Sun
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Pharmacology, College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Lihuan Liu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Meijuan Yan
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yujuan Song
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiaofan Zhang
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiangling Lu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yipei Xu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qiyue He
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chenglin Zhang
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yong Pan
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ying Li
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xifeng Lu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Jie Liu
- Department of pathophysiology, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
37
|
Jiang M, Ren X, Han L, Zheng X. Associations between sarcopenic obesity and risk of cardiovascular disease: A population-based cohort study among middle-aged and older adults using the CHARLS. Clin Nutr 2024; 43:796-802. [PMID: 38350287 DOI: 10.1016/j.clnu.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/08/2024] [Accepted: 02/02/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Previous studies have demonstrated that sarcopenia was associated with a high risk of cardiovascular disease (CVD). Nevertheless, little is known about the associations between sarcopenic obesity/possibly sarcopenic obesity and the risk of CVD among senior and middle-aged adults. METHODS Utilizing the nationally representative data from the China Health and Retirement Longitudinal Study (CHARLS), a sum of 7703 individuals aged at least 45 years were divided into four groups. The effects of sarcopenic obesity and possibly sarcopenic obesity on CVD were calculated using Cox proportional hazards regression models. Non-sarcopenic participants with optimal body mass index (BMI) or waist circumference (WC) served as a control group. RESULTS Sarcopenic obesity were related to increased risks of CVD (HR = 1.39; 95% CI = 1.16-1.67), heart disease (HR = 1.36; 95% CI = 1.10-1.67) and stroke (HR = 1.40; 95% CI = 1.02-1.92) compared with the optimal reference group. Similarly, the risk of CVD, heart disease and stroke increased by 0.34, 0.28 and 0.39 times in obese people with possible sarcopenia compared to the control group. Sensitivity analysis identified similar results to those described above. Patients with sarcopenia and a BMI ≥28.0 kg/m2 had a 1.47- and 1.48-fold risk of developing CVD and heart disease than controls. CONCLUSION Sarcopenic obesity and possibly sarcopenic obesity are positively associated with the development of CVD. The middle-aged and elderly population should prevent obesity and maintain muscle mass through some interventions such as weight control and moderate exercise, which may reduce the CVD risk.
Collapse
Affiliation(s)
- Minglan Jiang
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xiao Ren
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Longyang Han
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xiaowei Zheng
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
38
|
Zhang Y, Jiao X, Liu J, Feng G, Luo X, Zhang M, Zhang B, Huang L, Long Q. A new direction in Chinese herbal medicine ameliorates for type 2 diabetes mellitus: Focus on the potential of mitochondrial respiratory chain complexes. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117484. [PMID: 38012971 DOI: 10.1016/j.jep.2023.117484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetes is a common chronic disease. Chinese herbal medicine (CHM) has a history of several thousand years in the treatment of diabetes, and active components with hypoglycemic effects extracted from various CHM, such as polysaccharides, flavonoids, terpenes, and steroidal saponins, have been widely used in the treatment of diabetes. AIM OF THE STUDY Research exploring the potential of various CHM compounds to regulate the mitochondrial respiratory chain complex to improve type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS The literature data were primarily obtained from authoritative databases such as PubMed, CNKI, Wanfang, and others within the last decade. The main keywords used include "type 2 diabetes mellitus", "Chinese medicine", "Chinese herbal medicine", "mitochondrial respiratory chain complex", and "mitochondrial dysfunction". RESULTS Chinese herbal medicine primarily regulates the activity of mitochondrial respiratory chain complexes in various tissues such as liver, adipose tissue, skeletal muscle, pancreatic islets, and small intestine. It improves cellular energy metabolism through hypoglycemic, antioxidant, anti-inflammatory and lipid-modulating effects. Different components of CHM can regulate the same mitochondrial respiratory chain complexes, while the same components of a particular CHM can regulate different complex activities. The active components of CHM target different mitochondrial respiratory chain complexes, regulate their aberrant changes and effectively improve T2DM and its complications. CONCLUSION Chinese herbal medicine can modulate the function of mitochondrial respiratory chain complexes in various cell types and exert their hypoglycemic effects through various mechanisms. CHM has significant therapeutic potential in regulating mitochondrial respiratory chain complexes to improve T2DM, but further research is needed to explore the underlying mechanisms and conduct clinical trials to assess the safety and efficacy of these medications. This provides new perspectives and opportunities for personalized improvement and innovative developments in diabetes management.
