1
|
Furrer R, Handschin C. Biomarkers of aging: from molecules and surrogates to physiology and function. Physiol Rev 2025; 105:1609-1694. [PMID: 40111763 DOI: 10.1152/physrev.00045.2024] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.
Collapse
|
2
|
Gaesser GA, Hall SE, Angadi SS, Poole DC, Racette SB. Increasing the health span: unique role for exercise. J Appl Physiol (1985) 2025; 138:1285-1308. [PMID: 40244910 DOI: 10.1152/japplphysiol.00049.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
Health span, that period between birth and onset of major disease(s), when adequate physical and cognitive function permit those daily living activities essential to life quality, is lower in the United States than other developed countries. Physical inactivity and excessive calorie intake occupy dominant roles both in the problem, and by redressing them, in the solution. Consequently, this review focuses on evidence that appropriate exercise engagement and calorie restriction (CR) can improve physical and mental health with a view to extending the health span. Humanity, writ large, has grasped these underlying concepts for Millennia but has been largely intransigent to them. Thus, the final section proposes a novel Monty Python-esque approach that encompasses humanity's inimical sense of humor to increase physical fitness and mental health, restore energy balance, sustain better cognitive function, and extend the health span.
Collapse
Affiliation(s)
- Glenn A Gaesser
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States
| | - Stephanie E Hall
- Colleges of Veterinary Medicine and Health and Human Sciences, Kansas State University, Manhattan, Kansas, United States
| | - Siddhartha S Angadi
- School of Health Education and Human Development, University of Virginia, Charlottesville, Virginia, United States
| | - David C Poole
- Colleges of Veterinary Medicine and Health and Human Sciences, Kansas State University, Manhattan, Kansas, United States
| | - Susan B Racette
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States
| |
Collapse
|
3
|
Rong P, Mu Y, Wang M, Chen L, Liu F, Jin Y, Feng W, Zhou K, Liang H, Wang HY, Chen S. Targeting IGF1 to alleviate obesity through regulating energy expenditure and fat deposition. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1662-1675. [PMID: 39843847 DOI: 10.1007/s11427-024-2768-y] [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: 06/13/2024] [Accepted: 10/31/2024] [Indexed: 01/24/2025]
Abstract
Insulin-like growth factor 1 (IGF1) is a regulator of both cellular hypertrophy and lipogenesis, which are two key processes for pathogenesis of obesity. However, the in vivo role of IGF1 in the development of obesity remains unclear. Here, we show that IGF1 expression is increased in adipose tissue in obese human patients and animal models. Elevation of IGF1 is associated with increased lipogenic gene expression and decreased energy expenditure. Genetic down-regulation of IGF1 normalizes lipogenic gene expression, restores aberrant energy metabolism and alleviates obese phenotype of a genetic mouse model with IGF1-hypersecretion. Importantly, genetic down-regulation of IGF1 exerts similar effects on development of diet-induced obesity. Furthermore, berberine that is an AMP-activated protein kinase (AMPK) activator in medicinal herbs inhibits IGF1 secretion, decreases lipogenic gene expression and alleviates diet-induced adiposity. Collectively, our findings demonstrate that hypersecretion of IGF1 is a critical factor for the development of obesity and can be targeted using AMPK activators to alleviate obesity.
Collapse
Affiliation(s)
- Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Yinqiu Mu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Meiqin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Liang Chen
- College of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Fangtong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Yuxin Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Kun Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
4
|
Gjermeni E, Fiebiger R, Bundalian L, Garten A, Schöneberg T, Le Duc D, Blüher M. The impact of dietary interventions on cardiometabolic health. Cardiovasc Diabetol 2025; 24:234. [PMID: 40450314 DOI: 10.1186/s12933-025-02766-w] [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: 03/13/2025] [Accepted: 04/29/2025] [Indexed: 06/03/2025] Open
Abstract
Obesity and cardiometabolic diseases are leading causes of morbidity and mortality among adults worldwide. These conditions significantly contribute to and exacerbate other major causes of illness and death, including cancer, neurodegenerative diseases, and chronic kidney disease. The growing burden of these diseases has increased the interest of modern medicine in understanding metabolic processes and health, with diet emerging as a pivotal modifiable factor, alongside physical inactivity and smoking. In this review, we discuss the pathophysiological and evolutionary foundations of metabolic processes that may link "unhealthy" nutrition to obesity and cardiometabolic diseases and review the current literature to assess the effects of various diet interventions and patterns on cardiometabolic parameters. Special emphasis is placed on summarizing the latest, albeit partially contradictory, evidence to offer balanced dietary recommendations with the ultimate aim to improve cardiometabolic health.
Collapse
Affiliation(s)
- Erind Gjermeni
- Department of Cardiology, Median Center for Rehabilitation Schmannewitz, 04774, Dahlen, Germany.
| | - Raluca Fiebiger
- Department of Cardiology, Median Center for Rehabilitation Schmannewitz, 04774, Dahlen, Germany
| | - Linnaeus Bundalian
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Antje Garten
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103, Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| | - Diana Le Duc
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
- Department of Genetics, Center for Diagnostics at Chemnitz Clinics, 09116, Chemnitz, Germany
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103, Leipzig, Germany
| |
Collapse
|
5
|
van Galen I, Birkisdóttir MB, Ozinga RA, Brandt RMC, Barnhoorn S, Imholz S, van Oostrom CT, van der Marel RWGN, Smit K, Rijksen YMA, Reiling E, van Steeg H, Hoeijmakers JHJ, Dollé MET, Vermeij WP. High protein intake causes gene-length-dependent transcriptional decline, shortens lifespan and accelerates ageing in progeroid DNA repair-deficient mice. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:20. [PMID: 40416846 PMCID: PMC12098121 DOI: 10.1038/s44324-025-00064-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025]
Abstract
Dietary composition can significantly influence health and lifespan, however, robust knowledge on which food components, at what concentration exert which long-term health effects is still incomplete. Here, we explored the effects of dietary protein intake on Ercc1 Δ/- DNA-repair-deficient mice, which are an excellent model for accelerated ageing and are hyperresponsive to the anti-ageing effect of dietary restriction. Restricting dietary protein by 50% extended lifespan in male mice, but not in females. Restricting protein levels beyond 80% improved various neurological health parameters, while a further reduction to 95% affected appetite and became distinctly detrimental. Conversely, a near doubling of protein intake and isocaloric compensatory lowering with carbohydrates significantly shortened lifespan in both sexes. Gene expression analysis of liver from mice on a high-protein, low-carbohydrate diet to those on high-carbohydrate, low-protein revealed increased expression of oxidative phosphorylation, enrichment of processes associated with tissue injury, inflammation, and gene-length-dependent transcriptional decline (GLTD), recently shown to reflect DNA damage accumulation causing transcription stress, and cellular ageing. Finally, GLTD was also identified by reanalysis of publicly available data of wild-type mice, rats and humans on high-protein diets, suggesting that increased dietary protein enhances GLTD and accelerates systemic ageing. Together, our findings have implications for nutritional guidelines for progeroid DNA-repair-deficient human syndromes, warrant the use of excessive protein intake for sustaining health, and suggests GLTD as a sensitive read-out of overall health and predictor of biological ageing.
Collapse
Affiliation(s)
- Ivar van Galen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Laboratory for Experimental Orthopedics, Maastricht University, Maastricht, Netherlands
| | - Maria B. Birkisdóttir
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Rutger A. Ozinga
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Renata M. C. Brandt
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sandra Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Conny T. van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | | | - Kimberly Smit
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Yvonne M. A. Rijksen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Erwin Reiling
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Harry van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Jan H. J. Hoeijmakers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
- CECAD forschungszentrum, Köln, Germany
| | - Martijn E. T. Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Wilbert P. Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
6
|
Pang L, Wang D, Liu H, Zhang M, Yang X, Lu Q, Liu R. Antioxidant and anti-aging effect of queen bee larvae (Apis mellifera) protein hydrolysates in Drosophila melanogaster. Int J Biol Macromol 2025:144306. [PMID: 40383333 DOI: 10.1016/j.ijbiomac.2025.144306] [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: 02/14/2025] [Revised: 04/11/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
With increasing concern on the problem of aging, search for food-derived anti-aging compounds has become a hot research area. This study investigated the anti-aging potential and underlying mechanisms of queen bee larvae enzymatic hydrolysates (QBLE) in Drosophila through measurement of the natural lifespan and the mRNA level of related signaling pathways and widely targeted metabolomics analysis based on UPLC-MS/MS. The results showed that QBLE could prolong the mean, median, and maximum lifespan of Drosophila. Particularly, supplementation of QBLE at 5 mg/mL increased the median and mean lifespan by 12.74 % and 15.04 %, respectively, indicating that QBLE is effective in prolonging lifespan of Drosophila. Moreover, QBLE supplementation significantly improved the climbing ability and gut integrity, as well as effectively reduced MDA accumulation and increased the levels of CAT, GSH, T-SOD, and T-AOC in aged Drosophila. The effect of QBLE to extend the lifespan was jointly influenced by the intrinsic stress protection system (Nrf2/Keap1), TOR pathway (TOR/S6K, PI3K/Akt/TOR), autophagy-related genes (Atg8a, Atg5), and longevity genes (MTH). Furthermore, differential metabolite analysis revealed that QBLE mitigates Drosophila aging by regulating arginine biosynthesis, glycerophosphlipid metabolism, pyrimidine metabolism, purine metabolism, and cysteine, glycine, and methionine metabolism. Overall, these findings suggest that QBLE has great potential to be applied in the health food and biomedicine fields as a novel protein-derived anti-aging agent.
Collapse
Affiliation(s)
- Lili Pang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Danyang Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongshan Liu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Mo Zhang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinbiao Yang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Qun Lu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China.
| | - Rui Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China; Laboratory of Urban Agriculture in Central China, Ministry of Agriculture and Rural Affairs, Wuhan 430070, China.
| |
Collapse
|
7
|
Zhang S, Sun B, Sun L, Zou S, Chen Q. Effect of intermittent fasting on obesity and metabolic indices in patients with metabolic syndrome: a systematic review and meta analysis. BMC Endocr Disord 2025; 25:130. [PMID: 40369509 PMCID: PMC12076832 DOI: 10.1186/s12902-025-01952-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE Dietary patterns play a vital role in the health management of individuals with metabolic syndrome. Many recent studies have shown that intermittent fasting (IF) has better effects, such as improving obesity. Nevertheless, it warrants further investigation to determine which approach is more effective in comparison to continuous energy restriction (CR), particularly when total calorie intake shows minimal variation. Consequently, it is crucial to evaluate the degree of enhancement of the two dietary patterns concerning different aspects of metabolic syndrome. This study presents a meta-analysis of randomized controlled trials (RCTs) aimed at comparing the impacts of IF and CR on obesity and glucolipid metabolism in individuals diagnosed with metabolic syndrome. METHODS In August 2024, a thorough examination of English-language literature was performed across the PubMed, Medline, Embase, and Cochrane Library databases. The meta-analyses was performed according to the established guidelines and reported the results. Weight change, Body Mass Index (BMI) change, and triglyceride (TG) level change were designated as key assessment indicators, while blood pressure, blood glucose, hip circumference, and waist circumference served as supplementary indicators for comparative analysis. RESULT A total of nine studies involving 626 patients were analyzed, focusing on the influence of dietary patterns on obesity, cholesterol levels, and insulin resistance among individuals diagnosed with metabolic syndrome. Both dietary patterns were beneficial for patients with metabolic syndrome. However, IF was better than CRin terms of improvement in obesity over the trial period (mean -1.77, 95% CI [-3.06, -0.48]), and it was more conducive to a reduction in TG levels, which was beneficial in terms of improving insulin resistance (mean -10.16, 95% CI [-18.88, -1.45]). CONCLUSION Given its notable advantages for obesity, lipids, and insulin resistance, along with improved patient adherence, IF may be regarded as a more effective dietary approach for individuals with metabolic syndrome. Nonetheless, the long-term effectiveness still necessitates additional validation. PROSPERO REGISTRATION CRD42024587335.
