1
|
Mattson MP. The cyclic metabolic switching theory of intermittent fasting. Nat Metab 2025; 7:665-678. [PMID: 40087409 DOI: 10.1038/s42255-025-01254-5] [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: 09/25/2024] [Accepted: 02/19/2025] [Indexed: 03/17/2025]
Abstract
Intermittent fasting (IF) and ketogenic diets (KDs) have recently attracted much attention in the scientific literature and in popular culture and follow a longer history of exercise and caloric restriction (CR) research. Whereas IF involves cyclic metabolic switching (CMS) between ketogenic and non-ketogenic states, KDs and CR may not. In this Perspective, I postulate that the beneficial effects of IF result from alternating between activation of adaptive cellular stress response pathways during the fasting period, followed by cell growth and plasticity pathways during the feeding period. Thereby, I establish the cyclic metabolic switching (CMS) theory of IF. The health benefits of IF may go beyond those seen with continuous CR or KDs without CMS owing to the unique interplay between the signalling functions of the ketone β-hydroxybutyrate, mitochondrial adaptations, reciprocal activation of autophagy and mTOR pathways, endocrine and paracrine signalling, gut microbiota, and circadian biology. The CMS theory may have important implications for future basic research, clinical trials, development of pharmacological interventions, and healthy lifestyle practices.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Zhang X, Li Z, Wang S, Chen Y. Distinct Fgf21 Expression Patterns in Various Tissues in Response to Different Dietary Regimens Using a Reporter Mouse Model. Nutrients 2025; 17:1179. [PMID: 40218937 PMCID: PMC11990235 DOI: 10.3390/nu17071179] [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: 03/05/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Fibroblast growth factor 21 (FGF21), a secreted protein, plays a crucial role in regulating metabolism and energy homeostasis. Nevertheless, the expression pattern of Fgf21 across diverse tissues and its responsiveness to various dietary regimens remain incompletely understood. Methods: In this study, we developed a Fgf21-enhanced green fluorescent protein (EGFP) reporter mouse model to explore the expression of endogenous Fgf21 in different tissues under four dietary conditions: normal chow, low-protein diet, fasting, and fasting-refeeding. Results: A low-protein diet was found to induce Fgf21 expression in both the liver and skeletal muscle. Notably, Fgf21 was predominantly expressed in the periportal region of the liver. In the pancreas, Fgf21 exhibited a patchy expression pattern in the exocrine portion, but was absent in the endocrine part, regardless of the dietary regimens. Regarding the spleen, fasting triggered the expression of Fgf21, which was mainly localized in the red pulp area. Moreover, under fasting conditions, Fgf21 showed a scattered expression pattern in the small intestine. Conclusions: The Fgf21-EGFP reporter mouse model serves as a valuable tool for dissecting the expression of endogenous Fgf21 in different tissues under various dietary and stress conditions. Further investigations using this model may contribute to uncovering the hitherto unrecognized functions of locally produced FGF21.
Collapse
Affiliation(s)
- Xinhui Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zixuan Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuying Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
3
|
Jiang Q, Liu J, Huang S, Wang XY, Chen X, Liu GH, Ye K, Song W, Masters CL, Wang J, Wang YJ. Antiageing strategy for neurodegenerative diseases: from mechanisms to clinical advances. Signal Transduct Target Ther 2025; 10:76. [PMID: 40059211 PMCID: PMC11891338 DOI: 10.1038/s41392-025-02145-7] [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: 08/01/2024] [Revised: 11/29/2024] [Accepted: 01/15/2025] [Indexed: 05/13/2025] Open
Abstract
In the context of global ageing, the prevalence of neurodegenerative diseases and dementia, such as Alzheimer's disease (AD), is increasing. However, the current symptomatic and disease-modifying therapies have achieved limited benefits for neurodegenerative diseases in clinical settings. Halting the progress of neurodegeneration and cognitive decline or even improving impaired cognition and function are the clinically meaningful goals of treatments for neurodegenerative diseases. Ageing is the primary risk factor for neurodegenerative diseases and their associated comorbidities, such as vascular pathologies, in elderly individuals. Thus, we aim to elucidate the role of ageing in neurodegenerative diseases from the perspective of a complex system, in which the brain is the core and peripheral organs and tissues form a holistic network to support brain functions. During ageing, the progressive deterioration of the structure and function of the entire body hampers its active and adaptive responses to various stimuli, thereby rendering individuals more vulnerable to neurodegenerative diseases. Consequently, we propose that the prevention and treatment of neurodegenerative diseases should be grounded in holistic antiageing and rejuvenation means complemented by interventions targeting disease-specific pathogenic events. This integrated approach is a promising strategy to effectively prevent, pause or slow down the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Qiu Jiang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jie Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Shan Huang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xuan-Yue Wang
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Xiaowei Chen
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
- Brain Research Center, Third Military Medical University, Chongqing, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weihong Song
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province. Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China.
| |
Collapse
|
4
|
Nicolaisen TS, Lyster AE, Sjøberg KA, Haas DT, Voldstedlund CT, Lundsgaard AM, Jensen JK, Madsen EM, Nielsen CK, Bloch-Ibenfeldt M, Wewer Albrechtsen NJ, Rose AJ, Krahmer N, Clemmensen C, Richter EA, Fritzen AM, Kiens B. Dietary protein restriction elevates FGF21 levels and energy requirements to maintain body weight in lean men. Nat Metab 2025; 7:602-616. [PMID: 40050437 PMCID: PMC11946896 DOI: 10.1038/s42255-025-01236-7] [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: 04/03/2024] [Accepted: 02/07/2025] [Indexed: 03/28/2025]
Abstract
Dietary protein restriction increases energy expenditure and enhances insulin sensitivity in mice. However, the effects of a eucaloric protein-restricted diet in healthy humans remain unexplored. Here, we show in lean, healthy men that a protein-restricted diet meeting the minimum protein requirements for 5 weeks necessitates an increase in energy intake to uphold body weight, regardless of whether proteins are replaced with fats or carbohydrates. Upon reverting to the customary higher protein intake in the following 5 weeks, energy requirements return to baseline levels, thus preventing weight gain. We also show that fasting plasma FGF21 levels increase during protein restriction. Proteomic analysis of human white adipose tissue and in FGF21-knockout mice reveal alterations in key components of the electron transport chain within white adipose tissue mitochondria. Notably, in male mice, these changes appear to be dependent on FGF21. In conclusion, we demonstrate that maintaining body weight during dietary protein restriction in healthy, lean men requires a higher energy intake, partially driven by FGF21-mediated mitochondrial adaptations in adipose tissue.
Collapse
Affiliation(s)
- Trine S Nicolaisen
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aslak E Lyster
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kim A Sjøberg
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Daniel T Haas
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian T Voldstedlund
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie Lundsgaard
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk A/S, Novo Nordisk, Søborg, Denmark
| | - Jakob K Jensen
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ea M Madsen
- Novo Nordisk A/S, Novo Nordisk, Søborg, Denmark
| | - Casper K Nielsen
- Center for Clinical Metabolic Research, Gentofte Hospital, Hellerup, Denmark
| | - Mads Bloch-Ibenfeldt
- Institute of Sports Medicine Copenhagen (ISMC), Department of Orthopedic Surgery M81, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | | | - Adam J Rose
- Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Andreas M Fritzen
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
5
|
Senior AM, Raubenheimer D, Couteur DGL, Simpson SJ. The Geometric Framework for Nutrition and Its Application to Rodent Models. Annu Rev Anim Biosci 2025; 13:389-410. [PMID: 39546416 DOI: 10.1146/annurev-animal-111523-102327] [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: 11/17/2024]
Abstract
Rodents have been the primary model for mammalian nutritional physiology for decades. Despite an extensive body of literature, controversies remain around the effects of specific nutrients and total energy intake on several aspects of nutritional biology, even in this well-studied model. One approach that is helping to bring clarity to the field is the geometric framework for nutrition (GFN). The GFN is a multidimensional paradigm that can be used to conceptualize nutrition and nutritional effects, design experiments, and interpret results. To date, more than 30 publications have applied the GFN to data from rodent models of nutrition. Here we review the major conclusions from these studies. We pay particular attention to the effects of macronutrients on satiety, glucose metabolism, lifespan and the biology of aging, reproductive function, immune function, and the microbiome. We finish by highlighting several knowledge gaps that became evident upon reviewing this literature.
Collapse
Affiliation(s)
- Alistair M Senior
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - David Raubenheimer
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - David G Le Couteur
- ANZAC Research Institute, The Concord Hospital, Concord, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - Stephen J Simpson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| |
Collapse
|
6
|
Calubag MF, Ademi I, Grunow I, Breuer L, Babygirija R, Lialios P, Le S, Minton D, Sonsalla MM, Illiano J, Knopf BA, Xiao F, Konopka AR, Harris DA, Lamming DW. Tissue-specific effects of dietary protein on cellular senescence are mediated by branched-chain amino acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632607. [PMID: 39868338 PMCID: PMC11761368 DOI: 10.1101/2025.01.13.632607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Dietary protein is a key regulator of healthy aging in both mice and humans. In mice, reducing dietary levels of the branched-chain amino acids (BCAAs) recapitulates many of the benefits of a low protein diet; BCAA-restricted diets extend lifespan, reduce frailty, and improve metabolic health, while BCAA supplementation shortens lifespan, promotes obesity, and impairs glycemic control. Recently, high protein diets have been shown to promote cellular senescence, a hallmark of aging implicated in many age-related diseases, in the liver of mice. Here, we test the hypothesis that the effects of high protein diets on metabolic health and on cell senescence are mediated by BCAAs. We find that reducing dietary levels of BCAAs protects male and female mice from the negative metabolic consequences of both normal and high protein diets. Further, we identify tissue-specific effects of BCAAs on cellular senescence, with restriction of all three BCAAs - but not individual BCAAs - protecting from hepatic cellular senescence while potentiating cell senescence in white adipose tissue. We find that the effects of BCAAs on hepatic cellular senescence are cell-autonomous, with lower levels of BCAAs protecting cultured cells from antimycin-A induced senescence. Our results demonstrate a direct effect of a specific dietary component on a hallmark of aging and suggest that cellular senescence may be highly susceptible to dietary interventions.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ismail Ademi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Lucia Breuer
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Penelope Lialios
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Sandra Le
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Dennis Minton
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia Illiano
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Bailey A Knopf
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Fan Xiao
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David A Harris
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
7
|
Wang X, Zhang G. The mitochondrial integrated stress response: A novel approach to anti-aging and pro-longevity. Ageing Res Rev 2025; 103:102603. [PMID: 39608727 DOI: 10.1016/j.arr.2024.102603] [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: 08/16/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The ISR is a cellular signaling pathway that responds to various physiological changes and types of stimulation. The mitochondrial integrated stress response (ISRmt) is a stress response specific to mitochondria which is initiated by eIF2α phosphorylation and is responsive to mitochondrial stressors. The ISRmt triggers diverse metabolic responses reliant on activating transcription factor 4 (ATF4). The preliminary phases of ISRmt can provoke an adaptive stress response that antagonizes age-related diseases and promotes longevity. In this review, we provide an overview of the molecular mechanisms of the ISRmt, with a particular focus on its potential as a therapeutic target for age-related disease and the promotion of longevity.