Collapse
Affiliation(s)
- Yinghui Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xinyue Jiao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianying Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Gang Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Mingyue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Binzhi Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
39
|
Hu T, Shi Z, Sun Y, Hu F, Rong Y, Wang J, Wang L, Xu W, Zhang F, Zhang WZ. SEPHS1 attenuates intervertebral disc degeneration by delaying nucleus pulposus cell senescence through the Hippo-Yap/Taz pathway. Am J Physiol Cell Physiol 2024; 326:C386-C399. [PMID: 38105759 DOI: 10.1152/ajpcell.00571.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Nucleus pulposus cell (NPC) senescence is a major cause of intervertebral disc degeneration (IVDD). Oxidative stress and reactive oxygen species (ROS) play critical roles in regulating cell senescence. Selenophosphate synthetase 1 (SEPHS1) was reported to play an important role in mitigating oxidative stress in an osteoarthritis (OA) model by reducing the production of ROS, thereby, delaying the occurrence and development of osteoarthritis. In this study, we explored the, hitherto unknown, role of SEPHS1 in IVDD in vitro and in vivo using an interleukin-1β (IL-1β)-induced NPC senescence model and a rat needle puncture IVDD model, respectively. SEPHS1 delayed NPC senescence in vitro by reducing ROS production. Age-related dysfunction was also ameliorated by the overexpression of SEPHS1 and inhibition of the Hippo-Yap/Taz signaling pathway. In vivo experiments revealed that the overexpression of SEPHS1 and inhibition of Hippo-Yap/Taz alleviated IVDD in rats. Moreover, a selenium (Se)-deficient diet and lack of SEPHS1 synergistically aggravated IVDD progression. Taken together, our results demonstrate that SEPHS1 plays a significant role in NPC senescence. Overexpression of SEPHS1 and inhibition of Hippo-Yap/Taz can delay NPC senescence, restore the balance of extracellular matrix metabolism, and attenuate IVDD. SEPHS1 could be a promising therapeutic target for IVDD.NEW & NOTEWORTHY Selenophosphate synthetase 1 (SEPHS1) deficiency leads to an increase in reactive oxygen species levels and in the subsequent activation of the Hippo-Yap/Taz signaling pathway. In the rat model of intervertebral disc degeneration (IVDD), overexpression of SEPHS1 and inhibition of Hippo-YAP/Taz mitigated the progression of disc degeneration indicating the involvement of SEPHS1 in IVDD. SEPHS1 is a promising therapeutic target for IVDD.
Collapse
Affiliation(s)
- Tao Hu
- Department of Orthopedics, Provincial Hospital Affiliated to Anhui Medical University, Hefei, People's Republic of China
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Zhongming Shi
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yongjin Sun
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Feng Hu
- Department of Orthopedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China
| | - Yuluo Rong
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Jia Wang
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Liang Wang
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Wenbin Xu
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Feng Zhang
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Wen-Zhi Zhang
- Department of Orthopedics, Provincial Hospital Affiliated to Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
40
|
Mokhtar NFK, Shun YQ, Raja Nhari RMH, Mohamad NA, Shahidan NM, Warsanah IH, Mohd Hashim A. Nanoplate-based digital PCR for highly sensitive pork DNA detection targeting multi-copy nuclear and mitochondrial genes. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 2024; 41:120-133. [PMID: 38190283 DOI: 10.1080/19440049.2023.2298476] [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: 09/18/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024]
Abstract
The inclusion of ingredients derived from pigs in highly processed consumer products poses a significant challenge for DNA-targeted analytical enforcement, which could be overcome by using digital PCR. However, most species detection methods use digital PCR to target single-copy nuclear genes, which limits their sensitivity. In this work, we examined the performance of a nanoplate-based digital PCR method that targets multi-copy nuclear (MPRE42) and mitochondrial (Cytb) genes. Poor separation of positive and negative partitions, as well as a 'rain effect' were obtained in the porcine-specific MPRE42 assay. Among the optimization strategies examined, the inclusion of restriction enzymes slightly improved the separation of positive and negative partitions, but a more extensive 'rain effect' was observed. The high copy number of the MPRE42 amplicon is hypothesized to contribute to the saturation of the positive signal. In contrast, the porcine-specific Cytb assay achieved perfect separation of positive and negative partitions with no 'rain effect'. This assay can detect as little as 0.4 pg of pork DNA, with a sensitivity of 0.05% (w/w) in a pork-chicken mixture, proving its applicability for detecting pork in meat and meat-based products. For the MPRE42 assay, potential applications in highly degraded products such as gelatin and lard are anticipated.