Collapse
Affiliation(s)
- Shengxuan Zhang
- Department of Cardiology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Bixuan Sun
- Department of Cardiology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Lei Sun
- Department of Gastroenterology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Shijian Zou
- Department of Cardiology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Qilan Chen
- Department of Cardiology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China.
| |
Collapse
|
8
|
Zhang Y, Naaz A, Cheng TYN, Lin JJ, Gao M, Dorajoo R, Alfatah M. Systematic transcriptomics analysis of calorie restriction and rapamycin unveils their synergistic interaction in prolonging cellular lifespan. Commun Biol 2025; 8:753. [PMID: 40369174 PMCID: PMC12078523 DOI: 10.1038/s42003-025-08178-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
Aging is a multifaceted biological process marked by the decline in both mitotic and postmitotic cellular function, often central to the development of age-related diseases. In the pursuit of slowing or even reversing the aging process, a prominent strategy of significant interest is calorie restriction (CR), also known as dietary restriction, and the potential influence of a drug called rapamycin (RM). Both CR and RM have demonstrated the capacity to extend healthspan and lifespan across a diverse array of species, including yeast, worms, flies, and mice. Nevertheless, their individual and combined effects on mitotic and postmitotic cells, as well as their comparative analysis, remain areas that demand a thorough investigation. In this study, we employ RNA-sequencing methodologies to comprehensively analyze the impact of CR, RM, and their combination (CR + RM) on gene expression in yeast cells. Our analysis uncovers distinctive, overlapping, and even contrasting patterns of gene regulation, illuminating the unique and shared effects of CR and RM. Furthermore, the transcriptional synergistic interaction of CR + RM is validated in extending the lifespan of both yeast and human cells.
Collapse
Affiliation(s)
- Yizhong Zhang
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Arshia Naaz
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Trishia Yi Ning Cheng
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jovian Jing Lin
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mingtong Gao
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mohammad Alfatah
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Healthy Longevity, National University Health System, Singapore, Singapore.
| |
Collapse
|
9
|
Mao Z, Chen P, Ji Q, Zhao X, Zhong K, Zeng X. Hydrogen sulfide and ferroptosis inhibition underlies the dietary restriction-induced protection against cyclophosphamide cystitis. Front Pharmacol 2025; 16:1562852. [PMID: 40432898 PMCID: PMC12106363 DOI: 10.3389/fphar.2025.1562852] [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: 01/18/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Dietary restriction (DR) has emerged as a potential therapeutic intervention for various pathological conditions. This study investigated the effects of DR on cyclophosphamide-induced cystitis in mice. Animals were subjected to controlled food restriction for 1 week prior to cyclophosphamide administration. We evaluated changes in body weight, bladder pathology, redox status, and ferroptotic parameters. DR significantly attenuated cyclophosphamide-induced cystitis severity, as evidenced by reduced bladder weight, decreased lipid peroxidation, and diminished ferroptotic markers in bladder tissue. Mechanistic investigations revealed that DR upregulated hepatic hydrogen sulfide (H2S)-synthesizing enzymes and enhanced H2S production. Inhibition of H2S-synthesizing enzymes with DL-propargylglycine (PAG) and aminooxyacetic acid (AOAA) exacerbated cyclophosphamide-induced cystitis, whereas administration of diallyl trisulfide (DATS), an H2S donor, markedly ameliorated bladder pathology. In vitro studies demonstrated that H2S donors, NaHS and DATS, protected against cyclophosphamide metabolite acrolein (ACR)-induced urothelial cell death by suppressing oxidative stress, as indicated by reduced p38 MAPK activation and protein carbonylation. These findings suggest that DR confers protection against cyclophosphamide-induced cystitis through the induction of endogenous H2S production and inhibition of ferroptosis. Our study provides additional evidence supporting the health-promoting effects of DR as well as novel mechanistic insights into the beneficial effects of DR. Given H2S has anti-inflammatory and anti-oxidative properties and that oxidative stress and ferroptosis underlie various diseases, our finding could have broader implications.
Collapse
Affiliation(s)
- Zhimin Mao
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
10
|
Ye D, Feng S, Yang X, Su Y, Zhang J, Feng H, Zhou M, Zhou B, Duan L, Peng T, Wang C. Hedgehog-interacting protein orchestrates alveologenesis and protects against bronchopulmonary dysplasia and emphysema. SCIENCE ADVANCES 2025; 11:eadu2958. [PMID: 40333979 PMCID: PMC12057671 DOI: 10.1126/sciadv.adu2958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025]
Abstract
Most of the lung's gas-exchange surface forms during alveologenesis and its disruption causes bronchopulmonary dysplasia (BPD) in infants, characterized by alveolar simplification and myofibroblast accumulation. BPD also increases the risk of adult emphysema, marked by alveolar loss. Despite this connection, mechanisms linking these conditions and effective treatments are still lacking. We identify hedgehog-interacting protein (HHIP), associated with both BPD and emphysema, as a critical regulator of alveologenesis. During this process, Hhip-expressing cells expanded, accompanied by hedgehog (Hh) signaling inhibition and myofibroblast transition. Stromal-specific Hhip deletion led to hyperactivation of Hh-IGF1 signaling axis, causing persistent SMA+ myofibroblasts and epithelial stem/progenitor cell senescence. Hyperactivation of this pathway was also observed in human BPD and hyperoxia-induced BPD models. Early Hhip deficiency resulted in adult emphysema with myofibroblast accumulation. We developed a therapeutic Fc-fused HHIP protein that mitigated BPD in neonatal mice and prevented adult emphysema. These findings establish HHIP as a critical regulator of alveologenesis and a therapeutic target for BPD and emphysema.
Collapse
Affiliation(s)
- Datian Ye
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Shiyun Feng
- Perfect Life Science Research Institute, Perfect (GuangDong) Co. Ltd., Zhongshan 528402, China
| | - Xinguo Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanjing Su
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Jing Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Haixin Feng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Minqi Zhou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bin Zhou
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lihui Duan
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tien Peng
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chaoqun Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
11
|
Jaćević V, Grujić-Milanović J, Milovanović Z, Milanović S, Nežić L, Amidžić L, Vojinović N, Marković B, Dobričić V, Milosavljević P, Nepovimova E, Kuča K. Determination of paraoxonase activity and prooxidant-antioxidant balance in the brain tissue of rats following subacute administration of different K-oximes. Chem Biol Interact 2025; 416:111539. [PMID: 40334808 DOI: 10.1016/j.cbi.2025.111539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
This study aimed to determine the paraoxonase activity and prooxidant-antioxidant balance in the brain tissue of Wistar rats following subacute treatment with selected K-oximes. Each K-oxime was administered intramuscularly (0.1 LD50/kg) twice per week for four weeks, and 7 days after the last treatment, the paraoxonase activity (PON1), the prooxidant-antioxidant balance (PAB), the levels of superoxide anion radical (O2•-), the concentration of nitrite (NO2-) and the content of free protein thiol groups in the brain homogenates were evaluated. The PON1 and PAB activity were significantly reduced in almost all oxime-treated groups (p < 0.01 and p < 0.001, respectively). The concentrations of O2•- were significantly increased in the obidoxime-, K048-, K074- and K075-treated groups (p < 0.001), while the levels of NO2- was significantly decreased in asoxime-, obidoxime-, K074 and K075-treated rats (p < 0.01, p < 0.001, respectively). The content of Thiol groups was significantly elevated in all oxime-treated groups (p < 0.001). Continuing our previously published data, these results confirmed that applied K-oximes improved the oxidative status and further harmful systemic effects of rats after subacute administration.
Collapse
Affiliation(s)
- Vesna Jaćević
- Department for Experimental Toxicology and Pharmacology, National Poison Control Centre, Military Medical Academy, Crnotravska 17, 11040, Belgrade, Serbia; Medical Faculty of the Military Medical Academy, University of Defence, Crnotravska 17, 11040, Belgrade, Serbia.
| | - Jelica Grujić-Milanović
- University of Belgrade - Institute for Medical Research, National Institute of the Republic of Serbia, Department for Cardiovascular Research, Dr Subotića 4, 11 132 Belgrade, Serbia
| | - Zoran Milovanović
- Special Police Unit, Ministry of Interior, Trebevićka 12/A, 11 030, Belgrade, Serbia
| | - Sladjan Milanović
- University of Belgrade - Institute for Medical Research, National Institute of the Republic of Serbia, Group of Biomechanics, Biomedical Engineering, and Physics of Complex Systems, Dr Subotića 4, 11 000, Belgrade, Serbia
| | - Lana Nežić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Save Mrkalja 14, 78000, Banja Luka, Bosnia and Herzegovina
| | - Ljiljana Amidžić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Save Mrkalja 14, 78000, Banja Luka, Bosnia and Herzegovina; Department of Human Genetics, Faculty of Medicine, University of Banja Luka, Save Mrkalja 14, 78000, Banja Luka, Bosnia and Herzegovina
| | - Nataša Vojinović
- Department of Human Genetics, Faculty of Medicine, University of Banja Luka, Save Mrkalja 14, 78000, Banja Luka, Bosnia and Herzegovina
| | - Bojan Marković
- University of Belgrade - Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Vojvode Stepe 450, 11121, Belgrade, Serbia
| | - Vladimir Dobričić
- University of Belgrade - Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Vojvode Stepe 450, 11121, Belgrade, Serbia
| | - Petar Milosavljević
- Veterinary Services Center, Military Health Department, Crnotravska 17, 11040 Belgrade, Serbia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 70800, Ostrava-Poruba, Czech Republic
| | - Kamil Kuča
- Biomedical Research Center, University Hospital Hradec Kralove, 50005, Hradec Kralove, Czech Republic; Centre for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 70800, Ostrava-Poruba, Czech Republic
| |
Collapse
|
12
|
Cagigas ML, De Ciutiis I, Masedunskas A, Fontana L. Dietary and pharmacological energy restriction and exercise for healthspan extension. Trends Endocrinol Metab 2025:S1043-2760(25)00076-1. [PMID: 40318928 DOI: 10.1016/j.tem.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
Extending healthspan - the years lived in optimal health - holds transformative potential to reduce chronic diseases and healthcare costs. Dietary restriction (DR), particularly when combined with nutrient-rich diets and exercise, is among the most effective, evidence-based strategies for enhancing metabolic health and longevity. By targeting fundamental pathways, it mitigates the onset and progression of obesity, type 2 diabetes (T2D), cardiovascular disease (CVD), neurodegeneration, and cancer. This review synthesizes human data on the impact of DR and exercise on metabolic and age-related diseases, while emphasizing key biological mechanisms such as nutrient sensing, insulin sensitivity, inflammation, mitochondrial function, and gut microbiota. We also examine the emerging role of pharmacologically induced DR, focusing on glucagon-like peptide 1 (GLP-1) receptor agonists (RAs) that partially mimic DR and present opportunities for chronic disease prevention.
