151
|
Aon MA, Bernier M, Mitchell SJ, Di Germanio C, Mattison JA, Ehrlich MR, Colman RJ, Anderson RM, de Cabo R. Untangling Determinants of Enhanced Health and Lifespan through a Multi-omics Approach in Mice. Cell Metab 2020; 32:100-116.e4. [PMID: 32413334 PMCID: PMC8214079 DOI: 10.1016/j.cmet.2020.04.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/20/2019] [Accepted: 04/24/2020] [Indexed: 12/17/2022]
Abstract
The impact of chronic caloric restriction (CR) on health and survival is complex with poorly understood underlying molecular mechanisms. A recent study in mice addressing the diets used in nonhuman primate CR studies found that while diet composition did not impact longevity, fasting time and total calorie intake were determinant for increased survival. Here, integrated analysis of physiological and multi-omics data from ad libitum, meal-fed, or CR animals was used to gain insight into pathways associated with improved health and survival. We identified a potential involvement of the glycine-serine-threonine metabolic axis in longevity and related molecular mechanisms. Direct comparison of the different feeding strategies unveiled a pattern of shared pathways of improved health that included short-chain fatty acids and essential PUFA metabolism. These findings were recapitulated in the serum metabolome from nonhuman primates. We propose that the pathways identified might be targeted for their potential role in healthy aging.
Collapse
Affiliation(s)
- Miguel A Aon
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Laboratory of Cardiovascular Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Sarah J Mitchell
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Clara Di Germanio
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Margaux R Ehrlich
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ricki J Colman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Rozalyn M Anderson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53715, USA; Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
152
|
Tippairote T, Janssen S, Chunhabundit R. Restoration of metabolic tempo through time-restricted eating (TRE) as the preventive measure for metabolic diseases. Crit Rev Food Sci Nutr 2020; 61:2444-2453. [PMID: 32551943 DOI: 10.1080/10408398.2020.1781050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The characteristics of healthy bioenergetics are the overall balance of energy intake and expenditure, the alternate switching for different metabolic fuels, and the temporal rhythm of eating and fasting. These three bioenergetic attributes, herein, are termed as the metabolic tempo. Cumulative studies revealed the beneficial health effects of fasting. Most of the fasting regimens harness their innate mechanisms of enhancing metabolic fuel switching, thus improving metabolic flexibility. The emerging time-restricted eating (TRE) regimen includes the restoration of diurnal eating and fasting rhythms, improve the metabolic flexibility, while spontaneously reduces the food intake despite the ad-libitum eating. TRE thus simultaneously improves all three bioenergetic-tempo attributes when compared to the energy balance control in general obesity control. We reviewed fifteen human studies of TRE and TRE-liked interventions from 2007 to 2019. These studies reported promising beneficial metabolic effects on body weight, glycemic, and lipid controls while demonstrating most of the fasting-related metabolic and epigenetic responses in overweight and obese individuals. TRE is practically possible for long-termed implementation. Despite its potentials to restore the underlying dysregulated bioenergetics., there is no study confirming that TRE could prevent the development of common metabolic diseases in healthy subjects after long-term implementation. This gap of knowledge warrants future investigation.
Collapse
Affiliation(s)
- Torsak Tippairote
- Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok, Thailand.,Nutritional and Environmental Medicine Department, BBH Hospital, Bangkok, Thailand
| | - Sarah Janssen
- Nutritional and Environmental Medicine Department, BBH Hospital, Bangkok, Thailand
| | | |
Collapse
|
153
|
Bose S, Allen AE, Locasale JW. The Molecular Link from Diet to Cancer Cell Metabolism. Mol Cell 2020; 78:1034-1044. [PMID: 32504556 PMCID: PMC7305994 DOI: 10.1016/j.molcel.2020.05.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022]
Abstract
Malignant cells remodel their metabolism to meet the demands of uncontrolled cell proliferation. These demands lead to differential requirements in energy, biosynthetic precursors, and signaling intermediates. Both genetic programs arising from oncogenic events and transcriptional programs and epigenomic events are important in providing the necessary metabolic network activity. Accumulating evidence has established that environmental factors play a major role in shaping cancer cell metabolism. For metabolism, diet and nutrition are the major environmental aspects and have emerged as key components in determining cancer cell metabolism. In this review, we discuss these emerging concepts in cancer metabolism and how diet and nutrition influence cancer cell metabolism.
Collapse
Affiliation(s)
- Shree Bose
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Annamarie E Allen
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
154
|
Corina A, Abrudan MB, Nikolic D, Cӑtoi AF, Chianetta R, Castellino G, Citarrella R, Stoian AP, Pérez-Martínez P, Rizzo M. Effects of Aging and Diet on Cardioprotection and Cardiometabolic Risk Markers. Curr Pharm Des 2020; 25:3704-3714. [PMID: 31692432 DOI: 10.2174/1381612825666191105111232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022]
Abstract
The prevalence of several diseases increases by age, including cardiovascular diseases, which are the leading cause of morbidity and mortality worldwide. Aging, as a complex process characterized by senescence, triggers various pathways, such as oxidative stress, systemic inflammation, metabolism dysfunction, telomere shortening, mitochondrial dysfunction and deregulated autophagy. A better understanding of the mechanisms underlying senescence may lead to the development of new therapeutic targets and strategies for age-related pathologies and extend the healthy lifespan. Modulating lifestyle risk factors and adopting healthy dietary patterns remain significant tools in delaying the aging process, decreasing age-associated comorbidities and mortality, increasing life expectancy and consequently, preventing the development of cardiovascular disease. Furthermore, such a strategy represents the most cost-effective approach, and the quality of life of the subjects may be significantly improved. An integrated, personalized approach targeting cardiometabolic aging and frailty is suggested in daily clinical practice. However, it should be initiated from an early age. Moreover, there is a need for further well designed and controlled studies in order to elucidate a link between the time of feeding, longevity and cardiovascular prevention. In the future, it is expected that the pharmacological treatment in cardioprotective management will be necessary, accompanied by equally important lifestyle interventions and adjunctive exercise.