Collapse
Affiliation(s)
- Nur Fadhilah Khairil Mokhtar
- Laboratory of Halal Science Research, Halal Products Research Institute, Universiti Putra Malaysia, Serdang, Malaysia
| | | | - Raja Mohd Hafidz Raja Nhari
- Laboratory of Halal Science Research, Halal Products Research Institute, Universiti Putra Malaysia, Serdang, Malaysia
| | - Nurhidayatul Asma Mohamad
- Laboratory of Halal Services, Halal Products Research Institute, Universiti Putra Malaysia, Serdang, Malaysia
| | - Nur Maisarah Shahidan
- Laboratory of Halal Science Research, Halal Products Research Institute, Universiti Putra Malaysia, Serdang, Malaysia
| | - Irwan Hanish Warsanah
- Laboratory of Halal Science Research, Halal Products Research Institute, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Amalia Mohd Hashim
- Laboratory of Halal Science Research, Halal Products Research Institute, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
41
|
Hao J, Zhou J, Hu S, Zhang P, Wu H, Yang J, Zhao B, Liu H, Lin H, Chi J, Lou D. RTA 408 ameliorates diabetic cardiomyopathy by activating Nrf2 to regulate mitochondrial fission and fusion and inhibiting NF-κB-mediated inflammation. Am J Physiol Cell Physiol 2024; 326:C331-C347. [PMID: 38047307 DOI: 10.1152/ajpcell.00467.2023] [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: 09/19/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Diabetic cardiomyopathy (dCM) is a major complication of diabetes; however, specific treatments for dCM are currently lacking. RTA 408, a semisynthetic triterpenoid, has shown therapeutic potential against various diseases by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. We established in vitro and in vivo models using high glucose toxicity and db/db mice, respectively, to simulate dCM. Our results demonstrated that RTA 408 activated Nrf2 and alleviated various dCM-related cardiac dysfunctions, both in vivo and in vitro. Additionally, it was found that silencing the Nrf2 gene eliminated the cardioprotective effect of RTA 408. RTA 408 ameliorated oxidative stress in dCM mice and high glucose-exposed H9C2 cells by activating Nrf2, inhibiting mitochondrial fission, exerting anti-inflammatory effects through the Nrf2/NF-κB axis, and ultimately suppressing apoptosis, thereby providing cardiac protection against dCM. These findings provide valuable insights for potential dCM treatments.NEW & NOTEWORTHY We demonstrated first that the nuclear factor erythroid 2-related factor 2 (Nrf2) activator RTA 408 has a protective effect against diabetic cardiomyopathy. We found that RTA 408 could stimulate the nuclear entry of Nrf2 protein, regulate the mitochondrial fission-fusion balance, and redistribute p65, which significantly alleviated the oxidative stress level in cardiomyocytes, thereby reducing apoptosis and inflammation, and protecting the systolic and diastolic functions of the heart.
Collapse
Affiliation(s)
- Jinjin Hao
- Department of Endocrinology, Shaoxing People's Hospital, Shaoxing, China
| | - Jiedong Zhou
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Songqing Hu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Peipei Zhang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Haowei Wu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Juntao Yang
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Bingjie Zhao
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Hanxuan Liu
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Jufang Chi
- Department of Cardiology, Zhuji People's Hospital, Shaoxing, China
| | - Dajun Lou
- Department of Endocrinology, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
42
|
Kim S, Yoon H, Park SK. Butein Increases Resistance to Oxidative Stress and Lifespan with Positive Effects on the Risk of Age-Related Diseases in Caenorhabditis elegans. Antioxidants (Basel) 2024; 13:155. [PMID: 38397753 PMCID: PMC10886231 DOI: 10.3390/antiox13020155] [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: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Butein is a flavonoid found in many plants, including dahlia, butea, and coreopsis, and has both antioxidant and sirtuin-activating activities. In light of the postulated role of free radicals in aging, we examined the effects of butein on aging and on genetic or nutritional models of age-related diseases in Caenorhabditis elegans. Butein showed radical scavenging activity and increased resistance to oxidative stress in Caenorhabditis elegans. The mean lifespan of Caenorhabditis elegans was significantly increased by butein, from 22.7 days in the untreated control to 25.0 days in the butein-treated group. However, the lifespan-extending effect of butein was accompanied by reduced production of progeny as a trade-off. Moreover, the age-related decline in motility was delayed by butein supplementation. Genetic analysis showed that the lifespan-extending effect of butein required the autophagic protein BEC-1 and the transcription factor DAF-16 to regulate stress response and aging. At the genetic level, expression of the DAF-16 downstream target genes hsp-16.2 and sod-3 was induced in butein-treated worms. Butein additionally exhibited a preventive effect in models of age-related diseases. In an Alzheimer's disease model, butein treatment significantly delayed the paralysis caused by accumulation of amyloid-beta in muscle, which requires SKN-1, not DAF-16. In a high-glucose-diet model of diabetes mellitus, butein markedly improved survival, requiring both SKN-1 and DAF-16. In a Parkinson's disease model, dopaminergic neurodegeneration was completely inhibited by butein supplementation and the accumulation of α-synuclein was significantly reduced. These findings suggest the use of butein as a novel nutraceutical compound for aging and age-related diseases.