Collapse
Affiliation(s)
- Xiaoding Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China.
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China.
| |
Collapse
|
8
|
Yeh CY, Chini LCS, Davidson JW, Garcia GG, Gallagher MS, Freichels IT, Calubag MF, Rodgers AC, Green CL, Babygirija R, Sonsalla MM, Pak HH, Trautman ME, Hacker TA, Miller RA, Simcox JA, Lamming DW. Late-life protein or isoleucine restriction impacts physiological and molecular signatures of aging. NATURE AGING 2024; 4:1760-1771. [PMID: 39604703 PMCID: PMC11672203 DOI: 10.1038/s43587-024-00744-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024]
Abstract
Restricting the intake of protein or the branched-chain amino acid isoleucine promotes healthspan and extends lifespan in young or adult mice. However, their effects when initiated in aged animals are unknown. Here we investigate the consequences of consuming a diet with 67% reduction of all amino acids (low AA) or of isoleucine alone (low Ile), in male and female C57BL/6J.Nia mice starting at 20 months of age. Both dietary regimens effectively promote overall metabolic health without reducing calorie intake. Both low AA and low Ile diets improve aspects of frailty and slow multiple molecular indicators of aging rate; however, the low Ile diet reduces grip strength in both sexes and has mixed, sexually dimorphic effects on the heart. These results demonstrate that low AA and low Ile diets can promote aspects of healthy aging in aged mice and suggest that similar interventions might promote healthy aging in older adults.
Collapse
Affiliation(s)
- Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Lucas C S Chini
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jessica W Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Gonzalo G Garcia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Meredith S Gallagher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac T Freichels
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Allison C Rodgers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Judith A Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin-Madison Comprehensive Diabetes Center, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
9
|
Volcko KL, Taghipourbibalan H, McCutcheon JE. Intermittent protein restriction elevates food intake and plasma ghrelin in male mice. Appetite 2024; 203:107671. [PMID: 39265824 DOI: 10.1016/j.appet.2024.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
Low-protein diets affect body weight, body composition, food intake, and food preferences in mice. Furthermore, single periods of protein restriction can have lasting effects on these parameters. We sought to examine the effect of multiple, short, bouts of protein restriction, relative to long-term maintenance on either a control (NR) or protein-restricted (PR) diet. We found that male mice experiencing intermittent protein restriction (IPR) were indistinguishable from NR mice in terms of body weight and composition, but had food intake and plasma ghrelin as high as mice on PR diet, even when they were returned to control diet. This was not found in female mice. The results of this experiment highlight the importance of diet history on food intake and ghrelin levels in male mice, and the difference in how PR diet might affect male and female mice.
Collapse
Affiliation(s)
- K L Volcko
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway.
| | - H Taghipourbibalan
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway
| | - J E McCutcheon
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
10
|
Kip P, Sluiter TJ, MacArthur MR, Tao M, Kruit N, Mitchell SJ, Jung J, Kooijman S, Gorham J, Seidman JG, Quax PHA, Decano JL, Aikawa M, Ozaki CK, Mitchell JR, de Vries MR. Preoperative methionine restriction induces perivascular adipose tissue browning and improves vein graft remodeling in male mice. Nat Commun 2024; 15:9652. [PMID: 39511181 PMCID: PMC11544300 DOI: 10.1038/s41467-024-53844-8] [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: 12/07/2023] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Short-term preoperative methionine restriction (MetR) is a promising translatable strategy to mitigate surgical injury response. However, its application to improve post-interventional vascular remodeling remains underexplored. Here we find that MetR protects from arterial intimal hyperplasia in a focal stenosis model and pathologic vascular remodeling following vein graft surgery in male mice. RNA sequencing reveals that MetR enhances browning in arterial (thoracic aorta) perivascular adipose tissue (PVAT) and induces it in venous (caval vein) PVAT. Specifically, Ppara is highly upregulated in PVAT-adipocytes upon MetR. Furthermore, MetR dampens the postoperative pro-inflammatory response to surgery in PVAT-macrophages in vivo and in vitro. This study shows that the detrimental effects of dysfunctional PVAT on vascular remodeling can be reversed by MetR, and identifies pathways involved in MetR-induced browning of PVAT. Furthermore, we demonstrate the potential of short-term preoperative MetR as a simple intervention to ameliorate vascular remodeling after vascular surgery.
Collapse
Affiliation(s)
- Peter Kip
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Einthoven Laboratory for Experimental Vascular Medicine and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Thijs J Sluiter
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Einthoven Laboratory for Experimental Vascular Medicine and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael R MacArthur
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Ming Tao
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicky Kruit
- Einthoven Laboratory for Experimental Vascular Medicine and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sarah J Mitchell
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Jonathan Jung
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Josh Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Julius L Decano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - C Keith Ozaki
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James R Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Margreet R de Vries
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Einthoven Laboratory for Experimental Vascular Medicine and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
11
|
Loo J, Gunasekaran G, Tan JK, Goon JA. Elucidating the effective age for dietary restriction and the key metabolites involved. Exp Gerontol 2024; 197:112601. [PMID: 39362416 DOI: 10.1016/j.exger.2024.112601] [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: 06/04/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Dietary restriction (DR) extends lifespan in various species, but its effect at different ages, especially when started later, is unclear. This study used Caenorhabditis elegans to explore the impact of DR at different ages. Worms were divided into control and DR groups, with daily survival monitored. To confirm the occurrence of DR, the expression of DR-sensitive genes namely acdh-1, pyk-1, pck-2 and cts-1 were determined using RT-qPCR. Liquid chromatography mass spectrometry (LC-MS) was employed to observe the changes in metabolites affected by DR. The results indicated that young worms subjected to mild DR displayed the longest lifespan, highlighting the effectiveness of initiating DR at a young age. Increased expression of acdh-1 and pck-2 suggests activation of beta-oxidation and gluconeogenesis, while decreased cts-1 expression indicates a reduced citric acid cycle, further supporting the observed effects of DR in these worms. Metabolomic results indicated that DR decreased the activity of mechanistic Target of Rapamycin (mTOR) and the synthesis of amino acids namely leucine, tyrosine and tryptophan to conserve energy for cell repair and survival. DR also decreased levels of N-acetyl-L-methionine and S-adenosyl-methionine (SAM) in methionine metabolism, thereby promoting autophagy, reducing inflammation, and facilitating the removal of damaged cells and proteins. In conclusion, initiating dietary restriction early in life extends the lifespan by modulating amino acid metabolism and enhancing the autophagy pathway, thereby maintaining cellular wellbeing.
Collapse
Affiliation(s)
- Jazween Loo
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Geetha Gunasekaran
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Jen Kit Tan
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Jo Aan Goon
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
Elmansi AM, Miller RA. Oxidative phosphorylation and fatty acid oxidation in slow-aging mice. Free Radic Biol Med 2024; 224:246-255. [PMID: 39153667 DOI: 10.1016/j.freeradbiomed.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Oxidative metabolism declines with aging in humans leading to multiple metabolic ailments and subsequent inflammation. In mice, there is evidence of age-related suppression of fatty acid oxidation and oxidative phosphorylation in the liver, heart, and muscles. Many interventions that extend healthy lifespan of mice have been developed, including genetic, pharmacological, and dietary interventions. In this article, we review the literature on oxidative metabolism changes in response to those interventions. We also discuss the molecular pathways that mediate those changes, and their potential as targets for future longevity interventions.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA; University of Michigan Geriatrics Center, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA; University of Michigan Geriatrics Center, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Lamming DW. Quantification of healthspan in aging mice: introducing FAMY and GRAIL. GeroScience 2024; 46:4203-4215. [PMID: 38755467 PMCID: PMC11336093 DOI: 10.1007/s11357-024-01200-5] [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: 04/24/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
The population around the world is graying, and as many of these individuals will spend years suffering from the burdens of age associated diseases, understanding how to increase healthspan, defined as the period of life free from disease and disability, is an urgent priority of geroscience research. The lack of agreed-upon quantitative metrics for measuring healthspan in aging mice has slowed progress in identifying interventions that do not simply increase lifespan, but also healthspan. Here, we define FAMY (Frailty-Adjusted Mouse Years) and GRAIL (Gauging Robust Aging when Increasing Lifespan) as new summary statistics for quantifying healthspan in mice. FAMY integrates lifespan data with longitudinal measurements of a widely utilized clinical frailty index, while GRAIL incorporates these measures and also adds information from widely utilized healthspan assays and the hallmarks of aging. Both metrics are conceptually similar to quality-adjusted life years (QALY), a widely utilized measure of disease burden in humans, and can be readily calculated from data acquired during longitudinal and cross-sectional studies of mouse aging. We find that interventions generally thought to promote health, including calorie restriction, robustly improve healthspan as measured by FAMY and GRAIL. Finally, we show that the use of GRAIL provides new insights, and identify dietary restriction of protein or isoleucine as interventions that robustly promote healthspan but not longevity in female HET3 mice. We suggest that the routine integration of these measures into studies of aging in mice will allow the identification and development of interventions that promote healthy aging even in the absence of increased lifespan.
Collapse
Affiliation(s)
- Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA.
| |
Collapse
|
14
|
Wilson RB, Chen YJ, Zhang R, Maini S, Andrews TS, Wang R, Borradaile NM. Elongation factor 1A1 inhibition elicits changes in lipid droplet size, the bulk transcriptome, and cell type-associated gene expression in MASLD mouse liver. Am J Physiol Gastrointest Liver Physiol 2024; 327:G608-G622. [PMID: 39136056 PMCID: PMC11482270 DOI: 10.1152/ajpgi.00276.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/19/2024]
Abstract
Eukaryotic elongation factor 1A1 (EEF1A1), originally identified for its role in protein synthesis, has additional functions in diverse cellular processes. Of note, we previously discovered a role for EEF1A1 in hepatocyte lipotoxicity. We also demonstrated that a 2-wk intervention with the EEF1A1 inhibitor didemnin B (DB) (50 µg/kg) decreased liver steatosis in a mouse model of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) [129S6/SvEvTac mice fed Western diet (42% fat) for 26 wk]. Here, we further characterized the hepatic changes occurring in these mice by assessing lipid droplet (LD) size, bulk differential expression, and cell type-associated alterations in gene expression. Consistent with the previously demonstrated decrease in hepatic steatosis, we observed decreased median LD size in response to DB. Bulk RNA sequencing (RNA-Seq) followed by gene set enrichment analysis revealed alterations in pathways related to energy metabolism and proteostasis in DB-treated mouse livers. Deconvolution of bulk data identified decreased cell type association scores for cholangiocytes, mononuclear phagocytes, and mesenchymal cells in response to DB. Overrepresentation analyses of bulk data using cell type marker gene sets further identified hepatocytes and cholangiocytes as the primary contributors to bulk differential expression in response to DB. Thus, we show that chemical inhibition of EEF1A1 decreases hepatic LD size and decreases gene expression signatures associated with several liver cell types implicated in MASLD progression. Furthermore, changes in hepatic gene expression were primarily attributable to hepatocytes and cholangiocytes. This work demonstrates that EEF1A1 inhibition may be a viable strategy to target aspects of liver biology implicated in MASLD progression.NEW & NOTEWORTHY Chemical inhibition of EEF1A1 decreases hepatic lipid droplet size and decreases gene expression signatures associated with liver cell types that contribute to MASLD progression. Furthermore, changes in hepatic gene expression are primarily attributable to hepatocytes and cholangiocytes. This work highlights the therapeutic potential of targeting EEF1A1 in the setting of MASLD, and the utility of RNA-Seq deconvolution to reveal valuable information about tissue cell type composition and cell type-associated gene expression from bulk RNA-Seq data.