Collapse
Affiliation(s)
- Andreea Corina
- Lipids and Atherosclerosis Research Unit, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain.,CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Cordoba, Spain
| | - Maria B Abrudan
- Department of Pharmaceutical Technology and Biopharmaceutics, "Iuliu Hațieganu", University of Medicine and Pharmacy, Faculty of Pharmacy, Cluj-Napoca, Romania
| | - Dragana Nikolic
- PROMISE Department, University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Adriana F Cӑtoi
- Pathophysiology Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Roberta Chianetta
- PROMISE Department, University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Giuseppa Castellino
- PROMISE Department, University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | | | - Anca P Stoian
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Pablo Pérez-Martínez
- Lipids and Atherosclerosis Research Unit, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain.,CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Cordoba, Spain
| | - Manfredi Rizzo
- PROMISE Department, University of Palermo, Palermo, Italy
| |
Collapse
|
155
|
Ng LT, Ng LF, Tang RMY, Barardo D, Halliwell B, Moore PK, Gruber J. Lifespan and healthspan benefits of exogenous H 2S in C. elegans are independent from effects downstream of eat-2 mutation. NPJ Aging Mech Dis 2020; 6:6. [PMID: 32566245 PMCID: PMC7287109 DOI: 10.1038/s41514-020-0044-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 03/12/2020] [Indexed: 12/23/2022] Open
Abstract
Caloric restriction (CR) is one of the most effective interventions to prolong lifespan and promote health. Recently, it has been suggested that hydrogen sulfide (H2S) may play a pivotal role in mediating some of these CR-associated benefits. While toxic at high concentrations, H2S at lower concentrations can be biologically advantageous. H2S levels can be artificially elevated via H2S-releasing donor drugs. In this study, we explored the function of a novel, slow-releasing H2S donor drug (FW1256) and used it as a tool to investigate H2S in the context of CR and as a potential CR mimetic. We show that exposure to FW1256 extends lifespan and promotes health in Caenorhabditis elegans (C. elegans) more robustly than some previous H2S-releasing compounds, including GYY4137. We looked at the extent to which FW1256 reproduces CR-associated physiological effects in normal-feeding C. elegans. We found that FW1256 promoted healthy longevity to a similar degree as CR but with fewer fitness costs. In contrast to CR, FW1256 actually enhanced overall reproductive capacity and did not reduce adult body length. FW1256 further extended the lifespan of already long-lived eat-2 mutants without further detriments in developmental timing or fertility, but these lifespan and healthspan benefits required H2S exposure to begin early in development. Taken together, these observations suggest that FW1256 delivers exogenous H2S efficiently and supports a role for H2S in mediating longevity benefits of CR. Delivery of H2S via FW1256, however, does not mimic CR perfectly, suggesting that the role of H2S in CR-associated longevity is likely more complex than previously described.
Collapse
Affiliation(s)
- Li Theng Ng
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, 138527 Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600 Singapore.,Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| | - Li Fang Ng
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, 138527 Singapore
| | - Richard Ming Yi Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596 Singapore.,Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore.,NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, 117456 Singapore
| | - Diogo Barardo
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, 138527 Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596 Singapore
| | - Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596 Singapore.,Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| | - Philip Keith Moore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600 Singapore.,Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456 Singapore
| | - Jan Gruber
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, 138527 Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596 Singapore
| |
Collapse
|
156
|
Zheng D, Ratiner K, Elinav E. Circadian Influences of Diet on the Microbiome and Immunity. Trends Immunol 2020; 41:512-530. [DOI: 10.1016/j.it.2020.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 02/08/2023]
|
157
|
Long-Term Feeding of a High-Fat Diet Ameliorated Age-Related Phenotypes in SAMP8 Mice. Nutrients 2020; 12:nu12051416. [PMID: 32423039 PMCID: PMC7285040 DOI: 10.3390/nu12051416] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023] Open
Abstract
High-fat diets (HFD) have been thought to increase the risk of obesity and metabolic syndrome, as well as shorten lifespan. On the other hand, chrono-nutritional studies have shown that time-restricted feeding during active phase significantly suppresses the induction of HFD-induced obesity in mouse model. However, the long-term effects of time-restricted HFD feeding on aging are unknown. Therefore, in this study, we set up a total of four groups: mutual combination of ad libitum feeding or night-time-restricted feeding (NtRF) and an HFD or a control diet. We examined their long-term effects in a senescence-accelerated mouse strain, SAMP8, for over a year. Hearing ability, cognitive function, and other behavioral and physiological indexes were evaluated during the study. Unexpectedly, SAMP8 mice did not show early onset of death caused by the prolonged HFD intake, and both HFD and NtRF retarded age-related hearing loss (AHL). NtRF improved grip strength and cognitive memory scores, while HFD weakly suppressed age-related worsening of the appearance scores associated with the eyes. Notably, the HFD also retarded the progression of AHL in both DBA/2J and C57BL/6J mice. These results suggest that HFD prevents aging unless metabolic disorders occur and that HFD and NtRF are independently effective in retarding aging; thus, the combination of HFD and chrono-nutritional feeding may be an effective anti-aging strategy.
Collapse
|
158
|
Palliyaguru DL, Rudderow AL, Sossong AM, Lewis KN, Younts C, Pearson KJ, Bernier M, de Cabo R. Perinatal diet influences health and survival in a mouse model of leukemia. GeroScience 2020; 42:1147-1155. [PMID: 32394346 DOI: 10.1007/s11357-020-00199-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/29/2020] [Indexed: 11/28/2022] Open
Abstract
The goal of the current study was to determine the role of maternal diet in the perinatal period on the health and survival of the offspring. AKR/J mice, a model described to be susceptible to leukemia development, was used where females were maintained on either standard diet (SD), high sucrose diet, Western diet, or calorie restriction (CR) as they were mated with SD-fed males. Body weights, pregnancy rates, litter size, and litter survival were used as markers of successful pregnancy and pup health. Data indicated that maternal diet had significant effects on litter size, early pup survival, and early pup body weights. As pups matured, the makeup of their respective maternal diet was a predictor of adult metabolic health and survival. Overall, these results suggest that perinatal maternal diet is an important determinant of the health and survival of the offspring and that these effects continue well into adulthood, strongly correlating with lifespan.