Collapse
Affiliation(s)
- Seona Kim
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
| | - Hyemin Yoon
- Department of Medical Biotechnology, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
| | - Sang-Kyu Park
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
- Department of Medical Biotechnology, Soonchunhyang University, 22 Soonchunhyang-ro, Asan 31538, Republic of Korea
| |
Collapse
|
43
|
Balan AI, Halațiu VB, Scridon A. Oxidative Stress, Inflammation, and Mitochondrial Dysfunction: A Link between Obesity and Atrial Fibrillation. Antioxidants (Basel) 2024; 13:117. [PMID: 38247541 PMCID: PMC10812976 DOI: 10.3390/antiox13010117] [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: 12/18/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
The adipose tissue has long been thought to represent a passive source of triglycerides and fatty acids. However, extensive data have demonstrated that the adipose tissue is also a major endocrine organ that directly or indirectly affects the physiological functions of almost all cell types. Obesity is recognized as a risk factor for multiple systemic conditions, including metabolic syndrome, type 2 diabetes mellitus, sleep apnea, cardiovascular disorders, and many others. Obesity-related changes in the adipose tissue induce functional and structural changes in cardiac myocytes, promoting a wide range of cardiovascular disorders, including atrial fibrillation (AF). Due to the wealth of epidemiologic data linking AF to obesity, the mechanisms underlying AF occurrence in obese patients are an area of rich ongoing investigation. However, progress has been somewhat slowed by the complex phenotypes of both obesity and AF. The triad inflammation, oxidative stress, and mitochondrial dysfunction are critical for AF pathogenesis in the setting of obesity via multiple structural and functional proarrhythmic changes at the level of the atria. The aim of this paper is to provide a comprehensive view of the close relationship between obesity-induced oxidative stress, inflammation, and mitochondrial dysfunction and the pathogenesis of AF. The clinical implications of these mechanistic insights are also discussed.
Collapse
Affiliation(s)
- Alkora Ioana Balan
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Vasile Bogdan Halațiu
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Alina Scridon
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| |
Collapse
|
44
|
Luo L, An X, Xiao Y, Sun X, Li S, Wang Y, Sun W, Yu D. Mitochondrial-related microRNAs and their roles in cellular senescence. Front Physiol 2024; 14:1279548. [PMID: 38250662 PMCID: PMC10796628 DOI: 10.3389/fphys.2023.1279548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Aging is a natural aspect of mammalian life. Although cellular mortality is inevitable, various diseases can hasten the aging process, resulting in abnormal or premature senescence. As cells age, they experience distinctive morphological and biochemical shifts, compromising their functions. Research has illuminated that cellular senescence coincides with significant alterations in the microRNA (miRNA) expression profile. Notably, a subset of aging-associated miRNAs, originally encoded by nuclear DNA, relocate to mitochondria, manifesting a mitochondria-specific presence. Additionally, mitochondria themselves house miRNAs encoded by mitochondrial DNA (mtDNA). These mitochondria-residing miRNAs, collectively referred to as mitochondrial miRNAs (mitomiRs), have been shown to influence mtDNA transcription and protein synthesis, thereby impacting mitochondrial functionality and cellular behavior. Recent studies suggest that mitomiRs serve as critical sensors for cellular senescence, exerting control over mitochondrial homeostasis and influencing metabolic reprogramming, redox equilibrium, apoptosis, mitophagy, and calcium homeostasis-all processes intimately connected to senescence. This review synthesizes current findings on mitomiRs, their mitochondrial targets, and functions, while also exploring their involvement in cellular aging. Our goal is to shed light on the potential molecular mechanisms by which mitomiRs contribute to the aging process.