Collapse
Affiliation(s)
- Rachel B Wilson
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Yun Jin Chen
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Richard Zhang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Siddhant Maini
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Tallulah S Andrews
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rennian Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Nica M Borradaile
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
15
|
Chhabra Y, Bielefeldt-Ohmann H, Brooks TL, Brooks AJ, Waters MJ. Roles of Growth Hormone-Dependent JAK-STAT5 and Lyn Kinase Signaling in Determining Lifespan and Cancer Incidence. Endocrinology 2024; 165:bqae136. [PMID: 39378329 PMCID: PMC11500606 DOI: 10.1210/endocr/bqae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 10/10/2024]
Abstract
In rodents, loss of growth hormone (GH) or its receptor is associated with extended lifespan. We aimed to determine the signaling process resulting in this longevity using GH receptor (GHR)-mutant mice with key signaling pathways deleted and correlate this with cancer incidence and expression of genes associated with longevity. GHR uses both canonical janus kinase (JAK)2-signal transducer and activator of transcription (STAT) signaling as well as signaling via the LYN-ERK1/2 pathway. We used C57BL/6 mice with loss of key receptor tyrosines and truncation resulting in 1) loss of most STAT5 response to GH; 2) total inability to generate STAT5 to GH; 3) loss of Box1 to prevent activation of JAK2 but not LYN kinase; or 4) total knockout of the receptor. For each mutant we analyzed lifespan, histopathology to determine likely cause of death, and hepatic gene and protein expression. The extended lifespan is evident in the Box1-mutant males (retains Lyn activation), which have a median lifespan of 1016 days compared to 890 days for the Ghr-/- males. In the females, GhrBox1-/- mice have a median lifespan of 970 days compared to 911 days for the knockout females. Sexually dimorphic GHR-STAT5 is repressive for longevity, since its removal results in a median lifespan of 1003 days in females compared to 734 days for wild-type females. Numerous transcripts related to insulin sensitivity, oxidative stress response, and mitochondrial function are regulated by GHR-STAT5; however, LYN-responsive genes involve DNA repair, cell cycle control, and anti-inflammatory response. There appears to be a yin-yang relationship between JAK2 and LYN that determines lifespan.
Collapse
Affiliation(s)
- Yash Chhabra
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
- Faculty of Medicine, Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Helle Bielefeldt-Ohmann
- School of Chemistry & Molecular Biosciences, University of Queensland, St Lucia 4069, Australia
| | - Tania Louise Brooks
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
| | - Andrew James Brooks
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
- Faculty of Medicine, Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Michael J Waters
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
| |
Collapse
|
16
|
Calubag MF, Robbins PD, Lamming DW. A nutrigeroscience approach: Dietary macronutrients and cellular senescence. Cell Metab 2024; 36:1914-1944. [PMID: 39178854 PMCID: PMC11386599 DOI: 10.1016/j.cmet.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Cellular senescence, a process in which a cell exits the cell cycle in response to stressors, is one of the hallmarks of aging. Senescence and the senescence-associated secretory phenotype (SASP)-a heterogeneous set of secreted factors that disrupt tissue homeostasis and promote the accumulation of senescent cells-reprogram metabolism and can lead to metabolic dysfunction. Dietary interventions have long been studied as methods to combat age-associated metabolic dysfunction, promote health, and increase lifespan. A growing body of literature suggests that senescence is responsive to diet, both to calories and specific dietary macronutrients, and that the metabolic benefits of dietary interventions may arise in part through reducing senescence. Here, we review what is currently known about dietary macronutrients' effect on senescence and the SASP, the nutrient-responsive molecular mechanisms that may mediate these effects, and the potential for these findings to inform the development of a nutrigeroscience approach to healthy aging.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul D Robbins
- Institute On the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
17
|
Wang W, Qian J, Shang M, Qiao Y, Huang J, Gao X, Ye Z, Tong X, Xu K, Li X, Liu Z, Zhou L, Zheng S. Integrative analysis of the transcriptome and metabolome reveals the importance of hepatokine FGF21 in liver aging. Genes Dis 2024; 11:101161. [PMID: 39022127 PMCID: PMC11252782 DOI: 10.1016/j.gendis.2023.101161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 07/20/2024] Open
Abstract
Aging is a contributor to liver disease. Hence, the concept of liver aging has become prominent and has attracted considerable interest, but its underlying mechanism remains poorly understood. In our study, the internal mechanism of liver aging was explored via multi-omics analysis and molecular experiments to support future targeted therapy. An aged rat liver model was established with d-galactose, and two other senescent hepatocyte models were established by treating HepG2 cells with d-galactose and H2O2. We then performed transcriptomic and metabolomic assays of the aged liver model and transcriptome analyses of the senescent hepatocyte models. In livers, genes related to peroxisomes, fatty acid elongation, and fatty acid degradation exhibited down-regulated expression with aging, and the hepatokine Fgf21 expression was positively correlated with the down-regulation of these genes. In senescent hepatocytes, similar to the results found in aged livers, FGF21 expression was also decreased. Moreover, the expressions of cell cycle-related genes were significantly down-regulated, and the down-regulated gene E2F8 was the key cell cycle-regulating transcription factor. We then validated that FGF21 overexpression can protect against liver aging and that FGF21 can attenuate the declines in the antioxidant and regenerative capacities in the aging liver. We successfully validated the results from cellular and animal experiments using human liver and blood samples. Our study indicated that FGF21 is an important target for inhibiting liver aging and suggested that pharmacological prevention of the reduction in FGF21 expression due to aging may be used to treat liver aging-related diseases.
Collapse
Affiliation(s)
- Wenchao Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Mingge Shang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Xinxin Gao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Zhou Ye
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Xinyu Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Kangdi Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Xiang Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Zhengtao Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310000, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
18
|
Kosakamoto H, Sakuma C, Okada R, Miura M, Obata F. Context-dependent impact of the dietary non-essential amino acid tyrosine on Drosophila physiology and longevity. SCIENCE ADVANCES 2024; 10:eadn7167. [PMID: 39213345 PMCID: PMC11364096 DOI: 10.1126/sciadv.adn7167] [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: 03/11/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Dietary protein intake modulates growth, reproduction, and longevity by stimulating amino acid (AA)-sensing pathways. Essential AAs are often considered as limiting nutrients during protein scarcity, and the role of dietary non-essential AAs (NEAAs) is less explored. Although tyrosine has been reported to be crucial for sensing protein restriction in Drosophila larvae, its effect on adult physiology and longevity remains unclear. Here, using a synthetic diet, we perform a systematic investigation of the effect of single NEAA deprivation on nutrient-sensing pathways, reproductive ability, starvation resistance, feeding behavior, and life span in adult female flies. Specifically, dietary tyrosine deprivation decreases internal tyrosine levels and fecundity, influences AA-sensing machineries, and extends life span. These nutritional responses are not observed under higher total AA intake or in infertile female flies, suggesting a context-dependent influence of dietary tyrosine. Our findings highlight the unique role of tyrosine as a potentially limiting nutrient, underscoring its value for dietary interventions aimed at enhancing health span.
Collapse
Affiliation(s)
- Hina Kosakamoto
- RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Chisako Sakuma
- RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Rina Okada
- RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Fumiaki Obata
- RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
19
|
Lee AH, Orliaguet L, Youm YH, Maeda R, Dlugos T, Lei Y, Coman D, Shchukina I, Andhey S, Smith SR, Ravussin E, Stadler K, Hyder F, Artyomov MN, Sugiura Y, Dixit VD. Cysteine depletion triggers adipose tissue thermogenesis and weight-loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606880. [PMID: 39149397 PMCID: PMC11326254 DOI: 10.1101/2024.08.06.606880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Dietary interventions such as caloric restriction (CR)1 and methionine restriction2 that prolong lifespan induce the 'browning' of white adipose tissue (WAT), an adaptive metabolic response that increases heat production to maintain health3,4. However, how diet influences adipose browning and metabolic health is unclear. Here, we identified that weight-loss induced by CR in humans5 reduces cysteine concentration in WAT suggesting depletion of this amino-acid may be involved in metabolic benefits of CR. To investigate the role of cysteine on organismal metabolism, we created a cysteine-deficiency mouse model in which dietary cysteine was eliminated and cystathionine γ-lyase (CTH)6, the enzyme that synthesizes cysteine was conditionally deleted. Using this animal model, we found that systemic cysteine-depletion causes drastic weight-loss with increased fat utilization and browning of adipose tissue. The restoration of dietary cysteine in cysteine-deficient mice rescued weight loss together with reversal of adipose browning and increased food-intake in an on-demand fashion. Mechanistically, cysteine deficiency induced browning and weight loss is dependent on sympathetic nervous system derived noradrenaline signaling via β3-adrenergic-receptors and does not require UCP1. Therapeutically, in high-fat diet fed obese mice, one week of cysteine-deficiency caused 30% weight-loss and reversed inflammation. These findings thus establish that cysteine is essential for organismal metabolism as removal of cysteine in the host triggers adipose browning and rapid weight loss.