Collapse
Affiliation(s)
- Maria Lastra Cagigas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Isabella De Ciutiis
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
13
|
Biswas P, Bako JA, Liston JB, Yu H, Wat LW, Miller CJ, Gordon MD, Huan T, Stanley M, Rideout EJ. Insulin/insulin-like growth factor signaling pathway promotes higher fat storage in Drosophila females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.623936. [PMID: 40342968 PMCID: PMC12060994 DOI: 10.1101/2024.11.18.623936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
In Drosophila , adult females store more fat than males. While the mechanisms that restrict body fat in males are becoming clearer, less is known about how females achieve higher fat storage. Here, we perform a detailed investigation of the mechanisms that promote higher fat storage in females. We show greater intake of dietary sugar supports higher fat storage due to female-biased remodeling of the fat body lipidome. Dietary sugar stimulates a female-specific increase in Drosophila insulin-like peptide 3 (Dilp3), which acts together with greater peripheral insulin sensitivity to augment insulin/insulin-like growth factor signaling pathway (IIS) activity in adult females. Indeed, Dilp3 overexpression prevented the female-biased decrease in body fat after removal of dietary sugar. Given that adult-specific IIS inhibition caused a female-biased decrease in body fat, our data reveal IIS as a key determinant of female fat storage.
Collapse
|
14
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
15
|
Hu C. Prevention of cardiovascular disease for healthy aging and longevity: A new scoring system and related "mechanisms-hallmarks-biomarkers". Ageing Res Rev 2025; 107:102727. [PMID: 40096912 DOI: 10.1016/j.arr.2025.102727] [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: 05/09/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Healthy "environment-sleep-emotion-exercise-diet" intervention [E(e)SEEDi] lifestyle can improve the quality of life, prolong aging and promote longevity due to improvement of human immunity and prevention of cardiovascular diseases (CVD). Here, the author reviewed the associations between these core elements with CVD and cardiovascular aging, and developed a new scoring system based on the healthy E(e)SEEDi lifestyle for prediction and evaluation of life expectancy. These core factors are assigned 20 points each (120 points in total), and a higher score predicts healthier aging and longevity. The E(e)SEEDi represents "a tree of life" bearing the fruits of longevity as well as "a rocket of anti-ageing" carrying people around the world on a journey of longevity. In conclusion, the E(e)SEEDi can delay aging and increase the life expectancy due to the role of a series of cellular and molecular "mechanisms-hallmarks-biomarkers". It's believed that the novel scoring system has a huge potential and beautiful prospects.
Collapse
Affiliation(s)
- Chunsong Hu
- Department of Cardiovascular Medicine, Nanchang University, Hospital of Nanchang University, Jiangxi Academy of Medical Science, No. 461 Bayi Ave, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
16
|
Kilili H, Padilla-Morales B, Castillo-Morales A, Monzón-Sandoval J, Díaz-Barba K, Cornejo-Paramo P, Vincze O, Giraudeau M, Bush SJ, Li Z, Chen L, Mourkas E, Ancona S, Gonzalez-Voyer A, Cortez D, Gutierrez H, Székely T, Acuña-Alonzo AP, Urrutia AO. Maximum lifespan and brain size in mammals are associated with gene family size expansion related to immune system functions. Sci Rep 2025; 15:15087. [PMID: 40301502 PMCID: PMC12041557 DOI: 10.1038/s41598-025-98786-3] [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: 09/17/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
Mammals exhibit an unusual variation in their maximum lifespan potential, measured as the longest recorded longevity of any individual in a species. Evidence suggests that lifespan increases follow expansion in brain size relative to body mass. Here, we found significant gene family size expansions associated with maximum lifespan potential and relative brain size but not in gestation time, age of sexual maturity, and body mass in 46 mammalian species. Extended lifespan is associated with expanding gene families enriched in immune system functions. Our results suggest an association between gene duplication in immune-related gene families and the evolution of longer lifespans in mammals. These findings explore the genomic features linked with the evolution of lifespan in mammals and its association with life story and morphological traits.
Collapse
Affiliation(s)
- Huseyin Kilili
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
| | - Benjamin Padilla-Morales
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK.
| | | | | | - Karina Díaz-Barba
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
- Licenciatura en Ciencias Genómicas, Universidad Nacional Autónoma de México, CP62210, Cuernavaca, Mexico
| | - Paola Cornejo-Paramo
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
- Licenciatura en Ciencias Genómicas, Universidad Nacional Autónoma de México, CP62210, Cuernavaca, Mexico
| | - Orsolya Vincze
- Littoral, Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, 2 Rue Olympe de Gouges, FR-17000, La Rochelle, France
- Institute of Aquatic Ecology, Centre for Ecological Research, 4026, Debrecen, Hungary
- Evolutionary Ecology Group, Hungarian Department of Biology and Ecology, Babeş-Bolyai University, 400006, Cluj-Napoca, Romania
| | - Mathieu Giraudeau
- Littoral, Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, 2 Rue Olympe de Gouges, FR-17000, La Rochelle, France
| | - Stephen J Bush
- School of Automation Science and Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Zhidan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Evangelos Mourkas
- Zoonosis Science Centre, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sergio Ancona
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | | | - Diego Cortez
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, CP62210, Cuernavaca, México
| | - Humberto Gutierrez
- Instituto Nacional de Medicina Genomica, 14610, Ciudad de Mexico, Mexico
| | - Tamás Székely
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK
- Department of Evolutionary Zoology and Human Biology, University of Debrecen, Debrecen, Hungary
| | - Alín P Acuña-Alonzo
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| | - Araxi O Urrutia
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, BA2 7AY, UK.
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
17
|
Huang S, Shi H, Shi Z, Wu J, He L. Vorinostat, a potential hormetin, extends lifespan and enhances stress resistance via the SKN-1 pathway in Caenorhabditis elegans. Biogerontology 2025; 26:97. [PMID: 40278906 DOI: 10.1007/s10522-025-10236-9] [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: 03/03/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025]
Abstract
Vorinostat, a pan histone deacetylases (HDACs) inhibitor clinically approved for cutaneous T-cell lymphoma, exerts therapeutic effects by inducing tumor cell death and cycle arrest. Intriguingly, a previously unrecognized hormetic role of low-dose vorinostat in Caenorhabditis elegans. Subtoxic concentrations of vorinostat (1 μM) significantly extended lifespan, enhanced healthspan, and improved resistance to oxidative and heat stress, while ameliorating Aβ-induced paralysis. qPCR analysis demonstrated dose-dependent bidirectional regulation of stress-resistance genes (sod-3, hsp-16.2, skn-1, gst-4, act-1), with low doses of vorinostat upregulating these genes whereas higher doses (10 μM) exerted suppressive or neutral effects. Mechanistically, vorinostat-induced hormesis required functional SKN-1 signaling, as evidenced by its capacity to activate skn-1 and downstream targets (hsp-16.2, gst-4, act-1). Crucially, RNAi-mediated skn-1 knockdown completely abolished the pro-longevity and stress-resistant phenotypes. These findings establish vorinostat as a novel hormetin that enhances organismal resilience through SKN-1 pathway activation, providing new insights into HDAC inhibitor biology and aging intervention strategies.
Collapse
Affiliation(s)
- Shuai Huang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hang Shi
- Department of Pharmacology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhidan Shi
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiawei Wu
- Department of Gastroenterology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, China.
| | - Ling He
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
18
|
Stefens SJM, van der Linden J, Heredia-Genestar JM, Brandt RMC, Barnhoorn S, Nieuwenhuizen-Bakker I, van Vliet N, Odijk JHM, Ridwan Y, Stuijts D, Batenburg M, Hoeijmakers JHJ, Kanaar R, Essers J, van der Pluijm I. Dietary Restriction Mitigates Vascular Aging, Modulates the cGAS-STING Pathway and Reverses Macrophage-Like VSMC Phenotypes in Progeroid DNA-Repair-Deficient Ercc1 Δ /- Mice. Aging Cell 2025:e70062. [PMID: 40279334 DOI: 10.1111/acel.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/27/2025] Open
Abstract
Aging is a major risk factor for cardiovascular diseases, and the accumulation of DNA damage significantly contributes to the aging process. This study aimed to identify the underlying molecular mechanisms of vascular aging in DNA-repair-deficient progeroid Ercc1Δ/- mice and to explore the therapeutic effect of dietary restriction (DR). RNA sequencing analysis revealed that DR reversed gene expression of vascular aging processes, including extracellular matrix remodeling, in the Ercc1Δ/- aorta. Notably, this analysis indicated the presence of macrophage-like vascular smooth muscle cells (VSMCs) and suggested cGAS-STING pathway activation. The presence of macrophage-like VSMCs and increased STING1 expression were confirmed in Ercc1Δ/- aortic tissue and were both reduced by DR. In vitro, cisplatin-induced DNA damage activated the cGAS-STING pathway in Ercc1Δ/- VSMCs but not in wildtype VSMCs. These findings identify the involvement of the cGAS-STING pathway in DNA damage-driven vascular aging and underscore the therapeutic benefits of DR for vascular aging. Furthermore, upstream regulator analysis revealed compounds that may replicate the beneficial effects of DR, providing promising leads for further investigation.
Collapse
Affiliation(s)
- S J M Stefens
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J van der Linden
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J M Heredia-Genestar
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - R M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - I Nieuwenhuizen-Bakker
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - N van Vliet
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J H M Odijk
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Y Ridwan
- AMIE Core Facility, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - D Stuijts
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Batenburg
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Institute for Genome Stability in Aging and Disease, Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - R Kanaar
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - I van der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Hafeez E, Du D, Ni H, Zhu K, Hu F, Zhou J, Chen D. Regulation and mechanism of Bletilla striata polysaccharide on delaying aging in Drosophila melanogaster. Int J Biol Macromol 2025; 310:143382. [PMID: 40268010 DOI: 10.1016/j.ijbiomac.2025.143382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/12/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Bletilla striata polysaccharide (BSP) is a natural bioactive compound known for its promising health benefits, including antioxidant, immunomodulatory, and anti-inflammatory effects. However, its potential in combating aging remains largely unexplored. This study aims to investigate the anti-aging effects of BSP in the Drosophila melanogaster model. The results show that BSP supplementation significantly extends the lifespan of flies in a concentration-dependent manner, with the most pronounced effects observed at a concentration of 3 mg/mL. Lifespan extension is associated with enhanced antioxidative capacities, as evidenced by increased SOD and CAT activities, and decreased MDA content. Additionally, BSP ameliorates age-related symptoms, including improved climbing ability and enhanced intestinal barrier function. Furthermore, BSP supplementation enhances resistance to H2O2-induced oxidative and starvation stresses, attenuates the lead (Pb)-induced toxicity, and delays the onset of Alzheimer's phenotypes in flies. RNA-Seq analysis reveals that BSP supplementation leads to the differential expression of 992 genes. KEGG pathway analysis highlights significant changes in metabolic pathways, including galactose metabolism, starch and sucrose metabolism, and carbon metabolism. Key genes such as Mal-A1, Amy-d, Men-b, Pgm-1, Mdh1, and Hex-C are downregulated, while CG32026, CG11291, and Ald2 are upregulated. These findings suggest BSP exhibits significant anti-aging and protective properties, making it a potential therapeutic agent.