Collapse
Affiliation(s)
- Ling Luo
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingna An
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yinghui Xiao
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiguang Sun
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yingzhao Wang
- Department of Neurology, Qianwei Hospital of Jilin Province, Changchun, Jilin, China
| | - Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
45
|
Barba-Aliaga M, Bernal V, Rong C, Zid BM, Alepuz P. eIF5A controls mitoprotein import by relieving ribosome stalling at the TIM50 translocase mRNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572290. [PMID: 38187585 PMCID: PMC10769225 DOI: 10.1101/2023.12.19.572290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The efficient import of nuclear-encoded proteins into mitochondria is crucial for proper mitochondrial function. The conserved translation factor eIF5A is primarily known as an elongation factor which binds ribosomes to alleviate ribosome stalling at sequences encoding polyprolines or combinations of proline with glycine and charged amino acids. eIF5A is known to impact the mitochondrial function across a variety of species although the precise molecular mechanism underlying this impact remains unclear. We found that depletion of eIF5A in yeast drives reduced translation and levels of TCA cycle and oxidative phosphorylation proteins. We further found that loss of eIF5A leads to the accumulation of mitoprotein precursors in the cytosol as well as to the induction of a mitochondrial import stress response. Here we identify an essential polyproline-containing protein as a direct eIF5A target for translation: the mitochondrial inner membrane protein Tim50, which is the receptor subunit of the TIM23 translocase complex. We show how eIF5A directly controls mitochondrial protein import through the alleviation of ribosome stalling along TIM50 mRNA at the mitochondrial surface. Removal of the polyprolines from Tim50 rescues the mitochondrial import stress response, as well as the translation of oxidative phosphorylation reporter genes in an eIF5A loss of function. Overall, our findings elucidate how eIF5A impacts the mitochondrial function by reducing ribosome stalling and facilitating protein translation, thereby positively impacting the mitochondrial import process.
Collapse
Affiliation(s)
- Marina Barba-Aliaga
- Instituto de Biotecnología y Biomedicina (Biotecmed), Universitat de València, 46100 València, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, 46100 València, Spain
| | - Vanessa Bernal
- Instituto de Biotecnología y Biomedicina (Biotecmed), Universitat de València, 46100 València, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, 46100 València, Spain
| | - Cynthia Rong
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, United States
| | - Brian M Zid
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, United States
| | - Paula Alepuz
- Instituto de Biotecnología y Biomedicina (Biotecmed), Universitat de València, 46100 València, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, 46100 València, Spain
| |
Collapse
|
46
|
Xu J, Xing T, Li J, Zhang L, Gao F. Efficacy of creatine nitrate supplementation on redox status and mitochondrial function in pectoralis major muscle of preslaughter transported broilers. Anim Biotechnol 2023; 34:3988-3999. [PMID: 37747460 DOI: 10.1080/10495398.2023.2249957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
This study was purposed to investigate the efficacy of dietary creatine nitrate (CrN) supplementation on redox status and mitochondrial function in pectoralis major (PM) muscle of broilers that experienced preslaughter transport. A total of 288 Arbor Acres broilers (28-day-old) were randomly assigned into five dietary treatments, including a basal diet or the basal diet supplemented with 600 mg/kg guanidinoacetic acid (GAA), 300, 600, or 900 mg/kg CrN for 14 days, respectively. On the transportation day, the basal diet group was divided into two groups on average, resulting in six groups. The control group was transported for 0.5 h and the other groups for 3 h (identified as Control, T3h, GAA600, CrN300, CrN600, and CrN900 group, respectively), and all crates were randomly placed on the truck travelling at an average speed of 80 km/h. Our results showed that GAA600 and CrN treatments decreased the muscle ROS level and MDA content (P < 0.05) and increased the mitochondrial membrane potential (P < 0.001), as well as a higher mRNA expression of avUCP (P < 0.001) and lower mRNA expressions of Nrf2 (P < 0.001), Nrf2 and PGC-1α (P < 0.05) compared with T3h group. Meanwhile, the mRNA and protein expressions of Nrf1, TFAM, and PGC-1α in CrN600 and CrN900 groups were lower than those in the T3h group (P < 0.05). Conclusively, dietary supplementation with GAA and CrN decreased muscle oxidative products and enhanced mitochondrial uncoupling mechanism and mtDNA copy number, which relieved muscle oxidative damage and maintained mitochondrial function.