Collapse
Affiliation(s)
- Aileen H. Lee
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Lucie Orliaguet
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yun-Hee Youm
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Tamara Dlugos
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yuanjiu Lei
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, School of Engineering and Applied Science, Yale University
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University
| | - Irina Shchukina
- Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sairam Andhey
- Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, School of Engineering and Applied Science, Yale University
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University
| | - Maxim N. Artyomov
- Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Vishwa Deep Dixit
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
20
|
Wahl D, Risen SJ, Osburn SC, Emge T, Sharma S, Gilberto VS, Chatterjee A, Nagpal P, Moreno JA, LaRocca TJ. Nanoligomers targeting NF-κB and NLRP3 reduce neuroinflammation and improve cognitive function with aging and tauopathy. J Neuroinflammation 2024; 21:182. [PMID: 39068433 PMCID: PMC11283709 DOI: 10.1186/s12974-024-03182-9] [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: 02/13/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Neuroinflammation contributes to impaired cognitive function in brain aging and neurodegenerative disorders like Alzheimer's disease, which is characterized by the aggregation of pathological tau. One major driver of both age- and tau-associated neuroinflammation is the NF-κB and NLRP3 signaling axis. However, current treatments targeting NF-κB or NLRP3 may have adverse/systemic effects, and most have not been clinically translatable. In this study, we tested the efficacy of a novel, nucleic acid therapeutic (Nanoligomer) cocktail specifically targeting both NF-κB and NLRP3 in the brain for reducing neuroinflammation and improving cognitive function in old (aged 19 months) wildtype mice, and in rTg4510 tau pathology mice (aged 2 months). We found that 4 weeks of NF-κB/NLRP3-targeting Nanoligomer treatment strongly reduced neuro-inflammatory cytokine profiles in the brain and improved cognitive-behavioral function in both old and rTg4510 mice. These effects of NF-κB/NLRP3-targeting Nanoligomers were also associated with reduced glial cell activation and pathology, favorable changes in transcriptome signatures of glia-associated inflammation (reduced) and neuronal health (increased), and positive systemic effects. Collectively, our results provide a basis for future translational studies targeting both NF-κB and NLRP3 in the brain, perhaps using Nanoligomers, to inhibit neuroinflammation and improve cognitive function with aging and neurodegeneration.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| | - Sydney J Risen
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Brain Research Center, Colorado State University, Fort Collins, CO, USA
| | - Shelby C Osburn
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| | - Tobias Emge
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| | - Sadhana Sharma
- Sachi Bio, Colorado Technology Center, Louisville, CO, USA
| | | | | | | | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Brain Research Center, Colorado State University, Fort Collins, CO, USA
| | - Thomas J LaRocca
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA.
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
21
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
22
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Wahl D, Risen SJ, Osburn SC, Emge T, Sharma S, Gilberto VS, Chatterjee A, Nagpal P, Moreno JA, LaRocca TJ. Nanoligomers targeting NF-κB and NLRP3 reduce neuroinflammation and improve cognitive function with aging and tauopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.03.578493. [PMID: 38370618 PMCID: PMC10871285 DOI: 10.1101/2024.02.03.578493] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Neuroinflammation contributes to impaired cognitive function in brain aging and neurodegenerative disorders like Alzheimer's disease, which is characterized by the aggregation of pathological tau. One major driver of both age- and tau-associated neuroinflammation is the NF-κB and NLRP3 signaling axis. However, current treatments targeting NF-κB or NLRP3 may have adverse/systemic effects, and most have not been clinically translatable. In this study, we tested the efficacy of a novel, nucleic acid therapeutic (Nanoligomer) cocktail specifically targeting both NF-κB and NLRP3 in the brain for reducing neuroinflammation and improving cognitive function in old (aged 19 months) wildtype mice, and in rTg4510 tau pathology mice (aged 2 months). We found that 4 weeks of NF-κB/NLRP3-targeting Nanoligomer treatment strongly reduced neuro-inflammatory cytokine profiles in the brain and improved cognitive-behavioral function in both old and rTg4510 mice. These effects of NF-κB/NLRP3-targeting Nanoligomers were also associated with reduced glial cell activation and pathology, favorable changes in transcriptome signatures of glia-associated inflammation (reduced) and neuronal health (increased), and positive systemic effects. Collectively, our results provide a basis for future translational studies targeting both NF-κB and NLRP3 in the brain, perhaps using Nanoligomers, to inhibit neuroinflammation and improve cognitive function with aging and neurodegeneration.
Collapse
|
24
|
Mishra M, Wu J, Kane AE, Howlett SE. The intersection of frailty and metabolism. Cell Metab 2024; 36:893-911. [PMID: 38614092 PMCID: PMC11123589 DOI: 10.1016/j.cmet.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/15/2024]
Abstract
On average, aging is associated with unfavorable changes in cellular metabolism, which are the processes involved in the storage and expenditure of energy. However, metabolic dysregulation may not occur to the same extent in all older individuals as people age at different rates. Those who are aging rapidly are at increased risk of adverse health outcomes and are said to be "frail." Here, we explore the links between frailty and metabolism, including metabolic contributors and consequences of frailty. We examine how metabolic diseases may modify the degree of frailty in old age and suggest that frailty may predispose toward metabolic disease. Metabolic interventions that can mitigate the degree of frailty in people are reviewed. New treatment strategies developed in animal models that are poised for translation to humans are also considered. We suggest that maintaining a youthful metabolism into older age may be protective against frailty.
Collapse
Affiliation(s)
- Manish Mishra
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Judy Wu
- Institute for Systems Biology, Seattle, WA, USA
| | - Alice E Kane
- Institute for Systems Biology, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada; Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
25
|
Li S, Zou T, Chen J, Li J, You J. Fibroblast growth factor 21: An emerging pleiotropic regulator of lipid metabolism and the metabolic network. Genes Dis 2024; 11:101064. [PMID: 38292170 PMCID: PMC10825286 DOI: 10.1016/j.gendis.2023.06.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/20/2023] [Accepted: 06/27/2023] [Indexed: 02/01/2024] Open
Abstract
Fibroblast growth factor 21 (FGF21) was originally identified as an important metabolic regulator which plays a crucial physiological role in regulating a variety of metabolic parameters through the metabolic network. As a novel multifunctional endocrine growth factor, the role of FGF21 in the metabolic network warrants extensive exploration. This insight was obtained from the observation that the FGF21-dependent mechanism that regulates lipid metabolism, glycogen transformation, and biological effectiveness occurs through the coordinated participation of the liver, adipose tissue, central nervous system, and sympathetic nerves. This review focuses on the role of FGF21-uncoupling protein 1 (UCP1) signaling in lipid metabolism and how FGF21 alleviates non-alcoholic fatty liver disease (NAFLD). Additionally, this review reveals the mechanism by which FGF21 governs glucolipid metabolism. Recent research on the role of FGF21 in the metabolic network has mostly focused on the crucial pathway of glucolipid metabolism. FGF21 has been shown to have multiple regulatory roles in the metabolic network. Since an adequate understanding of the concrete regulatory pathways of FGF21 in the metabolic network has not been attained, this review sheds new light on the metabolic mechanisms of FGF21, explores how FGF21 engages different tissues and organs, and lays a theoretical foundation for future in-depth research on FGF21-targeted treatment of metabolic diseases.
Collapse
Affiliation(s)
| | | | - Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jiaming Li
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| |
Collapse
|
26
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
27
|
Fine KS, Wilkins JT, Sawicki KT. Circulating Branched Chain Amino Acids and Cardiometabolic Disease. J Am Heart Assoc 2024; 13:e031617. [PMID: 38497460 PMCID: PMC11179788 DOI: 10.1161/jaha.123.031617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Branched chain amino acids (BCAAs) are essential for protein homeostasis, energy balance, and signaling pathways. Changes in BCAA homeostasis have emerged as pivotal contributors in the pathophysiology of several cardiometabolic diseases, including type 2 diabetes, obesity, hypertension, atherosclerotic cardiovascular disease, and heart failure. In this review, we provide a detailed overview of BCAA metabolism, focus on molecular mechanisms linking disrupted BCAA homeostasis with cardiometabolic disease, summarize the evidence from observational and interventional studies investigating associations between circulating BCAAs and cardiometabolic disease, and offer valuable insights into the potential for BCAA manipulation as a novel therapeutic strategy for cardiometabolic disease.
Collapse
Affiliation(s)
- Keenan S. Fine
- Northwestern University Feinberg School of MedicineChicagoILUSA
| | - John T. Wilkins
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Division of Cardiology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Konrad T. Sawicki
- Northwestern University Feinberg School of MedicineChicagoILUSA
- Division of Cardiology, Department of MedicineNorthwestern University Feinberg School of MedicineChicagoILUSA
| |
Collapse
|
28
|
Lamming DW. Quantification of healthspan in aging mice: Introducing FAMY and GRAIL. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.07.566044. [PMID: 37986745 PMCID: PMC10659332 DOI: 10.1101/2023.11.07.566044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The population around the world is graying, and as many of these individuals will spend years suffering from the burdens of age associated diseases, understanding how to increase healthspan, defined as the period of life free from disease and disability, is an urgent priority of geroscience research. The lack of agreed-upon quantitative metrics for measuring healthspan in aging mice has slowed progress in identifying interventions that do not simply increase lifespan, but also healthspan. Here, we define FAMY (Frailty-Adjusted Mouse Years) and GRAIL (Gauging Robust Aging when Increasing Lifespan) as new summary statistics for quantifying healthspan in mice. FAMY integrates lifespan data with longitudinal measurements of a widely utilized clinical frailty index, while GRAIL incorporates these measures and also adds information from widely utilized healthspan assays and the hallmarks of aging. Both metrics are conceptually similar to quality-adjusted life years (QALY), a widely-utilized measure of disease burden in humans, and can be readily calculated from data acquired during longitudinal and cross-sectional studies of mouse aging. We find that interventions generally thought to promote health, including calorie restriction, robustly improve healthspan as measured by FAMY and GRAIL. Finally, we show that the use of GRAIL provides new insights, and identify dietary restriction of protein or isoleucine as interventions that robustly promote healthspan but not longevity in female HET3 mice. We suggest that the routine integration of these measures into studies of aging in mice will allow the identification and development of interventions that promote healthy aging even in the absence of increased lifespan.
Collapse
Affiliation(s)
- Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
29
|
Tokizane K, Brace CS, Imai SI. DMH Ppp1r17 neurons regulate aging and lifespan in mice through hypothalamic-adipose inter-tissue communication. Cell Metab 2024; 36:377-392.e11. [PMID: 38194970 PMCID: PMC10922643 DOI: 10.1016/j.cmet.2023.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/11/2024]
Abstract
Recent studies have shown that the hypothalamus functions as a control center of aging in mammals that counteracts age-associated physiological decline through inter-tissue communications. We have identified a key neuronal subpopulation in the dorsomedial hypothalamus (DMH), marked by Ppp1r17 expression (DMHPpp1r17 neurons), that regulates aging and longevity in mice. DMHPpp1r17 neurons regulate physical activity and WAT function, including the secretion of extracellular nicotinamide phosphoribosyltransferase (eNAMPT), through sympathetic nervous stimulation. Within DMHPpp1r17 neurons, the phosphorylation and subsequent nuclear-cytoplasmic translocation of Ppp1r17, regulated by cGMP-dependent protein kinase G (PKG; Prkg1), affect gene expression regulating synaptic function, causing synaptic transmission dysfunction and impaired WAT function. Both DMH-specific Prkg1 knockdown, which suppresses age-associated Ppp1r17 translocation, and the chemogenetic activation of DMHPpp1r17 neurons significantly ameliorate age-associated dysfunction in WAT, increase physical activity, and extend lifespan. Thus, these findings clearly demonstrate the importance of the inter-tissue communication between the hypothalamus and WAT in mammalian aging and longevity control.