Collapse
Affiliation(s)
- Dushani L Palliyaguru
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Annamaria L Rudderow
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alex M Sossong
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Kaitlyn N Lewis
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Caitlin Younts
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Kevin J Pearson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| |
Collapse
|
159
|
Hernandez A, Truckenbrod L, Federico Q, Campos K, Moon B, Ferekides N, Hoppe M, D’Agostino D, Burke S. Metabolic switching is impaired by aging and facilitated by ketosis independent of glycogen. Aging (Albany NY) 2020; 12:7963-7984. [PMID: 32369441 PMCID: PMC7244089 DOI: 10.18632/aging.103116] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/31/2020] [Indexed: 12/17/2022]
Abstract
The ability to switch between glycolysis and ketosis promotes survival by enabling metabolism through fat oxidation during periods of fasting. Carbohydrate restriction or stress can also elicit metabolic switching. Keto-adapting from glycolysis is delayed in aged rats, but factors mediating this age-related impairment have not been identified. We measured metabolic switching between glycolysis and ketosis, as well as glycogen dynamics, in young and aged rats undergoing time-restricted feeding (TRF) with a standard diet or a low carbohydrate ketogenic diet (KD). TRF alone reversed markers of insulin-related metabolic deficits and accelerated metabolic switching in aged animals. A KD+TRF, however, provided additive benefits on these variables. Remarkably, the ability to keto-adapt was not related to glycogen levels and KD-fed rats showed an enhanced elevation in glucose following epinephrine administration. This study provides new insights into the mechanisms of keto-adaptation demonstrating the utility of dietary interventions to treat metabolic impairments across the lifespan.
Collapse
Affiliation(s)
- Abbi Hernandez
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Leah Truckenbrod
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Quinten Federico
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Keila Campos
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Brianna Moon
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Nedi Ferekides
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Meagan Hoppe
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Dominic D’Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
- Institute for Human and Machine Cognition, Ocala, FL 34471, USA
| | - Sara Burke
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Institute on Aging, University of Florida, Gainesville, FL 32603, USA
| |
Collapse
|
160
|
Affiliation(s)
- Tiffany Dong
- Emory University School of Medicine, Atlanta, GA
| | | | | |
Collapse
|
161
|
Mechanisms of Lifespan Regulation by Calorie Restriction and Intermittent Fasting in Model Organisms. Nutrients 2020; 12:nu12041194. [PMID: 32344591 PMCID: PMC7230387 DOI: 10.3390/nu12041194] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Genetic and pharmacological interventions have successfully extended healthspan and lifespan in animals, but their genetic interventions are not appropriate options for human applications and pharmacological intervention needs more solid clinical evidence. Consequently, dietary manipulations are the only practical and probable strategies to promote health and longevity in humans. Caloric restriction (CR), reduction of calorie intake to a level that does not compromise overall health, has been considered as being one of the most promising dietary interventions to extend lifespan in humans. Although it is straightforward, continuous reduction of calorie or food intake is not easy to practice in real lives of humans. Recently, fasting-related interventions such as intermittent fasting (IF) and time-restricted feeding (TRF) have emerged as alternatives of CR. Here, we review the history of CR and fasting-related strategies in animal models, discuss the molecular mechanisms underlying these interventions, and propose future directions that can fill the missing gaps in the current understanding of these dietary interventions. CR and fasting appear to extend lifespan by both partially overlapping common mechanisms such as the target of rapamycin (TOR) pathway and circadian clock, and distinct independent mechanisms that remain to be discovered. We propose that a systems approach combining global transcriptomic, metabolomic, and proteomic analyses followed by genetic perturbation studies targeting multiple candidate pathways will allow us to better understand how CR and fasting interact with each other to promote longevity.
Collapse
|
162
|
Gerisch B, Tharyan RG, Mak J, Denzel SI, Popkes-van Oepen T, Henn N, Antebi A. HLH-30/TFEB Is a Master Regulator of Reproductive Quiescence. Dev Cell 2020; 53:316-329.e5. [PMID: 32302543 DOI: 10.1016/j.devcel.2020.03.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 01/28/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022]
Abstract
All animals have evolved the ability to survive nutrient deprivation, and nutrient signaling pathways are conserved modulators of health and disease. In C. elegans, late-larval starvation provokes the adult reproductive diapause (ARD), a long-lived quiescent state that enables survival for months without food, yet underlying molecular mechanisms remain unknown. Here, we show that ARD is distinct from other forms of diapause, showing little requirement for canonical longevity pathways, autophagy, and fat metabolism. Instead it requires the HLH-30/TFEB transcription factor to promote the morphological and physiological remodeling involved in ARD entry, survival, and recovery, suggesting that HLH-30 is a master regulator of reproductive quiescence. HLH-30 transcriptome and genetic analyses reveal that Max-like HLH factors, AMP-kinase, mTOR, protein synthesis, and mitochondrial fusion are target processes that promote ARD longevity. ARD thus rewires metabolism to ensure long-term survival and may illuminate similar mechanisms acting in stem cell quiescence and long-term fasting.