Collapse
Affiliation(s)
- Jiawen Xu
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, Jiangsu, P.R. China
| | - Tong Xing
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, Jiangsu, P.R. China
| | - Jiaolong Li
- Institute of Agricultural Products Processing, Jiangsu Academy of Agricultural Sciences, Nanjing, P.R. China
| | - Lin Zhang
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, Jiangsu, P.R. China
| | - Feng Gao
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
47
|
Chen CW, Chen LK, Chung YT, Liu SY, Chen SW, Chang YI, Hsieh PS, Juan CC. Cysteine-cysteine Chemokine Receptor Type 5 Plays a Critical Role in Exercise Performance by Regulating Mitochondrial Content in Skeletal Muscle. Inflammation 2023; 46:2089-2101. [PMID: 37436644 DOI: 10.1007/s10753-023-01864-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023]
Abstract
Cysteine-cysteine chemokine receptor type 5 (CCR5) is thought to play an important role in the trafficking of lymphoid cells but has recently also been associated with AMPK signaling pathways that are implicated in energy metabolism in skeletal muscle. We hypothesized that genetic deletions of CCR5 would alter mitochondria content and exercise performance in mice. CCR5-/- and wild-type mice with the same genetic background were subjected to endurance exercise and grip strength tests. The soleus muscle was stained with immunofluorescence for myosin heavy chain 7 (MYH7) and succinate dehydrogenase (SDH) analysis as well as the expression of genes associated with muscle atrophy and mitochondrial oxidative phosphorylation were measured using qPCR. Although there were no differences in the weight of the soleus muscle between the CCR5-/- group and the wild-type mice, the CCR5-/- mice showed the following muscular dysfunctions: (i) decreased MYH7 percentage and cross-section area, (ii) higher myostatin and atrogin-1 mRNA levels, (iii) dropped expression of mitochondrial DNA-encoded electron respiratory chain genes (cytochrome B, cytochrome c oxidase subunit III, and ATP synthase subunit 6) as well as mitochondrial generation genes (PPARγ and PGC-1α), and (iv) lower SDH activity and exercise performance when compared with wild-type mice. In addition, genes associated with mitochondrial biogenesis (PGC-1α, PPARγ, and MFN2) and mitochondrial complex (ND4 and Cytb) were upregulated when the skeletal muscle cell line C2C12 was exposed to cysteine-cysteine chemokine ligand 4 (a ligand of CCR5) in vitro. These findings suggested that attenuation of endurance exercise performance is related to the loss of mitochondrial content and lower SDH activity of soleus muscle in CCR5 knockout mice. The present study provides evidence indicating that the chemokine receptor CCR5 might modulate the skeletal muscle metabolic energy system during exercise.
Collapse
Affiliation(s)
- Chien-Wei Chen
- International Sport Science Master's Program, College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Luen-Kui Chen
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Yi-Ting Chung
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Shui-Yu Liu
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Shuoh-Wen Chen
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Yuan-I Chang
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan
| | - Po-Shiuan Hsieh
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
- Department of Medical Research, Tri-Service General Hospital, Taipei, Taiwan
| | - Chi-Chang Juan
- Institutes of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., 112304, Taipei, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
48
|
Owesny P, Grune T. The link between obesity and aging - insights into cardiac energy metabolism. Mech Ageing Dev 2023; 216:111870. [PMID: 37689316 DOI: 10.1016/j.mad.2023.111870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Obesity and aging are well-established risk factors for a range of diseases, including cardiovascular diseases and type 2 diabetes. Given the escalating prevalence of obesity, the aging population, and the subsequent increase in cardiovascular diseases, it is crucial to investigate the underlying mechanisms involved. Both aging and obesity have profound effects on the energy metabolism through various mechanisms, including metabolic inflexibility, altered substrate utilization for energy production, deregulated nutrient sensing, and mitochondrial dysfunction. In this review, we aim to present and discuss the hypothesis that obesity, due to its similarity in changes observed in the aging heart, may accelerate the process of cardiac aging and exacerbate the clinical outcomes of elderly individuals with obesity.