Collapse
Affiliation(s)
- Kyohei Tokizane
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cynthia S Brace
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shin-Ichiro Imai
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
30
|
Agius T, Emsley R, Lyon A, MacArthur MR, Kiesworo K, Faivre A, Stavart L, Lambelet M, Legouis D, de Seigneux S, Golshayan D, Lazeyras F, Yeh H, Markmann JF, Uygun K, Ocampo A, Mitchell SJ, Allagnat F, Déglise S, Longchamp A. Short-term hypercaloric carbohydrate loading increases surgical stress resilience by inducing FGF21. Nat Commun 2024; 15:1073. [PMID: 38316771 PMCID: PMC10844297 DOI: 10.1038/s41467-024-44866-3] [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: 08/18/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Dietary restriction promotes resistance to surgical stress in multiple organisms. Counterintuitively, current medical protocols recommend short-term carbohydrate-rich drinks (carbohydrate loading) prior to surgery, part of a multimodal perioperative care pathway designed to enhance surgical recovery. Despite widespread clinical use, preclinical and mechanistic studies on carbohydrate loading in surgical contexts are lacking. Here we demonstrate in ad libitum-fed mice that liquid carbohydrate loading for one week drives reductions in solid food intake, while nearly doubling total caloric intake. Similarly, in humans, simple carbohydrate intake is inversely correlated with dietary protein intake. Carbohydrate loading-induced protein dilution increases expression of hepatic fibroblast growth factor 21 (FGF21) independent of caloric intake, resulting in protection in two models of surgical stress: renal and hepatic ischemia-reperfusion injury. The protection is consistent across male, female, and aged mice. In vivo, amino acid add-back or genetic FGF21 deletion blocks carbohydrate loading-mediated protection from ischemia-reperfusion injury. Finally, carbohydrate loading induction of FGF21 is associated with the induction of the canonical integrated stress response (ATF3/4, NF-kB), and oxidative metabolism (PPARγ). Together, these data support carbohydrate loading drinks prior to surgery and reveal an essential role of protein dilution via FGF21.
Collapse
Affiliation(s)
- Thomas Agius
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raffaella Emsley
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Arnaud Lyon
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Michael R MacArthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kevin Kiesworo
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Anna Faivre
- Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Louis Stavart
- Transplantation Center, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Martine Lambelet
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - David Legouis
- Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Déla Golshayan
- Transplantation Center, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Francois Lazeyras
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
- Center for Biomedical Imaging (CIBM), Geneva, Switzerland
| | - Heidi Yeh
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James F Markmann
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Korkut Uygun
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sarah J Mitchell
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Florent Allagnat
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Pfefferkorn RM, Mortzfeld BM, Fink C, von Frieling J, Bossen J, Esser D, Kaleta C, Rosenstiel P, Heine H, Roeder T. Recurrent Phases of Strict Protein Limitation Inhibit Tumor Growth and Restore Lifespan in A Drosophila Intestinal Cancer Model. Aging Dis 2024; 15:226-244. [PMID: 37962464 PMCID: PMC10796089 DOI: 10.14336/ad.2023.0517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 11/15/2023] Open
Abstract
Diets that restrict caloric or protein intake offer a variety of benefits, including decreasing the incidence of cancer. However, whether such diets pose a substantial therapeutic benefit as auxiliary cancer treatments remains unclear. We determined the effects of severe protein depletion on tumorigenesis in a Drosophila melanogaster intestinal tumor model, using a human RAF gain-of-function allele. Severe and continuous protein restriction significantly reduced tumor growth but resulted in premature death. Therefore, we developed a diet in which short periods of severe protein restriction alternated cyclically with periods of complete feeding. This nutritional regime reduced tumor mass, restored gut functionality, and rescued the lifespan of oncogene-expressing flies to the levels observed in healthy flies on a continuous, fully nutritious diet. Furthermore, this diet reduced the chemotherapy-induced stem cell activity associated with tumor recurrence. Transcriptome analysis revealed long-lasting changes in the expression of key genes involved in multiple major developmental signaling pathways. Overall, the data suggest that recurrent severe protein depletion effectively mimics the health benefits of continuous protein restriction, without undesired nutritional shortcomings. This provides seminal insights into the mechanisms of the memory effect required to maintain the positive effects of protein restriction throughout the phases of a full diet. Finally, the repetitive form of strict protein restriction is an ideal strategy for adjuvant cancer therapy that is useful in many tumor contexts.
Collapse
Affiliation(s)
- Roxana M. Pfefferkorn
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Benedikt M. Mortzfeld
- Department of Cell and Developmental Biology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Christine Fink
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Jakob von Frieling
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Judith Bossen
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Daniela Esser
- Department of Neuroimmunology, Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel, Germany.
| | - Christoph Kaleta
- Department Medical Systems Biology, Institute for Experimental Medicine, Kiel University, Germany.
| | - Philip Rosenstiel
- Department Molecular Cell Biology, Institute for Clinical Molecular Biology, Kiel University, Germany.
| | - Holger Heine
- Division of Innate Immunity, Research Center Borstel - Leibniz Lung Center, Borstel, Germany.
| | - Thomas Roeder
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| |
Collapse
|
32
|
Delgado-Anglés A, Blasco-Roset A, Godoy-Nieto FJ, Cairó M, Villarroya F, Giralt M, Villarroya J. Parkin depletion prevents the age-related alterations in the FGF21 system and the decline in white adipose tissue thermogenic function in mice. J Physiol Biochem 2024; 80:41-51. [PMID: 37914970 PMCID: PMC10808413 DOI: 10.1007/s13105-023-00977-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/20/2023] [Indexed: 11/03/2023]
Abstract
Parkin is an ubiquitin-E3 ligase that is involved in cellular mitophagy and was recently shown to contribute to controlling adipose tissue thermogenic plasticity. We found that Parkin expression is induced in brown (BAT) and white (WAT) adipose tissues of aged mice. We determined the potential role of Parkin in the aging-associated decline in the thermogenic capacity of adipose tissues by analyzing subcutaneous WAT, interscapular BAT, and systemic metabolic and physiological parameters in young (5 month-old) and aged (16 month-old) mice with targeted invalidation of the Parkin (Park2) gene, and their wild-type littermates. Our data indicate that suppression of Parkin prevented adipose accretion, increased energy expenditure and improved the systemic metabolic derangements, such as insulin resistance, seen in aged mice. This was associated with maintenance of browning and reduction of the age-associated induction of inflammation in subcutaneous WAT. BAT in aged mice was much less affected by Parkin gene invalidation. Such protection was associated with a dramatic prevention of the age-associated induction of fibroblast growth factor-21 (FGF21) levels in aged Parkin-invalidated mice. This was associated with a parallel reduction in FGF21 gene expression in adipose tissues and liver in aged Parkin-invalidated mice. Additionally, Parkin invalidation prevented the protein down-regulation of β-Klotho (a key co-receptor mediating FGF21 responsiveness in tissues) in aged adipose tissues. We conclude that Parkin down-regulation leads to improved systemic metabolism in aged mice, in association with maintenance of adipose tissue browning and FGF21 system functionality.
Collapse
Affiliation(s)
- Alejandro Delgado-Anglés
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Albert Blasco-Roset
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Francisco J Godoy-Nieto
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Montserrat Cairó
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red "Fisiopatología de la Obesidad y Nutrición", Madrid, Spain
| | - Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red "Fisiopatología de la Obesidad y Nutrición", Madrid, Spain
- Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red "Fisiopatología de la Obesidad y Nutrición", Madrid, Spain
- Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Joan Villarroya
- Departament de Bioquímica i Biomedicina Molecular and Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red "Fisiopatología de la Obesidad y Nutrición", Madrid, Spain.
- Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
33
|
Garg S, Morehead LC, Bird JT, Graw S, Gies A, Storey AJ, Tackett AJ, Edmondson RD, Mackintosh SG, Byrum SD, Miousse IR. Characterization of methionine dependence in melanoma cells. Mol Omics 2024; 20:37-47. [PMID: 37782107 PMCID: PMC10903584 DOI: 10.1039/d3mo00087g] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Dietary methionine restriction is associated with a reduction in tumor growth in preclinical studies and an increase in lifespan in animal models. The mechanism by which methionine restriction inhibits tumor growth while sparing normal cells is incompletely understood. We do know that normal cells can utilize methionine or homocysteine interchangeably (methionine independence) while most cancer cells are strictly dependent on methionine availability. Here, we compared a typical methionine dependent and a rare methionine independent melanoma cell line. We show that replacing methionine, a methyl donor, with its precursor homocysteine generally induced hypomethylation in gene promoters. This decrease was similar in methionine dependent and methionine independent cells. There was only a low level of pathway enrichment, suggesting that the hypomethylation is generalized rather than gene specific. Whole proteome and transcriptome were also analyzed. This analysis revealed that contrarily to the effect on methylation, the replacement of methionine with homocysteine had a much greater effect on the transcriptome and proteome of methionine dependent cells than methionine independent cells. Interestingly, methionine adenosyltransferase 2A (MAT2A), responsible for the synthesis of S-adenosylmethionine from methionine, was equally strongly upregulated in both cell lines. This suggests that the absence of methionine is equally detected but triggers different outcomes in methionine dependent versus independent cells. Our analysis reveals the importance of cell cycle control, DNA damage repair, translation, nutrient sensing, oxidative stress and immune functions in the cellular response to methionine stress in melanoma.
Collapse
Affiliation(s)
- Sarita Garg
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Lauren C Morehead
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Jordan T Bird
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Stefan Graw
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Allen Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Aaron J Storey
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Rick D Edmondson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Isabelle R Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| |
Collapse
|
34
|
Na K, Park YJ. Protein Restriction in Metabolic Health: Lessons from Rodent Models. Nutrients 2024; 16:229. [PMID: 38257122 PMCID: PMC10819042 DOI: 10.3390/nu16020229] [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: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Consumption of protein-rich diets and supplements has been increasingly advocated by individuals seeking to optimize metabolic health and mitigate the effects of aging. Protein intake is postulated to support muscle mass retention and enhance longevity, underscoring its perceived benefits in age-related metabolic regulation. However, emerging evidence presents a paradox; while moderate protein consumption contributes to health maintenance, an excessive intake is associated with an elevated risk of chronic diseases, notably obesity and diabetes. Furthermore, recent studies suggest that reducing the ratio of protein intake to macronutrients improves metabolic parameters and extends lifespan. The aim of this study is to review the current evidence concerning the metabolic effects of protein-restricted diets and their potential mechanisms. Utilizing rodent models, investigations have revealed that protein-restricted diets exert a notable influence over food intake and energy consumption, ultimately leading to body weight loss, depending on the degree of dietary protein restriction. These phenotypic alterations are primarily mediated by the FGF21 signaling pathway, whose activation is likely regulated by ATF4 and the circadian clock. The evidence suggests that protein-restricted diets as an alternative approach to calorie-restricted regimes, particularly in overweight or obese adults. However, more research is needed to determine the optimal level of restriction, duration, and long-term effects of such interventions.