Collapse
Affiliation(s)
- Eqra Hafeez
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Dongsheng Du
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Hang Ni
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Kai Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Fan Hu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Jihai Zhou
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Dongsheng Chen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China.
| |
Collapse
|
20
|
Jeong DW, Lee DY, Kim SY, Jeoung SW, Zhao D, Knight J, Lam TT, Jin JH, Lee HS, Hochstrasser M, Ryu HY. Auto-sumoylation of the yeast Ubc9 E2 SUMO-conjugating enzyme extends cellular lifespan. Nat Commun 2025; 16:3735. [PMID: 40254622 PMCID: PMC12009981 DOI: 10.1038/s41467-025-58925-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Calorie restriction (CR) provides anti-aging benefits through diverse processes, such as reduced metabolism and growth and increased mitochondrial activity. Although controversy still exists regarding CR-mediated lifespan effects, many researchers are seeking interventions that mimic the effects of CR. Yeast has proven to be a useful model system for aging studies, including CR effects. We report here that yeast adapted through in vitro evolution to the severe cellular stress caused by loss of the Ulp2 SUMO-specific protease exhibit both enhanced growth rates and replicative lifespan, and they have altered gene expression profiles similar to those observed in CR. Notably, in certain evolved ulp2Δ lines, an increase in the auto-sumoylation of Ubc9 E2 SUMO-conjugating enzyme results in altered regulation of multiple targets involved in energy metabolism and translation at both transcriptional and post-translational levels. This increase is essential for the survival of aged cells and CR-mediated lifespan extension. Thus, we suggest that high Ubc9 auto-sumoylation exerts potent anti-aging effects by promoting efficient energy metabolism-driven improvements in cell replication abilities. This potential could be therapeutically explored for the development of promising CR-mimetic strategies.
Collapse
Affiliation(s)
- Dong-Won Jeong
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Do Yoon Lee
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Yeon Kim
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Seok-Won Jeoung
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Dejian Zhao
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - James Knight
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - TuKiet T Lam
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
| | - Jong Hwa Jin
- Osong Medical Innovation Foundation, New Drug Development Center, Cheongju, CT, Republic of Korea
| | - Hyun-Shik Lee
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Mark Hochstrasser
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT, USA.
| | - Hong-Yeoul Ryu
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
21
|
Wang A, Speakman JR. Potential downsides of calorie restriction. Nat Rev Endocrinol 2025:10.1038/s41574-025-01111-1. [PMID: 40247012 DOI: 10.1038/s41574-025-01111-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/19/2025]
Abstract
Although the potential benefits of calorie restriction on human lifespan remain uncertain, it is currently one of the most extensively researched non-genetic approaches to extending both lifespan and healthspan in animals. Calorie restriction offers numerous health benefits, including a reduced incidence of age-related diseases. However, calorie restriction also produces a range of negative effects, which are not fully documented and require further investigation, particularly in humans. As the viability of calorie restriction in humans will depend on the balance of benefits and detrimental effects, it is crucial to understand the nature of these negative effects and what drives them. In this Review, we summarize the effects of calorie restriction on wound healing, hunger, cold sensitivity, bone health, brain size, cognition, reproductive performance and infection, primarily based on studies of rodents with some data from other species and from humans. Overall, the detrimental effects of calorie restriction seem to stem directly from prioritization of vital functions and downregulation or suppression of energy-demanding processes, which helps preserve survival but can also lead to impaired physiological performance and increased vulnerability to stressors. The exact mechanisms underlying these effects remain unclear. Whether it might be possible to engage in calorie restriction but avoid these negative effects remains uncertain.
Collapse
Affiliation(s)
- Anyongqi Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Beijing Life Science Academy, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - John Roger Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China.
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK.
- Institute of Health Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
22
|
Rouskas K, Bocher O, Simistiras A, Emmanouil C, Mantas P, Skoulakis A, Park YC, Dimopoulos A, Glentis S, Kastenmüller G, Zeggini E, Dimas AS. Periodic dietary restriction of animal products induces metabolic reprogramming in humans with effects on cardiometabolic health. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:14. [PMID: 40225784 PMCID: PMC11981922 DOI: 10.1038/s44324-025-00057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Dietary interventions constitute powerful approaches for disease prevention and treatment. However, the molecular mechanisms through which diet affects health remain underexplored in humans. Here, we compare plasma metabolomic and proteomic profiles between dietary states for a unique group of individuals who alternate between omnivory and restriction of animal products for religious reasons. We find that short-term restriction drives reductions in levels of lipid classes and of branched-chain amino acids, not detected in a control group of individuals, and results in metabolic profiles associated with decreased risk for all-cause mortality. We show that 23% of proteins whose levels are affected by dietary restriction are druggable targets and reveal that pro-longevity hormone FGF21 and seven additional proteins (FOLR2, SUMF2, HAVCR1, PLA2G1B, OXT, SPP1, HPGDS) display the greatest magnitude of change. Through Mendelian randomization we demonstrate potentially causal effects of FGF21 and HAVCR1 on risk for type 2 diabetes, of HPGDS on BMI, and of OXT on risk for lacunar stroke. Collectively, we find that restriction-associated reprogramming improves metabolic health and emphasise high-value targets for pharmacological intervention.
Collapse
Affiliation(s)
- Konstantinos Rouskas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
- Institute of Applied Biosciences, Centre for Research & Technology Hellas, Thessaloniki, Greece
| | - Ozvan Bocher
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexandros Simistiras
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Christina Emmanouil
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Panagiotis Mantas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Anargyros Skoulakis
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexandros Dimopoulos
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Stavros Glentis
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine and Health, Munich, Germany
| | - Antigone S. Dimas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
23
|
Wu P, Vandemeulebroucke L, Cai H, Braeckman BP. The Proprotein Convertase BLI-4 Is Required for Axenic Dietary Restriction Mediated Longevity in Caenorhabditis elegans. Aging Cell 2025:e70058. [PMID: 40200707 DOI: 10.1111/acel.70058] [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: 06/17/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
Dietary restriction (DR) is a well-established method for extending lifespan across various species, including C. elegans. Among the different DR regimens, axenic dietary restriction (ADR), in which worms are grown in a nutrient-rich sterile liquid medium, yields the most powerful lifespan extension. However, the molecular mechanisms underlying this longevity phenotype remain largely unexplored. Through a pilot screen of candidate genes, we identified the proprotein convertase BLI-4 as a crucial factor in neurons for modulating lifespan under ADR conditions. BLI-4's role appears to be specific to ADR, as it does not significantly impact longevity under other DR regimens. We further explored the involvement of different bli-4 isoforms and found that isoforms b, f, i and j redundantly contribute to the ADR-mediated lifespan extension, while the bli-4d isoform is mainly involved in development. Proteomics analysis revealed that the loss of BLI-4 function under ADR conditions specifically downregulates GOLG-2, involved in Golgi complex organization. This gene also partially mediates the longevity effects of BLI-4 under ADR conditions. Our findings highlight the importance of neuronal BLI-4 and its downstream targets in regulating lifespan extension induced by ADR in C. elegans.
Collapse
Affiliation(s)
- Ping Wu
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
| | - Lieselot Vandemeulebroucke
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
| | - Huaihan Cai
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
- Overseas Pharmaceuticals, Ltd., Huangpu District, Guangzhou, China
| | - Bart P Braeckman
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Yang Y, Mayo A, Levy T, Raz N, Shenhar B, Jarosz DF, Alon U. Compression of morbidity by interventions that steepen the survival curve. Nat Commun 2025; 16:3340. [PMID: 40199852 PMCID: PMC11978790 DOI: 10.1038/s41467-025-57807-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/26/2025] [Indexed: 04/10/2025] Open
Abstract
Longevity research aims to extend the healthspan while minimizing the duration of disability and morbidity, known as the sickspan. Most longevity interventions in model organisms extend healthspan, but it is not known whether they compress sickspan relative to the lifespan. Here, we present a theory that predicts which interventions compress relative sickspan, based on the shape of the survival curve. Interventions such as caloric restriction that extend mean lifespan while preserving the shape of the survival curve, are predicted to extend the sickspan proportionally, without compressing it. Conversely, a subset of interventions that extend lifespan and steepen the shape of the survival curve are predicted to compress the relative sickspan. We explain this based on the saturating-removal mathematical model of aging, and present evidence from longitudinal health data in mice, Caenorhabditis elegans and Drosophila melanogaster. We apply this theory to identify potential interventions for compressing the sickspan in mice, and to combinations of longevity interventions. This approach offers potential strategies for compressing morbidity and extending healthspan.
Collapse
Affiliation(s)
- Yifan Yang
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
- Center for Interdisciplinary Studies, Westlake University, Hangzhou, Zhejiang, China.
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Naveh Raz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ben Shenhar
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel F Jarosz
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
25
|
Yilmaz Y. Green Tea Mitigates the Hallmarks of Aging and Age-Related Multisystem Deterioration. Aging Dis 2025:AD.2025.0398. [PMID: 40249928 DOI: 10.14336/ad.2025.0398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/20/2025] Open
Abstract
Aging is characterized by progressive multisystem deterioration driven by molecular and cellular mechanisms encapsulated in the twelve hallmarks of aging. Green tea (GT), derived from Camellia sinensis, has garnered significant scientific interest due to its rich polyphenolic composition, particularly epigallocatechin-3-gallate, and its pleiotropic health benefits. In this narrative review, we explored the multifaceted mechanisms through which GT may mitigate the aging hallmarks. Evidence from in vitro, animal, and human studies has shown that GT polyphenols can enhance DNA repair pathways, preserve telomere length, modulate epigenetic aging markers, improve proteostasis and autophagic flux, regulate nutrient-sensing networks, and rejuvenate mitochondrial function. Additionally, GT exhibits anti-inflammatory properties and may restore a physiological gut microbiota composition. Beyond molecular and cellular effects, GT consumption in humans has been associated with improved cognitive function, cardiovascular health, muscle preservation, and metabolic regulation in aging populations. Collectively, these findings highlight GT's potential as a naturally occurring geroscience intervention capable of addressing the interconnected network of aging processes more comprehensively than single-target pharmaceuticals. Future research should focus on optimizing dosing regimens, exploring synergies with other anti-aging strategies, and investigating personalized responses to GT interventions.
Collapse
|
26
|
Fiorito G, Tosti V, Polidoro S, Bertozzi B, Veronese N, Cava E, Spelta F, Piccio L, Early DS, Raftery D, Vineis P, Fontana L. Multi-omic analysis of biological aging biomarkers in long-term calorie restriction and endurance exercise practitioners: A cross-sectional study. Aging Cell 2025; 24:e14442. [PMID: 39692728 PMCID: PMC11984672 DOI: 10.1111/acel.14442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/10/2024] [Accepted: 11/04/2024] [Indexed: 12/19/2024] Open
Abstract
Calorie restriction (CR) and physical exercise (EX) are well-established interventions known to extend health span and lifespan in animal models. However, their impact on human biological aging remains unclear. With recent advances in omics technologies and biological age (BioAge) metrics, it is now possible to assess the impact of these lifestyle interventions without the need for long-term follow-up. This study compared BioAge biomarkers in 41 middle-aged and older adult long-term CR practitioners, 41 age- and sex-matched endurance athletes (EX), and 35 sedentary controls consuming Western diets (WD), through PhenoAge: a composite score derived from nine blood-biomarkers. Additionally, a subset of participants (12 CR, 11 EX, and 12 WD) underwent multi-omic profiling, including DNA methylation and RNAseq of colon mucosa, blood metabolomics, and stool metagenomics. A group of six young WD subjects (yWD) served as a reference for BioAge calculation using Mahalanobis distance across six omic layers. The results demonstrated consistently lower BioAge biomarkers in both CR and EX groups compared to WD controls across all layers. CR participants exhibited lower BioAge in gut microbiome and blood-derived omics, while EX participants had lower BioAge in colon mucosa-derived epigenetic and transcriptomic markers, suggesting potential tissue-specific effects. Multi-omic pathway enrichment analyses suggested both shared and intervention-specific mechanisms, including oxidative stress and basal transcription as common pathways, with ether lipid metabolism uniquely enriched in CR. Despite limitations due to sample size, these findings contribute to the broader understanding of the potential anti-aging effects of CR and EX, offering promising directions for further research.