Collapse
Affiliation(s)
- Birgit Gerisch
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Rebecca George Tharyan
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Jennifer Mak
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Sarah I Denzel
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany
| | - Till Popkes-van Oepen
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany
| | - Nadine Henn
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Adam Antebi
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany.
| |
Collapse
|
163
|
Freire T, Senior AM, Perks R, Pulpitel T, Clark X, Brandon AE, Wahl D, Hatchwell L, Le Couteur DG, Cooney GJ, Larance M, Simpson SJ, Solon-Biet SM. Sex-specific metabolic responses to 6 hours of fasting during the active phase in young mice. J Physiol 2020; 598:2081-2092. [PMID: 32198893 DOI: 10.1113/jp278806] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Night time/active phase food restriction for 6 h impaired glucose intolerance in young male and female mice. Females displayed increased capacity for lipogenesis and triglyceride storage in response to a short daily fast. Females had lower fasting insulin levels and an increased potential for utilizing fat for energy through β-oxidation compared to males. The need for the inclusion of both sexes, and the treatment of sex as an independent variable, is emphasized within the context of this fasting regime. ABSTRACT There is growing interest in understanding the mechanistic significance and benefits of fasting physiology in combating obesity. Increasing the fasting phase of a normal day can promote restoration and repair mechanisms that occur during the post-absorptive period. Most studies exploring the effect of restricting food access on mitigating obesity have done so with a large bias towards the use of male mice. Here, we disentangle the roles of sex, food intake and food withdrawal in the response to a short-term daily fasting intervention, in which food was removed for 6 h in the dark/active phase of young, 8-week-old mice. We showed that the removal of food during the dark phase impaired glucose tolerance in males and females, possibly due to the circadian disruption induced by this feeding protocol. Although both sexes demonstrated similar patterns of food intake, body composition and various metabolic markers, there were clear sex differences in the magnitude and extent of these responses. While females displayed enhanced capacity for lipogenesis and triglyceride storage, they also had low fasting insulin levels and an increased potential for utilizing available energy sources such as fat for energy through β-oxidation. Our results highlight the intrinsic biological and metabolic disparities between male and female mice, emphasizing the growing need for the inclusion of both sexes in scientific research. Furthermore, our results illustrate sex-specific metabolic pathways that regulate lipogenesis, obesity and overall metabolic health.
Collapse
Affiliation(s)
- Therese Freire
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - Ruth Perks
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - Ximonie Clark
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - Amanda E Brandon
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, NSW, Australia
| | - Devin Wahl
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, NSW, Australia
| | - Luke Hatchwell
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Ageing and Alzheimer's Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord, NSW, Australia
| | - Gregory J Cooney
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, NSW, Australia
| | - Mark Larance
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
164
|
Velingkaar N, Mezhnina V, Poe A, Makwana K, Tulsian R, Kondratov RV. Reduced caloric intake and periodic fasting independently contribute to metabolic effects of caloric restriction. Aging Cell 2020; 19:e13138. [PMID: 32159926 PMCID: PMC7189989 DOI: 10.1111/acel.13138] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/01/2020] [Accepted: 02/23/2020] [Indexed: 12/13/2022] Open
Abstract
Caloric restriction (CR) has positive effects on health and longevity. CR in mammals implements time‐restricted (TR) feeding, a short period of feeding followed by prolonged fasting. Periodic fasting, in the form of TR or mealtime, improves metabolism without reduction in caloric intake. In order to understand the relative contribution of reduced food intake and periodic fasting to the health benefits of CR, we compared physiological and metabolic changes induced by CR and TR (without reduced food intake) in mice. CR significantly reduced blood glucose and insulin around the clock, improved glucose tolerance, and increased insulin sensitivity (IS). TR reduced blood insulin and increased insulin sensitivity, but in contrast to CR, TR did not improve glucose homeostasis. Liver expression of circadian clock genes was affected by both diets while the mRNA expression of glucose metabolism genes was significantly induced by CR, and not by TR, which is in agreement with the minor effect of TR on glucose metabolism. Thus, periodic fasting contributes to some metabolic benefits of CR, but TR is metabolically different from CR. This difference might contribute to differential effects of CR and TR on longevity.
Collapse
Affiliation(s)
- Nikkhil Velingkaar
- Department of Biological, Geological, and Environmental Sciences and Center for Gene Regulation in Health and Disease Cleveland State University Cleveland OH USA
| | - Volha Mezhnina
- Department of Biological, Geological, and Environmental Sciences and Center for Gene Regulation in Health and Disease Cleveland State University Cleveland OH USA
- Institute of Genetics and Cytology National Academy of Science of Belarus Minsk Belarus
| | - Allan Poe
- Department of Biological, Geological, and Environmental Sciences and Center for Gene Regulation in Health and Disease Cleveland State University Cleveland OH USA
| | - Kuldeep Makwana
- Department of Biological, Geological, and Environmental Sciences and Center for Gene Regulation in Health and Disease Cleveland State University Cleveland OH USA
| | - Richa Tulsian
- Department of Biological, Geological, and Environmental Sciences and Center for Gene Regulation in Health and Disease Cleveland State University Cleveland OH USA
| | - Roman V. Kondratov
- Department of Biological, Geological, and Environmental Sciences and Center for Gene Regulation in Health and Disease Cleveland State University Cleveland OH USA
| |
Collapse
|
165
|
Li L, Su Y, Li F, Wang Y, Ma Z, Li Z, Su J. The effects of daily fasting hours on shaping gut microbiota in mice. BMC Microbiol 2020; 20:65. [PMID: 32209070 PMCID: PMC7092480 DOI: 10.1186/s12866-020-01754-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Background It has recently been reported that intermittent fasting shapes the gut microbiota to benefit health, but this effect may be influenced to the exact fasting protocols. The purpose of this study was to assess the effects of different daily fasting hours on shaping the gut microbiota in mice. Healthy C57BL/6 J male mice were subjected to 12, 16 or 20 h fasting per day for 1 month, and then fed ad libitum for an extended month. Gut microbiota was analyzed by 16S rRNA gene-based sequencing and food intake was recorded as well. Results We found that cumulative food intake was not changed in the group with 12 h daily fasting, but significantly decreased in the 16 and 20 h fasting groups. The composition of gut microbiota was altered by all these types of intermittent fasting. At genus level, 16 h fasting led to increased level of Akkermansia and decreased level of Alistipes, but these effects disappeared after the cessation of fasting. No taxonomic differences were identified in the other two groups. Conclusions These data indicated that intermittent fasting shapes gut microbiota in healthy mice, and the length of daily fasting interval may influence the outcome of intermittent fasting.