Collapse
Affiliation(s)
- Khuhee Na
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea;
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yoon Jung Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea;
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
35
|
Yeh CY, Chini LC, Davidson JW, Garcia GG, Gallagher MS, Freichels IT, Calubag MF, Rodgers AC, Green CL, Babygirija R, Sonsalla MM, Pak HH, Trautman M, Hacker TA, Miller RA, Simcox J, Lamming DW. Late-life isoleucine restriction promotes physiological and molecular signatures of healthy aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.06.527311. [PMID: 36798157 PMCID: PMC9934591 DOI: 10.1101/2023.02.06.527311] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
In defiance of the paradigm that calories from all sources are equivalent, we and others have shown that dietary protein is a dominant regulator of healthy aging. The restriction of protein or the branched-chain amino acid isoleucine promotes healthspan and extends lifespan when initiated in young or adult mice. However, many interventions are less efficacious or even deleterious when initiated in aged animals. Here, we investigate the physiological, metabolic, and molecular consequences of consuming a diet with a 67% reduction of all amino acids (Low AA), or of isoleucine alone (Low Ile), in male and female C57BL/6J.Nia mice starting at 20 months of age. We find that both diet regimens effectively reduce adiposity and improve glucose tolerance, which were benefits that were not mediated by reduced calorie intake. Both diets improve specific aspects of frailty, slow multiple molecular indicators of aging rate, and rejuvenate the aging heart and liver at the molecular level. These results demonstrate that Low AA and Low Ile diets can drive youthful physiological and molecular signatures, and support the possibility that these dietary interventions could help to promote healthy aging in older adults.
Collapse
Affiliation(s)
- Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Lucas C.S. Chini
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Jessica W. Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Gonzalo G. Garcia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Meredith S. Gallagher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Isaac T. Freichels
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Allison C. Rodgers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI 53706
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Michaela Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Timothy A. Hacker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI 53706
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
36
|
Hine C, Patel AK, Ponti AK. Diet-Modifiable Redox Alterations in Ageing and Cancer. Subcell Biochem 2024; 107:129-172. [PMID: 39693023 PMCID: PMC11753504 DOI: 10.1007/978-3-031-66768-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
With ageing comes some of life's best and worst moments. Those lucky enough to live out into the seventh, eighth, and nineth decades and perhaps beyond have more opportunities to experience the wonders and joys of the world. As the world's population shifts towards more and more of these individuals, this is something to be celebrated. However, it is not without negative consequences. Advanced age also ushers in health decline and the burden of non-communicable diseases such as cancer, heart disease, stroke, and organ function decay. Thus, alleviating or at least dampening the severity of ageing as a whole, as well as these individual age-related disorders will enable the improvement in lifespan and healthspan. In the following chapter, we delve into hypothesised causes of ageing and experimental interventions that can be taken to slow their progression. We also highlight cellular and subcellular mechanisms of ageing with a focus on protein thiol oxidation and posttranslational modifications that impact cellular homeostasis and the advent and progression of ageing-related cancers. By having a better understanding of the mechanisms of ageing, we can hopefully develop effective, safe, and efficient therapeutic modalities that can be used prophylactically and/or concurrent to the onset of ageing.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.
| | - Anand Kumar Patel
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Cardiovascular Genetics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - András K Ponti
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| |
Collapse
|
37
|
Trautman ME, Braucher LN, Elliehausen C, Zhu WG, Zelenovskiy E, Green M, Sonsalla MM, Yeh CY, Hornberger TA, Konopka AR, Lamming DW. Resistance exercise protects mice from protein-induced fat accretion. eLife 2023; 12:RP91007. [PMID: 38019262 PMCID: PMC10686620 DOI: 10.7554/elife.91007] [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] [Indexed: 11/30/2023] Open
Abstract
Low-protein (LP) diets extend the lifespan of diverse species and are associated with improved metabolic health in both rodents and humans. Paradoxically, many athletes and bodybuilders consume high-protein (HP) diets and protein supplements, yet are both fit and metabolically healthy. Here, we examine this paradox using weight pulling, a validated progressive resistance exercise training regimen, in mice fed either an LP diet or an isocaloric HP diet. We find that despite having lower food consumption than the LP group, HP-fed mice gain significantly more fat mass than LP-fed mice when not exercising, while weight pulling protected HP-fed mice from this excess fat accretion. The HP diet augmented exercise-induced hypertrophy of the forearm flexor complex, and weight pulling ability increased more rapidly in the exercised HP-fed mice. Surprisingly, exercise did not protect from HP-induced changes in glycemic control. Our results confirm that HP diets can augment muscle hypertrophy and accelerate strength gain induced by resistance exercise without negative effects on fat mass, and also demonstrate that LP diets may be advantageous in the sedentary. Our results highlight the need to consider both dietary composition and activity, not simply calories, when taking a precision nutrition approach to health.
Collapse
Affiliation(s)
- Michaela E Trautman
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Nutrition and Metabolism Graduate Program, University of Wisconsin- MadisonMadisonUnited States
| | - Leah N Braucher
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Christian Elliehausen
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Wenyuan G Zhu
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Esther Zelenovskiy
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Madelyn Green
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Troy A Hornberger
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- School of Veterinary Medicine, University of Wisconsin-MadisonMadisonUnited States
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Nutrition and Metabolism Graduate Program, University of Wisconsin- MadisonMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- University of Wisconsin Carbone Cancer CenterMadisonUnited States
| |
Collapse
|
38
|
Green CL, Trautman ME, Chaiyakul K, Jain R, Alam YH, Babygirija R, Pak HH, Sonsalla MM, Calubag MF, Yeh CY, Bleicher A, Novak G, Liu TT, Newman S, Ricke WA, Matkowskyj KA, Ong IM, Jang C, Simcox J, Lamming DW. Dietary restriction of isoleucine increases healthspan and lifespan of genetically heterogeneous mice. Cell Metab 2023; 35:1976-1995.e6. [PMID: 37939658 PMCID: PMC10655617 DOI: 10.1016/j.cmet.2023.10.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Low-protein diets promote health and longevity in diverse species. Restriction of the branched-chain amino acids (BCAAs) leucine, isoleucine, and valine recapitulates many of these benefits in young C57BL/6J mice. Restriction of dietary isoleucine (IleR) is sufficient to promote metabolic health and is required for many benefits of a low-protein diet in C57BL/6J males. Here, we test the hypothesis that IleR will promote healthy aging in genetically heterogeneous adult UM-HET3 mice. We find that IleR improves metabolic health in young and old HET3 mice, promoting leanness and glycemic control in both sexes, and reprograms hepatic metabolism in a sex-specific manner. IleR reduces frailty and extends the lifespan of male and female mice, but to a greater degree in males. Our results demonstrate that IleR increases healthspan and longevity in genetically diverse mice and suggests that IleR, or pharmaceuticals that mimic this effect, may have potential as a geroprotective intervention.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Krittisak Chaiyakul
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yasmine H Alam
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Anneliese Bleicher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Grace Novak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Teresa T Liu
- George M. O'Brien Center of Research Excellence, Department of Urology, University of Wisconsin, Madison, WI 93705, USA
| | - Sarah Newman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Will A Ricke
- George M. O'Brien Center of Research Excellence, Department of Urology, University of Wisconsin, Madison, WI 93705, USA
| | - Kristina A Matkowskyj
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Irene M Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA; Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
39
|
Prezotto LD, Keane JA, Cupp AS, Thorson JF. Fibroblast Growth Factor 21 Has a Diverse Role in Energetic and Reproductive Physiological Functions of Female Beef Cattle. Animals (Basel) 2023; 13:3185. [PMID: 37893910 PMCID: PMC10603626 DOI: 10.3390/ani13203185] [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: 08/16/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) has been identified in multiple mammalian species as a molecular marker of energy metabolism while also providing negative feedback to the gonads. However, the role of FGF21 in regulating the energetic and reproductive physiology of beef heifers and cows has yet to be characterized. Herein, we investigated the temporal concentrations of FGF21 in female beef cattle from the prepubertal period to early lactation. Circulating concentrations of FGF21, non-esterified fatty acids, plasma urea nitrogen, glucose, and progesterone were assessed. Ultrasonography was employed to determine the onset of puberty and resumption of postpartum ovarian cyclicity as well as to measure backfat thickness. Finally, cows and calves underwent the weigh-suckle-weigh technique to estimate rate of milk production. We have revealed that FGF21 has an expansive role in the physiology of female beef cattle, including pubertal onset, adaptation to nutritional transition, rate of body weight gain, circulating markers of metabolism, and rate of milk production. In conclusion, FGF21 plays a role in physiological functions in beef cattle that can be applied to advance the understanding of basic scientific processes governing the nutritional regulation of reproductive function but also provides a novel means for beef cattle producers to select parameters of financial interest.
Collapse
Affiliation(s)
- Ligia D. Prezotto
- Department of Animal Science, University of Nebraska-Lincoln, 3940 Fair Street, Lincoln, NE 68583-0908, USA; (L.D.P.); (J.A.K.); (A.S.C.)
| | - Jessica A. Keane
- Department of Animal Science, University of Nebraska-Lincoln, 3940 Fair Street, Lincoln, NE 68583-0908, USA; (L.D.P.); (J.A.K.); (A.S.C.)
| | - Andrea S. Cupp
- Department of Animal Science, University of Nebraska-Lincoln, 3940 Fair Street, Lincoln, NE 68583-0908, USA; (L.D.P.); (J.A.K.); (A.S.C.)
| | - Jennifer F. Thorson
- U.S. Meat Animal Research Center, Agricultural Research Service, United States Department of Agriculture, Clay Center, NE 68933-0166, USA
| |
Collapse
|
40
|
Munoz MD, Zamudio A, McCann M, Gil V, Xu P, Liew CW. Activation of brown adipose tissue by a low-protein diet ameliorates hyperglycemia in a diabetic lipodystrophy mouse model. Sci Rep 2023; 13:11808. [PMID: 37479751 PMCID: PMC10362023 DOI: 10.1038/s41598-023-37482-6] [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: 03/16/2023] [Accepted: 06/22/2023] [Indexed: 07/23/2023] Open
Abstract
Long-term ad libitum dietary restrictions, such as low-protein diets (LPDs), improve metabolic health and extend the life span of mice and humans. However, most studies conducted thus far have focused on the preventive effects of LPDs on metabolic syndromes. To test the therapeutic potential of LPD, we treated a lipodystrophy mouse model IRFKO (adipose-specific insulin receptor knockout) in this study. We have previously shown that IRFKO mice have profound insulin resistance, hyperglycemia, and whitening of interscapular brown adipose tissue (BAT), closely mimicking the phenotypes in lipoatrophic diabetic patients. Here, we demonstrate that 14-day of LPD (5.1% kcal from protein) feeding is sufficient to reduce postprandial blood glucose, improve insulin resistance, and normalize glucose tolerance in the IRFKO mice. This profound metabolic improvement is associated with BAT activation and increase in whole body energy expenditure. To confirm, we showed that surgical denervation of BAT attenuated the beneficial metabolic effects of LPD feeding in IRFKO mice, including the 'browning' effects on BAT and the glucose-ameliorating results. However, BAT denervation failed to affect the body weight-lowering effects of LPD. Together, our results imply a therapeutic potential to use LPD for the treatment of lipoatrophic diabetes.