Collapse
Affiliation(s)
- Giovanni Fiorito
- Clinical Bioinformatics UnitIRCCS Istituto Giannina GasliniGenoaItaly
| | - Valeria Tosti
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Silvia Polidoro
- Department of Translational Medicine (DiMET) CAAD Center for Translational Research on Autoimmune and Allergic DiseaseUniversity of Eastern Piedmont "Amedeo Avogadro"NovaraItaly
| | - Beatrice Bertozzi
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Nicola Veronese
- Geriatric Unit, Department of Internal Medicine and GeriatricsUniversity of PalermoPalermoItaly
| | - Edda Cava
- Unit of Dietetic and Clinical Nutrition, San CamilloForlanini HospitalRomeItaly
| | - Francesco Spelta
- Geriatric Unit, AULSS 9 Scaligera“Mater Salutis” Hospital, LegnagoVeronaItaly
| | - Laura Piccio
- Brain and Mind Centre, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Dayna S. Early
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain MedicineUniversity of WashingtonWashingtonUSA
| | - Paolo Vineis
- MRC‐PHE Centre for Environment and HealthImperial College LondonLondonUK
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Department of EndocrinologyRoyal Prince Alfred HospitalSydneyAustralia
| |
Collapse
|
27
|
Zarroug SHO. Caenorhabditis elegans as in vivo model for the screening of natural plants-derived novel anti-aging compounds: a short introduction. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:577-590. [PMID: 39404185 DOI: 10.1080/10286020.2024.2414189] [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: 04/25/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 03/18/2025]
Abstract
The global aging population highlights the need for effective anti-aging treatments. Natural products show promise, but thorough evaluation requires in vivo models due to the complexity of aging. Ethical concerns are driving a shift from traditional models like rabbits and mice to alternatives such as Caenorhabditis elegans. This microscopic nematode, with its short life cycle, genetic similarities to humans, and cost-effectiveness, is ideal for testing anti-aging compounds. We review studies using C. elegans to assess natural products, suggesting it could serve as a primary model for -evaluating the safety and efficacy of plant-derived anti-aging compounds.
Collapse
Affiliation(s)
- Samah H O Zarroug
- Department of Pharmacology, College of Medicine, Alfaisal University, Riyadh11533, Saudi Arabia
| |
Collapse
|
28
|
Guo Y, Zhang Q, Zhang B, Pan T, Ronan EA, Huffman A, He Y, Inoki K, Liu J, Xu XS. Dietary cinnamon promotes longevity and extends healthspan via mTORC1 and autophagy signaling. Aging Cell 2025; 24:e14448. [PMID: 39760475 PMCID: PMC11984692 DOI: 10.1111/acel.14448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Cinnamon, renowned for its aromatic flavor, represents one of the most widely used spices worldwide. Cinnamon is also considered beneficial to human health with therapeutic potential for treating various diseases, ranging from diabetes and cancer to neurodegenerative diseases. However, the mechanisms underlying cinnamon's health benefits remain elusive. It is also unclear whether cinnamon has any role in aging. Using C. elegans as a model, here we show that feeding worms cinnamaldehyde (CA), the active ingredient in cinnamon oil, prolongs longevity. CA also promotes stress resistance and reduces β-Amyloid toxicity in a C. elegans model of Alzheimer's disease. Mechanistically, CA exerts its beneficial effects through mTORC1 and autophagy signaling. Interestingly, CA promotes longevity by inducing a dietary restriction-like state without affecting food intake, suggesting CA as a dietary restriction mimetic. In human cells, CA exerts a similar effect on mTORC1 and autophagy signaling, suggesting a conserved mechanism. Our results demonstrate that dietary cinnamon promotes both lifespan and healthspan and does so by regulating mTORC1 and autophagy signaling.
Collapse
Affiliation(s)
- Yuling Guo
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Qing Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Bi Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Tong Pan
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Elizabeth A. Ronan
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Anthony Huffman
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
| | - Yongqun He
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
- Unit for Laboratory Animal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Ken Inoki
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Jianfeng Liu
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
- Bioland LaboratoryGuangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangzhouChina
| | - X.Z. Shawn Xu
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
29
|
Viollet B. Calorie restriction plugs lithocholic bile acid into the lysosomal AMPK pathway. LIFE METABOLISM 2025; 4:loaf005. [PMID: 40129483 PMCID: PMC11932140 DOI: 10.1093/lifemeta/loaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 03/26/2025]
Affiliation(s)
- Benoit Viollet
- Université Paris Cité, CNRS, Inserm, U1016, Institut Cochin, 24 rue du faubourg Saint Jacques, Paris 75014, France
| |
Collapse
|
30
|
Popescu I, Lenain M, Rovini E, Ruthsatz M. Lifestyle Interventions and Innovative Approaches for the Management of Neurodegenerative Disorders in Older Adults-State-Of-The-Art and Future Directions. Am J Lifestyle Med 2025:15598276251330170. [PMID: 40161281 PMCID: PMC11954138 DOI: 10.1177/15598276251330170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Aging increases the risk of neurodegenerative diseases (NDs) like Alzheimer's (AD) and Parkinson's (PD), characterized by neuronal loss and cognitive decline. Potential preventive/therapeutic interventions also include lifestyle changes like nutrition & diet, exercise, leisure and social engagement. Here, we discuss several lifestyle interventions for healthy brain aging, and in older adults with NDs. Balanced diets like the Mediterranean and MIND diets can reduce cognitive decline during aging. Long-term use of specific nutrient combinations in medical food may also exert benefits on memory. Metabolic interventions like calorie restriction (CR) and intermittent fasting (IF) have shown potential benefits in aging. However, their effects on cognitive function in older adults were modestly explored and remain unclear. Clinical trials are ongoing to test the cognitive and health outcomes of CR/IF variants in older adults with AD or PD (associated or not with metabolic disorders). We also highlight the role of cognitive reserve (CoR) in delaying dementia symptoms. Engaging in diverse leisure activities like music and bilingualism may enhance CoR and reduce AD risk. Finally, we outline future directions for promoting healthy brain aging through lifestyle interventions (e.g., personalized diets, precision nutrition, synergistic lifestyle approaches, AI-based technologies to monitor the effectiveness of lifestyle practices).
Collapse
Affiliation(s)
- Iuliana Popescu
- Barnstable Brown Diabetes Research Centre, University of Kentucky, Lexington, KY, USA (IP)
| | - Manon Lenain
- Centre de Prévention Santé Longévité (CPSL), Institut Pasteur de Lille, Sciences Cognitives et Sciences Affectives (SCALab), Université de Lille, Lille, France (ML)
| | - Erika Rovini
- Department of Industrial Engineering, University of Florence, Florence, Italy (ER)
| | | |
Collapse
|
31
|
Wang S, Huo T, Lu M, Zhao Y, Zhang J, He W, Chen H. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies. Cells 2025; 14:499. [PMID: 40214453 PMCID: PMC11987807 DOI: 10.3390/cells14070499] [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: 02/27/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Cellular senescence is an irreversible state of cell cycle arrest. Senescent cells (SCs) accumulate in the body with age and secrete harmful substances known as the senescence-associated secretory phenotype (SASP), causing chronic inflammation; at the same time, chronic inflammation leads to a decrease in immune system function, known as immunosenescence, which further accelerates the aging process. Cellular senescence and immunosenescence are closely related to a variety of chronic diseases, including cardiovascular diseases, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Studying the mechanisms of cellular senescence and immunosenescence and developing targeted interventions are crucial for improving the immune function and quality of life of elderly people. Here, we review a series of recent studies focusing on the molecular mechanisms of cellular senescence and immunosenescence, the regulation of aging by the immune system, and the latest advances in basic and clinical research on senolytics. We summarize the cellular and animal models related to aging research, as well as the mechanisms, strategies, and future directions of aging interventions from an immunological perspective, with the hope of laying the foundation for developing novel and practical anti-aging therapies.
Collapse
Affiliation(s)
- Shuaiqi Wang
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Tong Huo
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Mingyang Lu
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Yueqi Zhao
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Jianmin Zhang
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| | - Wei He
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
| | - Hui Chen
- Department of Immunology, CAMS Key Laboratory T-Cell and Cancer Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing 100005, China; (S.W.); (T.H.); (M.L.); (Y.Z.); (J.Z.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
| |
Collapse
|
32
|
Metwally H. STAT Signature Dish: Serving Immunity with a Side of Dietary Control. Biomolecules 2025; 15:487. [PMID: 40305224 PMCID: PMC12024614 DOI: 10.3390/biom15040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunity is a fundamental aspect of animal biology, defined as the host's ability to detect and defend against harmful pathogens and toxic substances to preserve homeostasis. However, immune defenses are metabolically demanding, requiring the efficient allocation of limited resources to balance immune function with other physiological and developmental needs. To achieve this balance, organisms have evolved sophisticated signaling networks that enable precise, context-specific responses to internal and external cues. These networks are essential for survival and adaptation in multicellular systems. Central to this regulatory architecture is the STAT (signal transducer and activator of Transcription) family, a group of versatile signaling molecules that govern a wide array of biological processes across eukaryotes. STAT signaling demonstrates remarkable plasticity, from orchestrating host defense mechanisms to regulating dietary metabolism. Despite its critical role, the cell-specific and context-dependent nuances of STAT signaling remain incompletely understood, highlighting a significant gap in our understanding. This review delves into emerging perspectives on immunity, presenting dynamic frameworks to explore the complexity and adaptability of STAT signaling and the underlying logic driving cellular decision-making. It emphasizes how STAT pathways integrate diverse physiological processes, from immune responses to dietary regulation, ultimately supporting organismal balance and homeostasis.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
33
|
Bulmer C, Avenell A. The effect of dietary weight-loss interventions on the inflammatory markers interleukin-6 and TNF-alpha in adults with obesity: A systematic review and meta-analysis of randomized controlled clinical trials. Obes Rev 2025:e13910. [PMID: 40090867 DOI: 10.1111/obr.13910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/30/2024] [Accepted: 01/04/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND A chronic inflammatory state characterizes a wide range of diseases for which obesity is a risk factor. Weight loss could reduce levels of circulating inflammatory markers potentially reducing the incidence of associated diseases and improving response to treatment. However, dietary weight loss studies have reported inconsistent effects on serum inflammatory makers and the long-term effects are unknown. OBJECTIVE To systematically review randomized controlled trials and analyze any differences in serum interleukin-6 and tumor necrosis factor-alpha between adults with obesity achieving weight loss through dietary intervention compared to those receiving none or standard care. METHODS Studies were identified by searching databases from 1966 to November 2024. Randomized controlled trials with at least 12 months' follow-up were included in this systematic review and meta-analysis with an assessment of Cochrane risk of bias version 1. RESULTS Twelve eligible studies were included. No trials reported a significant effect of weight loss on circulating tumor necrosis factor-alpha, whilst studies achieving greater than 5% weight loss significantly reduced circulating interleukin-6 in adults with obesity. CONCLUSION Weight loss interventions achieving and maintaining greater than 5% weight loss appear to be required to reduce circulating interleukin-6 levels in adults with obesity.