Collapse
Affiliation(s)
- Linghao Li
- China-Malaysia National Joint Laboratory, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Yuxin Su
- China-Malaysia National Joint Laboratory, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Fanglin Li
- China-Malaysia National Joint Laboratory, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Yueying Wang
- China-Malaysia National Joint Laboratory, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Zhongren Ma
- China-Malaysia National Joint Laboratory, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Zhuo Li
- China-Malaysia National Joint Laboratory, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Junhong Su
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, No.727 South Jingming Rd., Chenggong District, Kunming, China. .,Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
166
|
Bellantuono I, de Cabo R, Ehninger D, Di Germanio C, Lawrie A, Miller J, Mitchell SJ, Navas-Enamorado I, Potter PK, Tchkonia T, Trejo JL, Lamming DW. A toolbox for the longitudinal assessment of healthspan in aging mice. Nat Protoc 2020; 15:540-574. [PMID: 31915391 PMCID: PMC7002283 DOI: 10.1038/s41596-019-0256-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022]
Abstract
The number of people aged over 65 is expected to double in the next 30 years. For many, living longer will mean spending more years with the burdens of chronic diseases such as Alzheimer's disease, cardiovascular disease, and diabetes. Although researchers have made rapid progress in developing geroprotective interventions that target mechanisms of aging and delay or prevent the onset of multiple concurrent age-related diseases, a lack of standardized techniques to assess healthspan in preclinical murine studies has resulted in reduced reproducibility and slow progress. To overcome this, major centers in Europe and the United States skilled in healthspan analysis came together to agree on a toolbox of techniques that can be used to consistently assess the healthspan of mice. Here, we describe the agreed toolbox, which contains protocols for echocardiography, novel object recognition, grip strength, rotarod, glucose tolerance test (GTT) and insulin tolerance test (ITT), body composition, and energy expenditure. The protocols can be performed longitudinally in the same mouse over a period of 4-6 weeks to test how candidate geroprotectors affect cardiac, cognitive, neuromuscular, and metabolic health.
Collapse
Affiliation(s)
- I Bellantuono
- Department of Oncology and Metabolism, Healthy Lifespan Institute and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing, University of Sheffield, Sheffield, UK.
| | - R de Cabo
- Translational Gerontology Branch, National Institutes of Health, Baltimore, MD, USA
| | - D Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1, Bonn, Germany
| | - C Di Germanio
- Translational Gerontology Branch, National Institutes of Health, Baltimore, MD, USA
| | - A Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - J Miller
- Robert and Arlene KogodCenter on Aging, Mayo Clinic, Rochester, MN, USA
| | - S J Mitchell
- Department of Molecular Medicine, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - I Navas-Enamorado
- Translational Gerontology Branch, National Institutes of Health, Baltimore, MD, USA
| | - P K Potter
- Department of Biological and Life Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxfordshire, UK
| | - T Tchkonia
- Robert and Arlene KogodCenter on Aging, Mayo Clinic, Rochester, MN, USA
| | - J L Trejo
- Department of Translational Neuroscience, Cajal Institute (CSIC), Madrid, Spain
| | - D W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
167
|
McCracken AW, Adams G, Hartshorne L, Tatar M, Simons MJP. The hidden costs of dietary restriction: Implications for its evolutionary and mechanistic origins. SCIENCE ADVANCES 2020; 6:eaay3047. [PMID: 32128403 PMCID: PMC7034997 DOI: 10.1126/sciadv.aay3047] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/04/2019] [Indexed: 05/14/2023]
Abstract
Dietary restriction (DR) extends life span across taxa. Despite considerable research, universal mechanisms of DR have not been identified, limiting its translational potential. Guided by the conviction that DR evolved as an adaptive, pro-longevity physiological response to food scarcity, biomedical science has interpreted DR as an activator of pro-longevity molecular pathways. Current evolutionary theory predicts that organisms invest in their soma during DR, and thus when resource availability improves, should outcompete rich-fed controls in survival and/or reproduction. Testing this prediction in Drosophila melanogaster (N > 66,000 across 11 genotypes), our experiments revealed substantial, unexpected mortality costs when flies returned to a rich diet following DR. The physiological effects of DR should therefore not be interpreted as intrinsically pro-longevity, acting via somatic maintenance. We suggest DR could alternatively be considered an escape from costs incurred under nutrient-rich conditions, in addition to costs associated with DR.
Collapse
Affiliation(s)
- Andrew W. McCracken
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Gracie Adams
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Laura Hartshorne
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Mirre J. P. Simons
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
168
|
Short-term time-restricted feeding is safe and feasible in non-obese healthy midlife and older adults. GeroScience 2020; 42:667-686. [PMID: 31975053 DOI: 10.1007/s11357-020-00156-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/09/2020] [Indexed: 01/21/2023] Open
Abstract
Chronic calorie restriction (CR) improves cardiovascular function and several other physiological markers of healthspan. However, CR is impractical in non-obese older humans due to potential loss of lean mass and bone density, poor adherence, and risk of malnutrition. Time-restricted feeding (TRF), which limits the daily feeding period without requiring a reduction in calorie intake, may be a promising alternative healthspan-extending strategy for midlife and older adults; however, there is limited evidence for its feasibility and efficacy in humans. We conducted a randomized, controlled pilot study to assess the safety, tolerability, and overall feasibility of short-term TRF (eating <8 h day-1 for 6 weeks) without weight loss in healthy non-obese midlife and older adults, while gaining initial insight into potential efficacy for improving cardiovascular function and other indicators of healthspan. TRF was safe and well-tolerated, associated with excellent adherence and reduced hunger, and did not influence lean mass, bone density, or nutrient intake. Cardiovascular function was not enhanced by short-term TRF in this healthy cohort, but functional (endurance) capacity and glucose tolerance were modestly improved. These results provide a foundation for conducting larger clinical studies of TRF in midlife and older adults, including trials with a longer treatment duration.