Collapse
Affiliation(s)
- Patricia Owesny
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
49
|
Wen W, Guo C, Chen Z, Yang D, Zhu D, Jing Q, Zheng L, Sun C, Tang C. Regular exercise attenuates alcoholic myopathy in zebrafish by modulating mitochondrial homeostasis. PLoS One 2023; 18:e0294700. [PMID: 38032938 PMCID: PMC10688687 DOI: 10.1371/journal.pone.0294700] [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: 07/10/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Alcoholic myopathy is caused by chronic consumption of alcohol (ethanol) and is characterized by weakness and atrophy of skeletal muscle. Regular exercise is one of the important ways to prevent or alleviate skeletal muscle myopathy. However, the beneficial effects and the exact mechanisms underlying regular exercise on alcohol myopathy remain unclear. In this study, a model of alcoholic myopathy was established using zebrafish soaked in 0.5% ethanol. Additionally, these zebrafish were intervened to swim for 8 weeks at an exercise intensity of 30% of the absolute critical swimming speed (Ucrit), aiming to explore the beneficial effects and underlying mechanisms of regular exercise on alcoholic myopathy. This study found that regular exercise inhibited protein degradation, improved locomotion ability, and increased muscle fiber cross-sectional area (CSA) in ethanol-treated zebrafish. In addition, regular exercise increases the functional activity of mitochondrial respiratory chain (MRC) complexes and upregulates the expression levels of MRC complexes. Regular exercise can also improve oxidative stress and mitochondrial dynamics in zebrafish skeletal muscle induced by ethanol. Additionally, regular exercise can activate mitochondrial biogenesis and inhibit mitochondrial unfolded protein response (UPRmt). Together, our results suggest regular exercise is an effective intervention strategy to improve mitochondrial homeostasis to attenuate alcoholic myopathy.
Collapse
Affiliation(s)
- Wei Wen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Cheng Guo
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zhanglin Chen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Dong Yang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Danting Zhu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Quwen Jing
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Chenchen Sun
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
- School of Physical Education, Hunan First Normal University, Changsha, Hunan, China
| | - Changfa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| |
Collapse
|
50
|
Stern A, Fokra M, Sarvin B, Alrahem AA, Lee WD, Aizenshtein E, Sarvin N, Shlomi T. Inferring mitochondrial and cytosolic metabolism by coupling isotope tracing and deconvolution. Nat Commun 2023; 14:7525. [PMID: 37980339 PMCID: PMC10657349 DOI: 10.1038/s41467-023-42824-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/19/2023] [Indexed: 11/20/2023] Open
Abstract
The inability to inspect metabolic activities within distinct subcellular compartments has been a major barrier to our understanding of eukaryotic cell metabolism. Previous work addressed this challenge by analyzing metabolism in isolated organelles, which grossly bias metabolic activity. Here, we describe a method for inferring physiological metabolic fluxes and metabolite concentrations in mitochondria and cytosol based on isotope tracing experiments performed with intact cells. This is made possible by computational deconvolution of metabolite isotopic labeling patterns and concentrations into cytosolic and mitochondrial counterparts, coupled with metabolic and thermodynamic modelling. Our approach lowers the uncertainty regarding compartmentalized fluxes and concentrations by one and three orders of magnitude compared to existing modelling approaches, respectively. We derive a quantitative view of mitochondrial and cytosolic metabolic activities in central carbon metabolism across cultured cell lines without performing cell fractionation, finding major variability in compartmentalized malate-aspartate shuttle fluxes. We expect our approach for inferring metabolism at a subcellular resolution to be instrumental for a variety of studies of metabolic dysfunction in human disease and for bioengineering.
Collapse
Affiliation(s)
- Alon Stern
- Department of Computer Science, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Mariam Fokra
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Boris Sarvin
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Ahmad Abed Alrahem
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Won Dong Lee
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Elina Aizenshtein
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Nikita Sarvin
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Tomer Shlomi
- Department of Computer Science, Technion-Israel Institute of Technology, 32000, Haifa, Israel.
- Department of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel.
- Lokey Center for Life Science and Engineering, Technion-Israel Institute of Technology, 32000, Haifa, Israel.
| |
Collapse
|