Collapse
Affiliation(s)
- Cate Bulmer
- Health Services Research Unit, University of Aberdeen, UK
| | - Alison Avenell
- Health Services Research Unit, University of Aberdeen, UK
| |
Collapse
|
34
|
Li Q, Xiao N, Zhang H, Liang G, Lin Y, Qian Z, Yang X, Yang J, Fu Y, Zhang C, Liu A. Systemic aging and aging-related diseases. FASEB J 2025; 39:e70430. [PMID: 40022602 DOI: 10.1096/fj.202402479rrr] [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: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Aging is a biological process along with systemic and multiple organ dysfunction. It is more and more recognized that aging is a systemic disease instead of a single-organ functional disorder. Systemic aging plays a profound role in multiple diseases including neurodegenerative diseases, cardiovascular diseases, and malignant diseases. Aged organs communicate with other organs and accelerate aging. Skeletal muscle, heart, bone marrow, skin, and liver communicate with each other through organ-organ crosstalk. The crosstalk can be mediated by metabolites including lipids, glucose, short-chain fatty acids (SCFA), inflammatory cytokines, and exosomes. Metabolic disorders including hyperglycemia, hyperinsulinemia, and hypercholesterolemia caused by chronic diseases accelerate hallmarks of aging. Systemic aging leads to the destruction of systemic hemostasis, causes the release of inflammatory cytokines, senescence-associated secretory phenotype (SASP), and the imbalance of microbiota composition. Released inflammatory factors further aggregate senescence, which promotes the aging of multiple solid organs. Targeting senescence or delaying aging is emerging as a critical health strategy for solving age-related diseases, especially in the old population. In the current review, we will delineate the mechanisms of organ crosstalk in systemic aging and age-related diseases to provide therapeutic targets for delaying aging.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yanguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
35
|
Wang Q, Wang Z, Mizuguchi K, Takao T. Biological age prediction using a DNN model based on pathways of steroidogenesis. SCIENCE ADVANCES 2025; 11:eadt2624. [PMID: 40085695 PMCID: PMC11908500 DOI: 10.1126/sciadv.adt2624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Aging involves the progressive accumulation of cellular damage, leading to systemic decline and age-related diseases. Despite advances in medicine, accurately predicting biological age (BA) remains challenging due to the complexity of aging processes and the limitations of current models. This study introduces a method for predicting BA using a deep neural network (DNN) based on pathways of steroidogenesis. We analyzed 22 steroids from 148 serum samples of individuals aged 20 to 73, using 98 samples for model training and 50 for validation. Our model reflects the often-overlooked fact that aging heterogeneity expands over time and uncovers sex-specific variations in steroidogenesis. This study leveraged key markers, including cortisol (COL), which underscore the role of stress-related and sex-specific steroids in aging. The resulting model establishes a biologically meaningful and robust framework for predicting BA across diverse datasets, offering fresh insights and supporting more targeted strategies in aging research and disease management.
Collapse
Affiliation(s)
| | - Zi Wang
- Corresponding author. (Z.W.); (T.T.)
| | - Kenji Mizuguchi
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Toshifumi Takao
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
36
|
Kumari A, Rahaman A, Zeng XA, Baloch Z. Therapeutic potential and microRNA regulating properties of phytochemicals in Alzheimer's disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102439. [PMID: 40114707 PMCID: PMC11925107 DOI: 10.1016/j.omtn.2024.102439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly and is characterized by the aggregation of Aβ (peptide) and neurofibrillary tangles along with inflammatory processes. Aging is a significant driver of these alterations, and dementia is a major cause of disability and mortality. Despite extensive clinical trials over the past two decades, no effective drug has been developed to improve AD symptoms or slow its progression, indicating the inefficiency of current treatment targets. In AD development, the molecular microenvironment plays a significant role. MicroRNAs (miRNAs) are a key component of this microenvironment, regulate post-transcriptional gene expression, and are expressed more abundantly in the brain than in other tissues. Several dysregulated miRNAs in AD have been linked to neuropathological changes, such as plaque and tangle accrual, as well as altered expression of notorious molecules. Preclinical studies have confirmed the efficacy of phytochemicals/food bioactive compounds (PCs/FBCs) in regulating miRNA expression, which makes them immensely beneficial for targeting miRNA-altered expression patterns in neuronal diseases. This review highlights the potential of miRNAs in driving AD pathology and its development. Furthermore, it discusses the therapeutic efficacy of PCs/FBCs and their miRNA-regulatory properties, especially focusing on antiinflammatory and antioxidant capacities for their development as effective AD agents.
Collapse
Affiliation(s)
- Ankita Kumari
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Abdul Rahaman
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zulqarnain Baloch
- Faculty of Science and Technology, Kunming University of Science and Technology, Kunming, Yunan, China
| |
Collapse
|
37
|
Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, Tuo Z, Wang Z, Li D, Cho WC, Wei W, Feng D. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0612. [PMID: 40046513 PMCID: PMC11880593 DOI: 10.34133/research.0612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/17/2025]
Abstract
The intricate relationship between cancer, circadian rhythms, and aging is increasingly recognized as a critical factor in understanding the mechanisms underlying tumorigenesis and cancer progression. Aging is a well-established primary risk factor for cancer, while disruptions in circadian rhythms are intricately associated with the tumorigenesis and progression of various tumors. Moreover, aging itself disrupts circadian rhythms, leading to physiological changes that may accelerate cancer development. Despite these connections, the specific interplay between these processes and their collective impact on cancer remains inadequately explored in the literature. In this review, we systematically explore the physiological mechanisms of circadian rhythms and their influence on cancer development. We discuss how core circadian genes impact tumor risk and prognosis, highlighting the shared hallmarks of cancer and aging such as genomic instability, cellular senescence, and chronic inflammation. Furthermore, we examine the interplay between circadian rhythms and aging, focusing on how this crosstalk contributes to tumorigenesis, tumor proliferation, and apoptosis, as well as the impact on cellular metabolism and genomic stability. By elucidating the common pathways linking aging, circadian rhythms, and cancer, this review provides new insights into the pathophysiology of cancer and identifies potential therapeutic strategies. We propose that targeting the circadian regulation of cancer hallmarks could pave the way for novel treatments, including chronotherapy and antiaging interventions, which may offer important benefits in the clinical management of cancer.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Fanglin Shao
- Department of Rehabilitation,
The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qing Xin Yu
- Department of Pathology,
Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
- Department of Pathology,
Ningbo Medical Centre Lihuili Hospital, Ningbo, Zhejiang 315040, China
| | - Luxia Ye
- Department of Public Research Platform,
Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA,
Army Medical University, Chongqing, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - William C. Cho
- Department of Clinical Oncology,
Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
- Division of Surgery and Interventional Science,
University College London, London W1W 7TS, UK
| |
Collapse
|
38
|
Prescott J, Keyser AJ, Litwin P, Dunbar MD, McClelland R, Ruple A, Ernst H, Butler BL, Kauffman M, Avery A, Harrison BR, Partida-Aguilar M, McCoy BM, Slikas E, Greenier AK, Muller E, Algavi YM, Bamberger T, Creevy KE, Borenstein E, Snyder-Mackler N, Promislow DEL. Rationale and design of the Dog Aging Project precision cohort: a multi-omic resource for longitudinal research in geroscience. GeroScience 2025:10.1007/s11357-025-01571-3. [PMID: 40038157 DOI: 10.1007/s11357-025-01571-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
A significant challenge in multi-omic geroscience research is the collection of high quality, fit-for-purpose biospecimens from a diverse and well-characterized study population with sufficient sample size to detect age-related changes in physiological biomarkers. The Dog Aging Project designed the precision cohort to study the mechanisms underlying age-related change in the metabolome, microbiome, and epigenome in companion dogs, an emerging model system for translational geroscience research. One thousand dog-owner pairs were recruited into cohort strata based on life stage, sex, size, and geography. We designed and built a novel implementation of the REDCap electronic data capture system to manage study participants, logistics, and biospecimen and survey data collection in a secure online platform. In collaboration with primary care veterinarians, we collected and processed blood, urine, fecal, and hair samples from 976 dogs. The resulting data include complete blood count, chemistry profile, immunophenotyping by flow cytometry, metabolite quantification, fecal microbiome characterization, epigenomic profile, urinalysis, and associated metadata characterizing sample conditions at collection and during lab processing. The project, which has already begun collecting second- and third-year samples from precision cohort dogs, demonstrates that scientifically useful biospecimens can be collected from a geographically dispersed population through collaboration with private veterinary clinics and downstream labs. The data collection infrastructure developed for the precision cohort can be leveraged for future studies. Most important, the Dog Aging Project is an open data project. We encourage researchers around the world to apply for data access and utilize this rich, constantly growing dataset in their own work.
Collapse
Affiliation(s)
- Jena Prescott
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Amber J Keyser
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Paul Litwin
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Matthew D Dunbar
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Robyn McClelland
- Biostatistics and Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Audrey Ruple
- Department of Population Health Science, Virginia Tech, Blacksburg, VA, USA
| | - Holley Ernst
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Brianna L Butler
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Mandy Kauffman
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Anne Avery
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Benjamin R Harrison
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Partida-Aguilar
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Brianah M McCoy
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Elizabeth Slikas
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | | | - Efrat Muller
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Yadid M Algavi
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tal Bamberger
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Kate E Creevy
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Elhanan Borenstein
- Blavatnik School of Computer Science and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel E L Promislow
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA.
| |
Collapse
|
39
|
Gaugel J, Jähnert M, Neumann A, Heyd F, Schürmann A, Vogel H. Alternative splicing landscape in mouse skeletal muscle and adipose tissue: Effects of intermittent fasting and exercise. J Nutr Biochem 2025; 137:109837. [PMID: 39725041 DOI: 10.1016/j.jnutbio.2024.109837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/28/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Alternative splicing contributes to diversify the cellular protein landscape, but aberrant splicing is implicated in many diseases. To which extent mis-splicing contributes to insulin resistance as the causal defect of type 2 diabetes and whether this can be reversed by lifestyle interventions is largely unknown. Therefore, RNA sequencing data from skeletal muscle and adipose tissue of diabetes-susceptible NZO mice treated with or without intermittent fasting and of healthy C57BL/6J mice subjected to exercise were analyzed for alternative splicing differences using Whippet and rMATS. Diet and exercise interventions triggered comparable levels of splicing changes, although the splicing profile of skeletal muscle appeared to be more flexible than that of adipose tissue, with 72-114 differential splicing events in muscle and less than 25 in adipose tissue. Splicing changes induced by time-restricted feeding, alternate-day fasting and exercise were generally mild, with a maximal percent spliced in (PSI) difference of 67%, indicating that alternative splicing plays a rather minor role in lifestyle-induced adaptations of muscle and adipose tissue in mice. However, intron retention contributed to the regulation of gene expression, influencing genes whose expression was directly linked to phenotypic parameters (e.g. Eno2 and Pan2). Alternate-day fasting promoted skipping of exon 7 in Mlxipl (coding for ChREBP), thereby affecting the glucose sensing module of this carbohydrate-responsive transcription factor. Both intermittent fasting and exercise training led to alternative splicing of known diabetes-related GWAS genes (e.g. Abcc8, Ifnar2, Smarcad1), highlighting the potential metabolic relevance of these changes.