Collapse
Affiliation(s)
- Marcos David Munoz
- Department of Physiology and Biophysics, The University of Illinois at Chicago, 909 S Wolcott Ave, RM 2099, Chicago, IL, 60612, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Alexa Zamudio
- Department of Physiology and Biophysics, The University of Illinois at Chicago, 909 S Wolcott Ave, RM 2099, Chicago, IL, 60612, USA
| | - Maximilian McCann
- Department of Physiology and Biophysics, The University of Illinois at Chicago, 909 S Wolcott Ave, RM 2099, Chicago, IL, 60612, USA
| | - Victoria Gil
- Department of Physiology and Biophysics, The University of Illinois at Chicago, 909 S Wolcott Ave, RM 2099, Chicago, IL, 60612, USA
| | - Pingwen Xu
- Department of Physiology and Biophysics, The University of Illinois at Chicago, 909 S Wolcott Ave, RM 2099, Chicago, IL, 60612, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Chong Wee Liew
- Department of Physiology and Biophysics, The University of Illinois at Chicago, 909 S Wolcott Ave, RM 2099, Chicago, IL, 60612, USA.
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
41
|
Cao Y, Yang M, Song J, Jiang X, Xu S, Che L, Fang Z, Lin Y, Jin C, Feng B, Wu D, Hua L, Zhuo Y. Dietary Protein Regulates Female Estrous Cyclicity Partially via Fibroblast Growth Factor 21. Nutrients 2023; 15:3049. [PMID: 37447375 DOI: 10.3390/nu15133049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21), a hormone predominantly released in the liver, has emerged as a critical endocrine signal of dietary protein intake, but its role in the control of estrous cyclicity by dietary protein remains uncertain. To investigated the role of FGF21 and hypothalamic changes in the regulation of estrous cyclicity by dietary protein intake, female adult Sprague-Dawley rats with normal estrous cycles were fed diets with protein contents of 4% (P4), 8% (P8), 13% (P13), 18% (P18), and 23% (P23). FGF21 liver-specific knockout or wild-type mice were fed P18 or P4 diets to examine the role of liver FGF21 in the control of estrous cyclicity. Dietary protein restriction resulted in no negative effects on estrous cyclicity or ovarian follicular development when the protein content was greater than 8%. Protein restriction at 4% resulted in decreased bodyweight, compromised Kiss-1 expression in the hypothalamus, disturbed estrous cyclicity, and inhibited uterine and ovarian follicular development. The disturbed estrous cyclicity in rats that received the P4 diet was reversed after feeding with the P18 diet. Liver Fgf21 mRNA expressions and serum FGF21 levels were significantly increased as dietary protein content decreased, and loss of hepatic FGF21 delayed the onset of cyclicity disruption in rats fed with the P4 diet, possibly due to the regulation of insulin-like growth factor-1. Collectively, severe dietary protein restriction results in the cessation of estrous cyclicity and ovarian follicle development, and hepatic FGF21 and hypothalamic Kiss-1 were partially required for this process.
Collapse
Affiliation(s)
- Yaxue Cao
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Min Yang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Pet Nutrition and Health Research Center, Chengdu Agricultural College, Chengdu 611130, China
| | - Jie Song
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuemei Jiang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Shengyu Xu
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Jin
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - De Wu
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lun Hua
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
42
|
Lee AH, Sugiura Y, Youm YH, Dlugos T, Maeda R, Coman D, Spadaro O, Sidorov S, Shchukina I, Andhey S, Smith SR, Ravussin E, Hyder F, Artyomov MN, Dixit VD. [WITHDRAWN] Activation of transsulfuration pathway to maintain cysteine is a thermogenic checkpoint for the conservation of energy. RESEARCH SQUARE 2023:rs.3.rs-3069713. [PMID: 37461682 PMCID: PMC10350108 DOI: 10.21203/rs.3.rs-3069713/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
The authors have requested that this preprint be removed from Research Square.
Collapse
Affiliation(s)
- Aileen H. Lee
- Department of Pathology
- Department of Comparative Medicine
- Department of Immunobiology, Yale School of Medicine, New
Haven, CT 06520, USA
| | | | - Yun-Hee Youm
- Department of Pathology
- Department of Comparative Medicine
- Department of Immunobiology, Yale School of Medicine, New
Haven, CT 06520, USA
| | - Tamara Dlugos
- Department of Pathology
- Department of Comparative Medicine
- Department of Immunobiology, Yale School of Medicine, New
Haven, CT 06520, USA
| | | | - Daniel Coman
- Department of Radiology and Biomedical Imaging
- Department of Biomedical Engineering, School of Engineering
and Applied Science, Yale University
| | - Olga Spadaro
- Department of Pathology
- Department of Comparative Medicine
- Department of Immunobiology, Yale School of Medicine, New
Haven, CT 06520, USA
| | - Sviatoslav Sidorov
- Department of Pathology
- Department of Comparative Medicine
- Department of Immunobiology, Yale School of Medicine, New
Haven, CT 06520, USA
| | - Irina Shchukina
- Department of Pathology and Immunology Washington
University School of Medicine, St. Louis, MO 63110, USA
| | - Sairam Andhey
- Department of Pathology and Immunology Washington
University School of Medicine, St. Louis, MO 63110, USA
| | - Steven R. Smith
- Translational Research Institute for Metabolism and
Diabetes, AdventHealth, Orlando, FL, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA,
USA
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging
- Department of Biomedical Engineering, School of Engineering
and Applied Science, Yale University
| | - Maxim N. Artyomov
- Department of Pathology and Immunology Washington
University School of Medicine, St. Louis, MO 63110, USA
| | - Vishwa Deep Dixit
- Department of Pathology
- Department of Comparative Medicine
- Department of Immunobiology, Yale School of Medicine, New
Haven, CT 06520, USA
- Yale Center for Research on Aging, Yale School of
Medicine, New Haven, CT 06520, USA
| |
Collapse
|
43
|
Moraitis I, Guiu J, Rubert J. Gut microbiota controlling radiation-induced enteritis and intestinal regeneration. Trends Endocrinol Metab 2023:S1043-2760(23)00108-X. [PMID: 37336645 DOI: 10.1016/j.tem.2023.05.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/21/2023]
Abstract
Cancer remains the second leading cause of mortality, with nearly 10 million deaths worldwide in 2020. In many cases, radiotherapy is used for its anticancer effects. However, radiation causes healthy tissue toxicity as a side effect. In intra-abdominal and pelvic malignancies, the healthy bowel is inevitably included in the radiation field, causing radiation-induced enteritis and dramatically affecting the gut microbiome. This condition is associated with significant morbidity and mortality that impairs cancer patients' and survivors' quality of life. This Review provides a critical overview of the main drivers in modulating the gut microenvironment in homeostasis, disease, and injury, focusing on gut microbial metabolites and microorganisms that influence epithelial regeneration upon radiation injury.
Collapse
Affiliation(s)
- Ilias Moraitis
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain; Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet de Llobregat, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain; Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet de Llobregat, Spain.
| | - Josep Rubert
- Division of Human Nutrition and Health, Wageningen University & Research, Stippeneng 4, Wageningen, 6708, WE, Netherlands; Food Quality and Design, Wageningen University & Research, Bornse Weilanden 9, Wageningen, 6708, WG, Netherlands.
| |
Collapse
|
44
|
Rafiq T, Stearns JC, Shanmuganathan M, Azab SM, Anand SS, Thabane L, Beyene J, Williams NC, Morrison KM, Teo KK, Britz-McKibbin P, de Souza RJ. Integrative multiomics analysis of infant gut microbiome and serum metabolome reveals key molecular biomarkers of early onset childhood obesity. Heliyon 2023; 9:e16651. [PMID: 37332914 PMCID: PMC10272340 DOI: 10.1016/j.heliyon.2023.e16651] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Evidence supports a complex interplay of gut microbiome and host metabolism as regulators of obesity. The metabolic phenotype and microbial metabolism of host diet may also contribute to greater obesity risk in children early in life. This study aimed to identify features that discriminated overweight/obese from normal weight infants by integrating gut microbiome and serum metabolome profiles. This prospective analysis included 50 South Asian children living in Canada, selected from the SouTh Asian biRth cohorT (START). Serum metabolites were measured by multisegment injection-capillary electrophoresis-mass spectrometry and the relative abundance of bacterial 16S rRNA gene amplicon sequence variant was evaluated at 1 year. Cumulative body mass index (BMIAUC) and skinfold thickness (SSFAUC) scores were calculated from birth to 3 years as the total area under the growth curve (AUC). BMIAUC and/or SSFAUC >85th percentile was used to define overweight/obesity. Data Integration Analysis for Biomarker discovery using Latent cOmponent (DIABLO) was used to identify discriminant features associated with childhood overweight/obesity. The associations between identified features and anthropometric measures were examined using logistic regression. Circulating metabolites including glutamic acid, acetylcarnitine, carnitine, and threonine were positively, whereas γ-aminobutyric acid (GABA), symmetric dimethylarginine (SDMA), and asymmetric dimethylarginine (ADMA) were negatively associated with childhood overweight/obesity. The abundance of the Pseudobutyrivibrio and Lactobacillus genera were positively, and Clostridium sensu stricto 1 and Akkermansia were negatively associated with childhood overweight/obesity. Integrative analysis revealed that Akkermansia was positively whereas Lactobacillus was inversely correlated with GABA and SDMA, and Pseudobutyrivibrio was inversely correlated with GABA. This study provides insights into metabolic and microbial signatures which may regulate satiety, energy metabolism, inflammatory processes, and/or gut barrier function, and therefore, obesity trajectories in childhood. Understanding the functional capacity of these molecular features and potentially modifiable risk factors such as dietary exposures early in life may offer a novel approach for preventing childhood obesity.