Collapse
|
169
|
von Frieling J, Roeder T. Factors that affect the translation of dietary restriction into a longer life. IUBMB Life 2019; 72:814-824. [PMID: 31889425 DOI: 10.1002/iub.2224] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023]
Abstract
Nutritional interventions, such as dietary or calorie restriction, are known to have a variety of health-promoting effects. The most impressive are the direct effects on life expectancy, which have been reproduced in many animal models. A variety of dietary restriction protocols have been described, which differ either in their macronutrient composition or in the time window for consumption. Mechanistically, the effects of dietary restriction are mediated mainly through signaling pathways that have central roles in the maintenance of cellular energy balance. Among these, target of rapamycin and insulin signaling appear to be the most important. Such nutritional interventions can have their effects in two different ways: either by direct interaction with the metabolism of the host organism, or by modulating the composition and performance of its endogenous microbiome. Various dietary restriction regimens have been identified that significantly alter the microbiome and thus profoundly modulate host metabolism. This review aims to discuss the mechanisms by which dietary restriction can affect life expectancy, and in particular the role of the microbiome.
Collapse
Affiliation(s)
- Jakob von Frieling
- Department of Zoology, Molecular Physiology, Kiel University, Kiel, Germany
| | - Thomas Roeder
- Department of Zoology, Molecular Physiology, Kiel University, Kiel, Germany.,DZL, German Center for Lung Research, ARCN, Kiel, Germany
| |
Collapse
|
170
|
Brandhorst S, Longo VD. Dietary Restrictions and Nutrition in the Prevention and Treatment of Cardiovascular Disease. Circ Res 2019; 124:952-965. [PMID: 30870119 DOI: 10.1161/circresaha.118.313352] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in many developed countries and remains one of the major diseases strongly affected by the diet. Nutrition can affect CVD directly by contributing to the accumulation of vascular plaques and also indirectly by regulating the rate of aging. This review summarizes research on nutrition and CVD incidence based on a multipillar system that includes basic research focused on aging, epidemiological studies, clinical studies, and studies of centenarians. The relevant research linking nutrition and CVD with focus on macronutrients and aging will be highlighted. We will review some of the most relevant studies on nutrition and CVD treatment, also focusing on interventions known to delay aging. We will discuss both everyday dietary compositions, as well as intermittent and periodic fasting interventions with the potential to prevent and treat CVD.
Collapse
Affiliation(s)
- Sebastian Brandhorst
- From the Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles (S.B., V.D.L.)
| | - Valter D Longo
- From the Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles (S.B., V.D.L.).,Institute of Molecular Oncology, Italian Foundation for Cancer Research, Milan (V.D.L.)
| |
Collapse
|
171
|
Sabbatinelli J, Prattichizzo F, Olivieri F, Procopio AD, Rippo MR, Giuliani A. Where Metabolism Meets Senescence: Focus on Endothelial Cells. Front Physiol 2019; 10:1523. [PMID: 31920721 PMCID: PMC6930181 DOI: 10.3389/fphys.2019.01523] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Despite the decline in their proliferative potential, senescent cells display a high metabolic activity. Senescent cells have been shown to acquire a more glycolytic state even in presence of high oxygen levels, in a way similar to cancer cells. The diversion of pyruvate, the final product of glycolysis, away from oxidative phosphorylation results in an altered bioenergetic state and may occur as a response to the enhanced oxidative stress caused by the accumulation of dysfunctional mitochondria. This metabolic shift leads to increased AMP/ATP and ADP/ATP ratios, to the subsequent AMPK activation, and ultimately to p53-mediated growth arrest. Mounting evidences suggest that metabolic reprogramming is critical to direct considerable amounts of energy toward specific activities related to the senescent state, including the senescence-associated secretory phenotype (SASP) and the modulation of immune responses within senescent cell tissue microenvironment. Interestingly, despite the relative abundance of oxygen in the vascular compartment, healthy endothelial cells (ECs) produce most of their ATP content from the anaerobic conversion of glucose to lactate. Their high glycolytic rate further increases during senescence. Alterations in EC metabolism have been identified in age-related diseases (ARDs) associated with a dysfunctional vasculature, including atherosclerosis, type 2 diabetes and cardiovascular diseases. In particular, higher production of reactive oxygen species deriving from a variety of enzymatic sources, including uncoupled endothelial nitric oxide synthase and the electron transport chain, causes DNA damage and activates the NAD+-consuming enzymes polyADP-ribose polymerase 1 (PARP1). These non-physiological mechanisms drive the impairment of the glycolytic flux and the diversion of glycolytic intermediates into many pathological pathways. Of note, accumulation of senescent ECs has been reported in the context of ARDs. Through their pro-oxidant, pro-inflammatory, vasoconstrictor, and prothrombotic activities, they negatively impact on vascular physiology, promoting both the onset and development of ARDs. Here, we review the current knowledge on the cellular senescence-related metabolic changes and their contribution to the mechanisms underlying the pathogenesis of ARDs, with a particular focus on ECs. Moreover, current and potential interventions aimed at modulating EC metabolism, in order to prevent or delay ARD onset, will be discussed.
Collapse
Affiliation(s)
- Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| | | | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
172
|
Current nutritional and pharmacological anti-aging interventions. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165612. [PMID: 31816437 DOI: 10.1016/j.bbadis.2019.165612] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022]
Abstract
Aging is the main risk factor for chronic diseases and disablement in human societies with a great impact in social and health care expenditures. So far, aging and, eventually, death are unavoidable. Nevertheless, research efforts on aging-associated diseases with the aim not only to extend life span but also to increment health span in an attempt to delay, stop and even reverse the aging process have not stopped growing. Caloric restriction extends both health and life span in several short-lived experimental models and has brought to light the role of different molecular effectors involved in nutrient sensing pathways and longevity. This opens the possibility of modulating these molecular effectors also in humans to increase longevity and health span. The difficulty to implement caloric restricted diets in humans has led to the development of new bearable diets such as time-restricted feeding, intermittent fasting or diets with limited amounts of some nutrients and to the search of pharmacological agents, targeted to the effectors that mediate the extension of life and health span in response to these anti-aging diets. Pharmacological approaches that eliminate senescent cells or prevent primary causes of aging such as telomere attrition also emerge as potential anti-aging strategies. In the present article, we review these possible nutritional and pharmacological interventions designed to mitigate and/or delay the aging process and to increase health and life span.