Collapse
Affiliation(s)
- Jasmin Gaugel
- Research Group Nutrigenomics of Obesity and Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Research Group Molecular and Clinical Life Science of Metabolic Diseases, Faculty of Health Sciences Brandenburg, University of Potsdam, Brandenburg, Germany
| | - Markus Jähnert
- Research Group Nutrigenomics of Obesity and Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Alexander Neumann
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Omiqa Bioinformatics, Berlin, Germany
| | - Florian Heyd
- Laboratory of RNA Biochemistry, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Annette Schürmann
- Research Group Nutrigenomics of Obesity and Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Heike Vogel
- Research Group Nutrigenomics of Obesity and Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Research Group Molecular and Clinical Life Science of Metabolic Diseases, Faculty of Health Sciences Brandenburg, University of Potsdam, Brandenburg, Germany.
| |
Collapse
|
40
|
Bakowska-Zywicka K, Rzepczak A, Plawgo K, Sobanska D, Tyczewska A. tRNA-Derived Fragments in Age-Related Diseases: A Systematic Review. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70013. [PMID: 40263934 DOI: 10.1002/wrna.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
Aging is a progressive weakening of numerous functions of organisms resulting in diminished abilities to safeguard against environmental damage and augment physiological harmony. It is not a disease in itself; however, it is a main cause of debilitating and life-threatening chronic aging-related diseases (ARDs). tRNA-derived fragments (tDRs) are stable forms of tRNAs of 14-35 nt in length that function as regulatory small-RNA molecules. Here we aimed to perform a systematic review of original articles on the involvement of tDRs in the etiology of ARDs: their identification and characterization. The systematic review was conducted according to the Cochrane Handbook guidelines and the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. Based on the eligibility criteria defined for the study, 21 original articles were included in this systematic review, covering 11 ARDs. The preferred research method used to study tDRs was high-throughput sequencing combined with RT-qPCR, and as a result, a number of tDRs were implicated in ARDs. Importantly, an in-depth analysis of the articles allowed us to identify several shortcomings: (i) the tDRs nomenclature varies between studies and articles, making it often difficult to precisely identify molecules differentiating in a given disease; (ii) the chosen tDRs have all been studied for a miRNA-like mechanism of action; however, tDRs also function in RNAi-independent ways, which need to be studied as well; (iii) to precisely identify tDRs, the sequencing techniques that overcome the issues of modifications harbored by tRNAs must be used.
Collapse
Affiliation(s)
| | - Alicja Rzepczak
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Kinga Plawgo
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Daria Sobanska
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Agata Tyczewska
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
41
|
Wang R, Liao Y, Deng Y, Shuang R. Unraveling the Health Benefits and Mechanisms of Time-Restricted Feeding: Beyond Caloric Restriction. Nutr Rev 2025; 83:e1209-e1224. [PMID: 38954563 DOI: 10.1093/nutrit/nuae074] [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: 07/04/2024] Open
Abstract
Time-restricted feeding (TRF) is a lifestyle intervention that aims to maintain a consistent daily cycle of feeding and fasting to support robust circadian rhythms. Recently, it has gained scientific, medical, and public attention due to its potential to enhance body composition, extend lifespan, and improve overall health, as well as induce autophagy and alleviate symptoms of diseases like cardiovascular diseases, type 2 diabetes, neurodegenerative diseases, cancer, and ischemic injury. However, there is still considerable debate on the primary factors that contribute to the health benefits of TRF. Despite not imposing strict limitations on calorie intake, TRF consistently led to reductions in calorie intake. Therefore, while some studies suggest that the health benefits of TRF are primarily due to caloric restriction (CR), others argue that the key advantages of TRF arise not only from CR but also from factors like the duration of fasting, the timing of the feeding period, and alignment with circadian rhythms. To elucidate the roles and mechanisms of TRF beyond CR, this review incorporates TRF studies that did not use CR, as well as TRF studies with equivalent energy intake to CR, which addresses the previous lack of comprehensive research on TRF without CR and provides a framework for future research directions.
Collapse
Affiliation(s)
- Ruhan Wang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Yan Deng
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Rong Shuang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| |
Collapse
|
42
|
Zhang L, Yu J, Gao X, Yan Y, Wang X, Shi H, Fang M, Liu Y, Kim YB, Zhu H, Wu X, Huang C, Fan S. Targeting farnesoid X receptor as aging intervention therapy. Acta Pharm Sin B 2025; 15:1359-1382. [PMID: 40370561 PMCID: PMC12069902 DOI: 10.1016/j.apsb.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/21/2024] [Accepted: 11/14/2024] [Indexed: 05/16/2025] Open
Abstract
Environmental toxicants have been linked to aging and age-related diseases. The emerging evidence has shown that the enhancement of detoxification gene expression is a common transcriptome marker of long-lived mice, Drosophila melanogaster, and Caenorhabditis elegans. Meanwhile, the resistance to toxicants was increased in long-lived animals. Here, we show that farnesoid X receptor (FXR) agonist obeticholic acid (OCA), a marketed drug for the treatment of cholestasis, may extend the lifespan and healthspan both in C. elegans and chemical-induced early senescent mice. Furthermore, OCA increased the resistance of worms to toxicants and activated the expression of detoxification genes in both mice and C. elegans. The longevity effects of OCA were attenuated in Fxr -/- mice and Fxr homologous nhr-8 and daf-12 mutant C. elegans. In addition, metabolome analysis revealed that OCA increased the endogenous agonist levels of the pregnane X receptor (PXR), a major nuclear receptor for detoxification regulation, in the liver of mice. Together, our findings suggest that OCA has the potential to lengthen lifespan and healthspan by activating nuclear receptor-mediated detoxification functions, thus, targeting FXR may offer to promote longevity.
Collapse
Affiliation(s)
- Lijun Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Yu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoyan Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yingxuan Yan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinyi Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hang Shi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Minglv Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Huanhu Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaojun Wu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
43
|
Lv R, Liu B, Jiang Z, Zhou R, Liu X, Lu T, Bao Y, Huang C, Zou G, Zhang Z, Lu L, Yin Q. Intermittent fasting and neurodegenerative diseases: Molecular mechanisms and therapeutic potential. Metabolism 2025; 164:156104. [PMID: 39674569 DOI: 10.1016/j.metabol.2024.156104] [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: 08/13/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neurodegenerative disorders are straining public health worldwide. During neurodegenerative disease progression, aberrant neuronal network activity, bioenergetic impairment, adaptive neural plasticity impairment, dysregulation of neuronal Ca2+ homeostasis, oxidative stress, and immune inflammation manifest as characteristic pathological changes in the cellular milieu of the brain. There is no drug for the treatment of neurodegenerative disorders, and therefore, strategies/treatments for the prevention or treatment of neurodegenerative disorders are urgently needed. Intermittent fasting (IF) is characterized as an eating pattern that alternates between periods of fasting and eating, requiring fasting durations that vary depending on the specific protocol implemented. During IF, depletion of liver glycogen stores leads to the production of ketone bodies from fatty acids derived from adipocytes, thereby inducing an altered metabolic state accompanied by cellular and molecular adaptive responses within neural networks in the brain. At the cellular level, adaptive responses can promote the generation of synapses and neurons. At the molecular level, IF triggers the activation of associated transcription factors, thereby eliciting the expression of protective proteins. Consequently, this regulatory process governs central and peripheral metabolism, oxidative stress, inflammation, mitochondrial function, autophagy, and the gut microbiota, all of which contribute to the amelioration of neurodegenerative disorders. Emerging evidence suggests that weight regulation significantly contributes to the neuroprotective effects of IF. By alleviating obesity-related factors such as blood-brain barrier dysfunction, neuroinflammation, and β-amyloid accumulation, IF enhances metabolic flexibility and insulin sensitivity, further supporting its potential in mitigating neurodegenerative disorders. The present review summarizes animal and human studies investigating the role and underlying mechanisms of IF in physiology and pathology, with an emphasis on its therapeutic potential. Furthermore, we provide an overview of the cellular and molecular mechanisms involved in regulating brain energy metabolism through IF, highlighting its potential applications in neurodegenerative disorders. Ultimately, our findings offer novel insights into the preventive and therapeutic applications of IF for neurodegenerative disorders.
Collapse
Affiliation(s)
- Renjun Lv
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan 250014, China
| | - Ziying Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Runfa Zhou
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehlstr. 13-17, Mannheim 68167, Germany
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Chunxia Huang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zongyong Zhang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871 Beijing, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
44
|
Stamatakis E, Koemel NA, Biswas RK, Ahmadi MN, Allman-Farinelli M, Trost SG, Inan-Eroglu E, Del Pozo Cruz B, Bin YS, Postnova S, Duncan MJ, Dumuid D, Brown H, Maher C, Fontana L, Simpson S, Cistulli PA. Minimum and optimal combined variations in sleep, physical activity, and nutrition in relation to all-cause mortality risk. BMC Med 2025; 23:111. [PMID: 40001093 PMCID: PMC11863424 DOI: 10.1186/s12916-024-03833-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/18/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Sleep, physical activity, and nutrition (SPAN) are critical behaviours for health, although they have traditionally been studied separately. We examined the combined associations of SPAN and the minimum between-individual variations associated with meaningfully lower all-cause mortality risk. METHODS This prospective cohort analysis included 59,078 participants from the UK Biobank (median age: 64.0 years; 45.4% male) who wore trackers for 7 days and self-reported dietary data. Wearable-measured sleep (hours/day) and moderate to vigorous physical activity (MVPA; mins/day) were calculated using a machine learning based schema. A 10-item diet quality score (DQS) assessed the intake of vegetables, fruits, fish, dairy, whole grains, vegetable oils, refined grains, processed and unprocessed meats, and sugary beverages (0-100 for all components with higher values indicating higher quality). Cox proportional hazards models were used to estimate hazard ratios (HR) for all-cause mortality risk across 27 separate joint tertile combinations of SPAN behaviours with the lowest tertile for all three as the referent group. For more granular clinical interpretations, we examined combined incremental dose-response changes of the SPAN behaviours using the 5th percentile of each behaviour as the referent point. RESULTS Over the 8.1-year median follow-up time, 2,458 mortality events occurred. Compared to the referent group of combined SPAN exposure (lowest tertiles for all three), the optimal SPAN combination involving moderate sleep duration (7.2-8.0 h/day), high MVPA (42-103 min/day), and a DQS between 57.5 and 72.5 was associated with an HR of 0.36 (95% CI: 0.26-0.50). Relative to the 5th percentile of sleep (5.5 h/day), physical activity (7.3 min/day), and nutrition (36.9 DQS), a theoretical minimum combined increase of 15 min/day of sleep, 1.6 min/day MVPA, and 5 DQS points (corresponding to e.g., extra 1/2 serving of vegetables per day or 1 less serving of processed meat per week) was associated with 10% lower all-cause mortality risk (0.90; 0.88-0.93). Combined increases of 75 min/day of sleep, 12.5 min/day MVPA, and 25 DQS points were associated with 50% lower all-cause mortality risk (0.50; 0.44-0.58). CONCLUSIONS This study highlights the potential health value of subtle combined SPAN modification in relation to mortality risk and expands opportunities for more holistic recommendations.