Collapse
Affiliation(s)
- Talha Rafiq
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - Jennifer C. Stearns
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Sandi M. Azab
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pharmacognosy, Alexandria University, Alexandria 21521, Egypt
| | - Sonia S. Anand
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Lehana Thabane
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biostatistics Unit, Father Sean O’Sullivan Research Centre, The Research Institute, St Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
- Faculty of Health Sciences, University of Johannesburg, Johannesburg 524, South Africa
| | - Joseph Beyene
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | - Katherine M. Morrison
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Koon K. Teo
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Russell J. de Souza
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
45
|
Flores V, Spicer AB, Sonsalla MM, Richardson NE, Yu D, Sheridan GE, Trautman ME, Babygirija R, Cheng EP, Rojas JM, Yang SE, Wakai MH, Hubbell R, Kasza I, Tomasiewicz JL, Green CL, Dantoin C, Alexander CM, Baur JA, Malecki KC, Lamming DW. Regulation of metabolic health by dietary histidine in mice. J Physiol 2023; 601:2139-2163. [PMID: 36086823 PMCID: PMC9995620 DOI: 10.1113/jp283261] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/01/2022] [Indexed: 11/08/2022] Open
Abstract
Low-protein (LP) diets are associated with a decreased risk of diabetes in humans, and promote leanness and glycaemic control in both rodents and humans. While the effects of an LP diet on glycaemic control are mediated by reduced levels of the branched-chain amino acids, we have observed that reducing dietary levels of the other six essential amino acids leads to changes in body composition. Here, we find that dietary histidine plays a key role in the response to an LP diet in male C57BL/6J mice. Specifically reducing dietary levels of histidine by 67% reduces the weight gain of young, lean male mice, reducing both adipose and lean mass without altering glucose metabolism, and rapidly reverses diet-induced obesity and hepatic steatosis in diet-induced obese male mice, increasing insulin sensitivity. This normalization of metabolic health was associated not with caloric restriction or increased activity, but with increased energy expenditure. Surprisingly, the effects of histidine restriction do not require the energy balance hormone Fgf21. Histidine restriction that was started in midlife promoted leanness and glucose tolerance in aged males but not females, but did not affect frailty or lifespan in either sex. Finally, we demonstrate that variation in dietary histidine levels helps to explain body mass index differences in humans. Overall, our findings demonstrate that dietary histidine is a key regulator of weight and body composition in male mice and in humans, and suggest that reducing dietary histidine may be a translatable option for the treatment of obesity. KEY POINTS: Protein restriction (PR) promotes metabolic health in rodents and humans and extends rodent lifespan. Restriction of specific individual essential amino acids can recapitulate the benefits of PR. Reduced histidine promotes leanness and increased energy expenditure in male mice. Reduced histidine does not extend the lifespan of mice when begun in midlife. Dietary levels of histidine are positively associated with body mass index in humans.
Collapse
Affiliation(s)
- Victoria Flores
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alexandra B. Spicer
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Michelle M. Sonsalla
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nicole E. Richardson
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Deyang Yu
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Grace E. Sheridan
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michaela E. Trautman
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Reji Babygirija
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eunhae P. Cheng
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jennifer M. Rojas
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shany E. Yang
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Matthew H. Wakai
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ryan Hubbell
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Cara L. Green
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Claudia Dantoin
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristen C. Malecki
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Dudley W. Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA
| |
Collapse
|
46
|
Sonsalla MM, Lamming DW. Geroprotective interventions in the 3xTg mouse model of Alzheimer's disease. GeroScience 2023; 45:1343-1381. [PMID: 37022634 PMCID: PMC10400530 DOI: 10.1007/s11357-023-00782-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: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disease. As the population ages, the increasing prevalence of AD threatens massive healthcare costs in the coming decades. Unfortunately, traditional drug development efforts for AD have proven largely unsuccessful. A geroscience approach to AD suggests that since aging is the main driver of AD, targeting aging itself may be an effective way to prevent or treat AD. Here, we discuss the effectiveness of geroprotective interventions on AD pathology and cognition in the widely utilized triple-transgenic mouse model of AD (3xTg-AD) which develops both β-amyloid and tau pathologies characteristic of human AD, as well as cognitive deficits. We discuss the beneficial impacts of calorie restriction (CR), the gold standard for geroprotective interventions, and the effects of other dietary interventions including protein restriction. We also discuss the promising preclinical results of geroprotective pharmaceuticals, including rapamycin and medications for type 2 diabetes. Though these interventions and treatments have beneficial effects in the 3xTg-AD model, there is no guarantee that they will be as effective in humans, and we discuss the need to examine these interventions in additional animal models as well as the urgent need to test if some of these approaches can be translated from the lab to the bedside for the treatment of humans with AD.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, 2500 Overlook Terrace, VAH C3127 Research 151, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 2500 Overlook Terrace, VAH C3127 Research 151, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA.
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
47
|
Wang L, Wang F, Xiong L, Song H, Ren B, Shen X. A nexus of dietary restriction and gut microbiota: Recent insights into metabolic health. Crit Rev Food Sci Nutr 2023; 64:8649-8671. [PMID: 37154021 DOI: 10.1080/10408398.2023.2202750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
In recent times, dietary restriction (DR) has received considerable attention for its promising effects on metabolism and longevity. Previous studies on DR have mainly focused on the health benefits produced by different restriction patterns, whereas comprehensive reviews of the role of gut microbiota during DR are limited. In this review, we discuss the effects of caloric restriction, fasting, protein restriction, and amino acid restriction from a microbiome perspective. Furthermore, the underlying mechanisms by which DR affects metabolic health by regulating intestinal homeostasis are summarized. Specifically, we reviewed the impacts of different DRs on specific gut microbiota. Additionally, we put forward the limitations of the current research and suggest the development of personalized microbes-directed DR for different populations and corresponding next-generation sequencing technologies for accurate microbiological analysis. DR effectively modulates the composition of the gut microbiota and microbial metabolites. In particular, DR markedly affects the rhythmic oscillation of microbes which may be related to the circadian clock system. Moreover, increasing evidence supports that DR profoundly improves metabolic syndrome, inflammatory bowel disease, and cognitive impairment. To summarize, DR may be an effective and executable dietary manipulation strategy for maintaining metabolic health, however, further investigation is needed to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
48
|
Wang Y, Li J, Zhang Z, Wang R, Bo H, Zhang Y. Exercise Improves the Coordination of the Mitochondrial Unfolded Protein Response and Mitophagy in Aging Skeletal Muscle. Life (Basel) 2023; 13:life13041006. [PMID: 37109535 PMCID: PMC10142204 DOI: 10.3390/life13041006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) and mitophagy are two mitochondrial quality control (MQC) systems that work at the molecular and organelle levels, respectively, to maintain mitochondrial homeostasis. Under stress conditions, these two processes are simultaneously activated and compensate for each other when one process is insufficient, indicating mechanistic coordination between the UPRmt and mitophagy that is likely controlled by common upstream signals. This review focuses on the molecular signals regulating this coordination and presents evidence showing that this coordination mechanism is impaired during aging and promoted by exercise. Furthermore, the bidirectional regulation of reactive oxygen species (ROS) and AMPK in modulating this mechanism is discussed. The hierarchical surveillance network of MQC can be targeted by exercise-derived ROS to attenuate aging, which offers a molecular basis for potential therapeutic interventions for sarcopenia.
Collapse
Affiliation(s)
- Yan Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
- School of Physical Education, Guangdong Institute of Petrochemical Technology, Maoming 525000, China
| | - Jialin Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| | - Ziyi Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| | - Runzi Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| | - Hai Bo
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
- Department of Military Training Medicines, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, China
| | - Yong Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| |
Collapse
|
49
|
Garg S, Morehead LC, Bird JT, Graw S, Gies A, Storey AJ, Tackett AJ, Edmondson RD, Mackintosh SG, Byrum SD, Miousse IR. Characterization of methionine dependence in melanoma cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.05.535723. [PMID: 37066392 PMCID: PMC10104025 DOI: 10.1101/2023.04.05.535723] [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] [Indexed: 06/19/2023]
Abstract
Dietary methionine restriction is associated with a reduction in tumor growth in preclinical studies and an increase in lifespan in animal models. The mechanism by which methionine restriction inhibits tumor growth while sparing normal cells is incompletely understood. We do know that normal cells can utilize methionine or homocysteine interchangeably (methionine independence) while most cancer cells are strictly dependent on methionine availability. Here, we compared a typical methionine dependent and a rare methionine independent melanoma cell line. We show that replacing methionine, a methyl donor, with its precursor homocysteine generally induced hypomethylation in gene promoters. This decrease was similar in methionine dependent and methionine independent cells. There was only a low level of pathway enrichment, suggesting that the hypomethylation is generalized rather than gene specific. Whole proteome and transcriptome were also analyzed. This analysis revealed that contrarily to the effect on methylation, the replacement of methionine with homocysteine had a much greater effect on the transcriptome and proteome of methionine dependent cells than methionine independent cells. Interestingly, methionine adenosyltransferase 2A (MAT2A), responsible for the synthesis of s-adenosylmethionine from methionine, was equally strongly upregulated in both cell lines. This suggests that the absence of methionine is equally detected but triggers different outcomes in methionine dependent versus independent cells. Our analysis reveals the importance of cell cycle control, DNA damage repair, translation, nutrient sensing, oxidative stress and immune functions in the cellular response to methionine stress in melanoma.
Collapse
Affiliation(s)
- Sarita Garg
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Lauren C Morehead
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Jordan T Bird
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Stefan Graw
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Allen Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Aaron J Storey
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Rick D Edmondson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| | - Isabelle R Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences
| |
Collapse
|
50
|
Li C, Zhu J, Wei S, Ye X, Yang L, Wang Z, Chen Y. Intermittent protein restriction improves glucose homeostasis in Zucker diabetic fatty rats and single-cell sequencing reveals distinct changes in β cells. J Nutr Biochem 2023; 114:109275. [PMID: 36669706 DOI: 10.1016/j.jnutbio.2023.109275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 12/04/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Diabetes is caused by the interplay between genetic and environmental factors, therefore changes of lifestyle and dietary patterns are the most common practices for diabetes intervention. Protein restriction and caloric restriction have been shown to improve diabetic hyperglycemia in both animal models and humans. We report here the effectiveness of intermittent protein restriction (IPR) for the intervention of diabetes in Zucker diabetic fatty (ZDF) rats. Administration of IPR significantly reduced hyperglycemia and decreased glucose production in the liver. IPR protected pancreatic islets from diabetes-mediated damages as well as elevated the number and the proliferation activity of β cells. Single-cell RNA sequencing performed with isolated islets from the ZDF rats revealed that IPR was able to reverse the diabetes-associated β cell dedifferentiation. In addition, diabetic β cells in ZDF rats were associated with increased expressions of islet amyloid polypeptide, chromogranin and genes involved in endoplasmic reticulum stress. A β cell dedifferentiation marker Cd81 was also increased in the β cells of diabetic rats. In contrast, the expressions of D-box binding PAR bZIP transcription factor Dbp and immediate-early response genes were reduced in the diabetic β cells. In conclusion, these results indicated that IPR is effective in glycemic control and β cell protection in a diabetic rat model. In addition, diabetes in ZDF rats is associated with changes in the expression of genes involved in many facets of β cell functions.
Collapse
Affiliation(s)
- Chenchen Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jing Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Siying Wei
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyi Ye
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lanzexin Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zinan Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|