Collapse
|
173
|
Timing of Calorie Restriction in Mice Impacts Host Metabolic Phenotype with Correlative Changes in Gut Microbiota. mSystems 2019; 4:4/6/e00348-19. [PMID: 31796564 PMCID: PMC6890928 DOI: 10.1128/msystems.00348-19] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aberrant feeding patterns whereby people eat more frequently throughout the day and with a bias toward late-night eating are prevalent in society today. However, whether restriction of food to daytime in comparison to nighttime, coupled with restricted calorie intake, can influence gut microbiota, metabolism, and overall health requires further investigation. We surveyed the effects of the shift in feeding time on gut microbiota and metabolic phenotype in calorie-restricted mice and found that avoiding eating during the rest period may generate more beneficial effects in mice. This work strengthens the evidence for using “when to eat” as an intervention to improve health during calorie restriction. Calorie restriction (CR) is accompanied by self-imposed daily restriction of food intake and an extended fasting period between meals. The impact of restricting feeding to the dark or light phase on the effects of CR remains elusive. Here, light-fed CR mice showed physiological changes, such as muscle loss, concomitant with changes in the gut microbiota structure and composition. After switching to ad libitum access to food, light-fed mice had a period of food-craving behavior and short-lived physiological changes, while dark-fed mice displayed lasting changes in fat accumulation, glucose metabolism, intestinal barrier function, and systemic inflammatory markers. Moreover, the gut microbiota was modulated by when the food was consumed, and the most abundant Lactobacillus operational taxonomic unit (OTU) promoted by CR was enhanced in dark-fed mice. After switching to ad libitum feeding, the gut microbiota of dark-fed mice returned to the state resembling that of mice fed normal chow ad libitum, but that of light-fed mice was still significantly different from the other two groups. Together, these data indicate that for CR, restricting food consumption to the active phase brought better metabolic phenotype associated with potentially beneficial structural shifts in the gut microbiota. IMPORTANCE Aberrant feeding patterns whereby people eat more frequently throughout the day and with a bias toward late-night eating are prevalent in society today. However, whether restriction of food to daytime in comparison to nighttime, coupled with restricted calorie intake, can influence gut microbiota, metabolism, and overall health requires further investigation. We surveyed the effects of the shift in feeding time on gut microbiota and metabolic phenotype in calorie-restricted mice and found that avoiding eating during the rest period may generate more beneficial effects in mice. This work strengthens the evidence for using “when to eat” as an intervention to improve health during calorie restriction.
Collapse
|
174
|
Yong-Quan Ng G, Yang-Wei Fann D, Jo DG, Sobey CG, Arumugam TV. Dietary Restriction and Epigenetics: Part I. CONDITIONING MEDICINE 2019; 2:284-299. [PMID: 32039345 PMCID: PMC7007115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Biological aging occurs concomitantly with chronological aging and is commonly burdened by the development of age-related conditions, such as neurodegenerative, cardiovascular, and a myriad of metabolic diseases. With a current global shift in disease epidemiology associated with aging and the resultant social, economic, and healthcare burdens faced by many countries, the need to achieve successful aging has fueled efforts to address this problem. Aging is a complex biological phenomenon that has confounded much of the historical research effort to understand it, with still limited knowledge of the underlying molecular mechanisms. Interestingly, dietary restriction (DR) is one intervention that produces anti-aging effects from simple organisms to mammals. Research into DR has revealed robust systemic effects that can result in attenuation of age-related diseases via a myriad of molecular mechanisms. Given that numerous age-associated diseases are often polygenic and affect individuals differently, it is possible that they are confounded by interactions between environmental influences and the genome, a process termed 'epigenetics'. In part one of the review, we summarize the different variants of DR regimens and their corresponding mechanism(s) and resultant effects, as well as in-depth analysis of current knowledge of the epigenetic landscape.
Collapse
Affiliation(s)
- Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Christopher G. Sobey
- Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V. Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
175
|
Molecular mechanisms and physiological importance of circadian rhythms. Nat Rev Mol Cell Biol 2019; 21:67-84. [PMID: 31768006 DOI: 10.1038/s41580-019-0179-2] [Citation(s) in RCA: 604] [Impact Index Per Article: 120.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 12/12/2022]
Abstract
To accommodate daily recurring environmental changes, animals show cyclic variations in behaviour and physiology, which include prominent behavioural states such as sleep-wake cycles but also a host of less conspicuous oscillations in neurological, metabolic, endocrine, cardiovascular and immune functions. Circadian rhythmicity is created endogenously by genetically encoded molecular clocks, whose components cooperate to generate cyclic changes in their own abundance and activity, with a periodicity of about a day. Throughout the body, such molecular clocks convey temporal control to the function of organs and tissues by regulating pertinent downstream programmes. Synchrony between the different circadian oscillators and resonance with the solar day is largely enabled by a neural pacemaker, which is directly responsive to certain environmental cues and able to transmit internal time-of-day representations to the entire body. In this Review, we discuss aspects of the circadian clock in Drosophila melanogaster and mammals, including the components of these molecular oscillators, the function and mechanisms of action of central and peripheral clocks, their synchronization and their relevance to human health.