Collapse
Affiliation(s)
- Emmanuel Stamatakis
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Nicholas A Koemel
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Raaj K Biswas
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Matthew N Ahmadi
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Margaret Allman-Farinelli
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Nutrition and Dietetics, School of Nursing, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Stewart G Trost
- School of Human Movement and Nutrition Sciences, The University of Queensland, and Children's Health Queensland, Brisbane, QLD, Australia
| | - Elif Inan-Eroglu
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Borja Del Pozo Cruz
- Faculty of Sports Sciences, Universidad Europea de Madrid, Madrid, Spain
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Yu Sun Bin
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Svetlana Postnova
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Mitch J Duncan
- School of Medicine and Public Health, College of Health, Medicine, and Wellbeing, The University of Newcastle, University Drive, Callaghan, NSW, Australia
- Centre for Active Living and Learning, University of Newcastle, Callaghan, NSW, Australia
| | - Dorothea Dumuid
- Alliance for Research in Exercise, Nutrition and Activity, Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
| | - Helen Brown
- School of Exercise and Nutrition Sciences, Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Carol Maher
- Alliance for Research in Exercise, Nutrition and Activity, Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
| | - Luigi Fontana
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Stephen Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter A Cistulli
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, NSW, Australia
| |
Collapse
|
45
|
Yang HM. Mitochondrial Dysfunction in Cardiovascular Diseases. Int J Mol Sci 2025; 26:1917. [PMID: 40076543 PMCID: PMC11900462 DOI: 10.3390/ijms26051917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Mitochondrial dysfunction is increasingly recognized as a central contributor to the pathogenesis of cardiovascular diseases (CVDs), including heart failure, ischemic heart disease, hypertension, and cardiomyopathy. Mitochondria, known as the powerhouses of the cell, play a vital role in maintaining cardiac energy homeostasis, regulating reactive oxygen species (ROS) production and controlling cell death pathways. Dysregulated mitochondrial function results in impaired adenosine triphosphate (ATP) production, excessive ROS generation, and activation of apoptotic and necrotic pathways, collectively driving the progression of CVDs. This review provides a detailed examination of the molecular mechanisms underlying mitochondrial dysfunction in CVDs, including mutations in mitochondrial DNA (mtDNA), defects in oxidative phosphorylation (OXPHOS), and alterations in mitochondrial dynamics (fusion, fission, and mitophagy). Additionally, the role of mitochondrial dysfunction in specific cardiovascular conditions is explored, highlighting its impact on endothelial dysfunction, myocardial remodeling, and arrhythmias. Emerging therapeutic strategies targeting mitochondrial dysfunction, such as mitochondrial antioxidants, metabolic modulators, and gene therapy, are also discussed. By synthesizing recent advances in mitochondrial biology and cardiovascular research, this review aims to enhance understanding of the role of mitochondria in CVDs and identify potential therapeutic targets to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Han-Mo Yang
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| |
Collapse
|
46
|
Popa AD, Gherasim A, Mihalache L, Arhire LI, Graur M, Niță O. Fasting Mimicking Diet for Metabolic Syndrome: A Narrative Review of Human Studies. Metabolites 2025; 15:150. [PMID: 40137116 PMCID: PMC11943686 DOI: 10.3390/metabo15030150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic syndrome (MetS) is an association of risk factors that share insulin resistance (IR), exerting a super cumulative effect on the risk of developing cardiometabolic diseases. Lifestyle optimization is a key element in the prevention and non-pharmacological therapy of MetS. Certain studies have concluded that some dietary patterns could be more beneficial as an adjunctive treatment for MetS. Fasting mimicking diet (FMD) is a form of periodic fasting in which caloric intake is restricted for 5 days each month. It has been studied for its beneficial effects not only in patients with neoplasia and neurodegenerative diseases but also for its effects on IR and metabolism. In this narrative review, the effects of FMD in patients with MetS were analyzed, focusing on its impact on key metabolic components and summarizing findings from human studies. FMD has demonstrated beneficial effects on MetS by reducing BMI and waist circumference, preserving lean mass, and improving the metabolic profile. Moreover, individuals with a higher BMI or a greater number of MetS components appear to derive greater benefits from this intervention. However, limitations such as high dropout rates, small sample sizes, and methodological constraints restrict the generalizability of current findings. Further large-scale studies are needed to confirm these effects and establish FMD as a viable non-pharmacological strategy for managing MetS.
Collapse
Affiliation(s)
- Alina Delia Popa
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Andreea Gherasim
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Laura Mihalache
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Lidia Iuliana Arhire
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Mariana Graur
- Faculty of Medicine and Biological Sciences, University “Ștefan cel Mare” of Suceava, 720229 Suceava, Romania;
| | - Otilia Niță
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| |
Collapse
|
47
|
Santos M, Moreira JAF, Santos SS, Solá S. Sustaining Brain Youth by Neural Stem Cells: Physiological and Therapeutic Perspectives. Mol Neurobiol 2025:10.1007/s12035-025-04774-z. [PMID: 39985708 DOI: 10.1007/s12035-025-04774-z] [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: 04/09/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
In the last two decades, stem cells (SCs) have attracted considerable interest for their research value and therapeutic potential in many fields, namely in neuroscience. On the other hand, the discovery of adult neurogenesis, the process by which new neurons are generated in the adult brain, challenged the traditional view that the brain is a static structure after development. The recent findings showing that adult neurogenesis has a significant role in brain plasticity, learning and memory, and emotional behavior, together with the fact that it is strongly dependent on several external and internal factors, have sparked more interest in this area. The mechanisms of adult neural stem cell (NSC) regulation, the physiological role of NSC-mediated neuroplasticity throughout life, and the most recent NSC-based therapeutic applications will be concisely reviewed. Noteworthy, due to their multipotency, self-renewal potential, and ability to secrete growth and immunomodulatory factors, NSCs have been mainly suggested for (1) transplantation, (2) neurotoxicology tests, and (3) drug screening approaches. The clinical trials of NSC-based therapy for different neurologic conditions are, nonetheless, mostly in the early phases and have not yet demonstrated conclusive efficacy or safety. Here, we provide an outlook of the major challenges and limitations, as well as some promising directions that could help to move toward stem cell widespread use in the treatment and prevention of several neurological disorders.
Collapse
Affiliation(s)
- Matilde Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - João A Ferreira Moreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Sónia Sá Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal.
| |
Collapse
|
48
|
Gao X, Yu J, Li Y, Shi H, Zhang L, Fang M, Liu Y, Huang C, Fan S. 27-Hydroxymangiferolic Acid Extends Lifespan and Improves Neurodegeneration in Caenorhabditis elegans by Activating Nuclear Receptors. Molecules 2025; 30:1010. [PMID: 40076235 PMCID: PMC11902184 DOI: 10.3390/molecules30051010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
27-Hydroxymangiferolic acid (27-HMA) is a naturally occurring compound in mango fruits that exhibits diverse biological functions. Here, we show that 27-HMA activates the transcriptional activity of farnesoid X receptor (FXR), a nuclear receptor transcription factor, extending the lifespan and healthspan in Caenorhabditis elegans (C. elegans). Meanwhile, the longevity-promoting effect of 27-HMA was attenuated in the mutants of nhr-8 and daf-12, the FXR homologs, indicating that the longevity effects of 27-HMA in C. elegans may depend on nuclear hormone receptors (NHRs). Further analysis revealed potential associations between the longevity effects of 27-HMA and the insulin/insulin-like growth factor-1 signaling (IIS)/TORC1 pathway. Moreover, 27-HMA increased the toxin resistance of nematodes and activated the expression of detoxification genes, which rely on NHRs. Finally, 27-HMA improved the age-related neurodegeneration in Alzheimer's disease (AD) and Parkinson's disease (PD) C. elegans models. Taken together, our findings suggest that 27-HMA is a novel FXR agonist and may prolong lifespan and healthspan via activating NHRs.
Collapse
Affiliation(s)
- Xiaoyan Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Jing Yu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Yin Li
- School of Agriculture and Medicine, Hebei Open University, Shijiazhuang 050080, China;
| | - Hang Shi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Lijun Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Minglv Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (X.G.); (J.Y.); (H.S.); (L.Z.); (M.F.); (Y.L.)
| |
Collapse
|
49
|
Yasuda K. The usefulness of Caenorhabditis elegans lifespan analysis in screening for functional foods. Biosci Biotechnol Biochem 2025; 89:319-323. [PMID: 39592222 DOI: 10.1093/bbb/zbae178] [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: 09/14/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024]
Abstract
Caenorhabditis elegans is a type of nematode that has significantly contributed to aging research as a multicellular animal model because of its high reproductive rate, ease of cultivation, low cost, short generation cycle, body transparency, and eukaryotic nature. Since the discovery of long-lived mutant strains of C. elegans, signaling pathways involved in lifespan have been elucidated. Some of these pathways are shared with mammals, indicating that aging research in C. elegans may be applied to other animals, including humans. Studies on the mechanisms of aging have advanced with the availability of mutants for these pathways. In recent years, C. elegans has also contributed to the discovery of antioxidants and the elucidation of the molecular mechanisms responsible for the anti-aging effects of foods and traditional medicines with lifespan as an indicator. This review summarizes the characteristics of C. elegans for lifespan analysis associated with functional foods.
Collapse
Affiliation(s)
- Kayo Yasuda
- Department of Health Management, Undergraduate School of Health Studies, Tokai University, 4-1-1 Kitakaname, Hiratsuka-shi, Kanagawa 259-1292, Japan
| |
Collapse
|
50
|
Zhang X, Zhang L, Xiang W. The impact of mitochondrial dysfunction on ovarian aging. J Transl Med 2025; 23:211. [PMID: 39980008 PMCID: PMC11844166 DOI: 10.1186/s12967-025-06223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
IMPORTANCE Ovarian aging has become a focal point in current research on female aging and refers to the gradual decline in ovarian function as women age. Numerous factors influence ovarian aging, among which mitochondrial function is one because it plays a crucial role by affecting oocytes and granulosa cells. Mitochondrial deterioration not only leads to a decrease in oocyte quality but also hinders follicle development, further impacting women's reproductive health and fertility. OBJECTIVE This review summarizes and integrates research on the impact of mitochondrial function on ovarian aging, outlining the mechanisms by which mitochondria regulate the functions of oocytes and granulosa cells. This study aims to provide potential therapeutic directions to mitigate mitochondrial decline and support female reproductive health. EVIDENCE REVIEW According to a 2023 study published in Cell, factors such as oxidative stress, mitochondrial dysfunction, chronic inflammation, and telomere shortening collectively drive ovarian aging, directly affecting female fertility. Among these factors, mitochondrial dysfunction plays a key role. This study reviewed literature from databases such as PubMed, Google Scholar, and CNKI, using keywords such as "mitochondrial dysfunction", "decline in oocyte quality and quantity", and "ovarian aging", aiming to summarize current research on the mechanisms of the impact of mitochondrial dysfunction on ovarian aging and provide theoretical support for future exploration of related therapeutic strategies. FINDINGS The main characteristics of ovarian aging include a decline in oocyte quantity and quality, fluctuations in hormone levels, and a reduction in granulosa cell function. Studies have shown that mitochondria affect fertility by regulating cellular energy metabolism, exacerbating oxidative stress, causing mitochondrial DNA (mtDNA) damage, and impacting the physiological function of granulosa cells within the ovary, gradually diminishing the ovarian reserve. CONCLUSION This review focuses on analyzing the effects of mitochondrial decline on energy production in oocytes and granulosa cells, the accumulation of reactive oxygen species (ROS), and the calcium ion (Ca2+) concentration, which all contribute to the ovarian aging process, and understanding them will provide new insights into the mechanisms of ovarian aging. RELEVANCE Therapeutic interventions targeting mitochondrial dysfunction may help delay ovarian aging and improve female reproductive health.
Collapse
Affiliation(s)
- Xiaoyue Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Wenpei Xiang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|