Collapse
|
176
|
Hu S, Wang L, Togo J, Yang D, Xu Y, Wu Y, Douglas A, Speakman JR. The carbohydrate-insulin model does not explain the impact of varying dietary macronutrients on the body weight and adiposity of mice. Mol Metab 2019; 32:27-43. [PMID: 32029228 PMCID: PMC6938849 DOI: 10.1016/j.molmet.2019.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives The carbohydrate-insulin model (CIM) predicts that increases in fasting and post-prandial insulin in response to dietary carbohydrates stimulate energy intake and lower energy expenditures, leading to positive energy balance and weight gain. The objective of the present study was to directly test the CIM's predictions using C57BL/6 mice. Methods Diets were designed by altering dietary carbohydrates with either fixed protein or fat content and were fed to C57BL/6 mice acutely or chronically for 12 weeks. The body weight, body composition, food intake, and energy expenditures of the mice were measured. Their fasting and post-prandial glucose and insulin levels were also measured. RNA-seq was performed on RNA from the hypothalamus and subcutaneous white adipose tissue. Pathway analysis was conducted using IPA. Results Only the post-prandial insulin and fasting glucose levels followed the CIM's predictions. The lipolysis and leptin signaling pathways in the sWAT were inhibited in relation to the elevated fasting insulin, supporting the CIM's predicted impact of high insulin. However, because higher fasting insulin was unrelated to carbohydrate intake, the overall pattern did not support the model. Moreover, the hypothalamic hunger pathways were inhibited in relation to the increased fasting insulin, and the energy intake was not increased. The browning pathway in the sWAT was inhibited at higher insulin levels, but the daily energy expenditure was not altered. Conclusions Two of the predictions were partially supported (and hence also partially not supported) and the other three predictions were not supported. We conclude that the CIM does not explain the impact of dietary macronutrients on adiposity in mice. Higher fasting insulin related to inhibited lipolysis and leptin pathways in sWAT, supporting CIM. Higher fasting insulin related to inhibited hypothalamic hunger pathway, contrasting CIM. Fasting insulin decreased with higher dietary carbohydrate, overall contrasting CIM. Higher dietary carbohydrate did not lead to greater EI/adiposity, or lowered EE.
Collapse
Affiliation(s)
- Sumei Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Jacques Togo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yanchao Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yingga Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK; CAS Center for Excellence in Animal Evolution and Genetics (CCEAEG), Kunming, PR China.
| |
Collapse
|
177
|
Zinöcker MK, Lindseth IA. Two Bad Diets for Monkeys. Obesity (Silver Spring) 2019; 27:1553. [PMID: 31411382 DOI: 10.1002/oby.22588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
178
|
Shively CA, Appt SE, Vitolins MZ, Uberseder B, Michalson KT, Silverstein-Metzler MG, Register TC. Response to "Two Bad Diets for Monkeys". Obesity (Silver Spring) 2019; 27:1554. [PMID: 31420940 DOI: 10.1002/oby.22587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Carol A Shively
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Susan E Appt
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mara Z Vitolins
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Beth Uberseder
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kristofer T Michalson
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Marnie G Silverstein-Metzler
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Thomas C Register
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
179
|
Natarajan N, Vujic A, Das J, Wang AC, Phu KK, Kiehm SH, Ricci-Blair EM, Zhu AY, Vaughan KL, Colman RJ, Mattison JA, Lee RT. Effect of dietary fat and sucrose consumption on cardiac fibrosis in mice and rhesus monkeys. JCI Insight 2019; 4:128685. [PMID: 31415241 PMCID: PMC6795382 DOI: 10.1172/jci.insight.128685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/12/2019] [Indexed: 01/11/2023] Open
Abstract
Calorie restriction (CR) improved health span in 2 longitudinal studies in nonhuman primates (NHPs), yet only the University of Wisconsin (UW) study demonstrated an increase in survival in CR monkeys relative to controls; the National Institute on Aging (NIA) study did not. Here, analysis of left ventricle samples showed that CR did not reduce cardiac fibrosis relative to controls. However, there was a 5.9-fold increase of total fibrosis in UW hearts, compared with NIA hearts. Diet composition was a prominent difference between the studies; therefore, we used the NHP diets to characterize diet-associated molecular and functional changes in the hearts of mice. Consistent with the findings from the NHP samples, mice fed a UW or a modified NIA diet with increased sucrose and fat developed greater cardiac fibrosis compared with mice fed the NIA diet, and transcriptomics analysis revealed diet-induced activation of myocardial oxidative phosphorylation and cardiac muscle contraction pathways.
Collapse
Affiliation(s)
- Niranjana Natarajan
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Ana Vujic
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jishnu Das
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Annie C. Wang
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Krystal K. Phu
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Spencer H. Kiehm
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Elisabeth M. Ricci-Blair
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Anthony Y. Zhu
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Kelli L. Vaughan
- National Institute on Aging, NIH, Baltimore, Maryland, USA
- SoBran BioSciences, SoBran Inc., Burtonsville, Maryland, USA
| | - Ricki J. Colman
- Department of Cell and Regenerative Biology and Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
180
|
Stekovic S, Hofer SJ, Tripolt N, Aon MA, Royer P, Pein L, Stadler JT, Pendl T, Prietl B, Url J, Schroeder S, Tadic J, Eisenberg T, Magnes C, Stumpe M, Zuegner E, Bordag N, Riedl R, Schmidt A, Kolesnik E, Verheyen N, Springer A, Madl T, Sinner F, de Cabo R, Kroemer G, Obermayer-Pietsch B, Dengjel J, Sourij H, Pieber TR, Madeo F. Alternate Day Fasting Improves Physiological and Molecular Markers of Aging in Healthy, Non-obese Humans. Cell Metab 2019; 30:462-476.e6. [PMID: 31471173 DOI: 10.1016/j.cmet.2019.07.016] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/17/2019] [Accepted: 07/30/2019] [Indexed: 01/11/2023]
Abstract
Caloric restriction and intermittent fasting are known to prolong life- and healthspan in model organisms, while their effects on humans are less well studied. In a randomized controlled trial study (ClinicalTrials.gov identifier: NCT02673515), we show that 4 weeks of strict alternate day fasting (ADF) improved markers of general health in healthy, middle-aged humans while causing a 37% calorie reduction on average. No adverse effects occurred even after >6 months. ADF improved cardiovascular markers, reduced fat mass (particularly the trunk fat), improving the fat-to-lean ratio, and increased β-hydroxybutyrate, even on non-fasting days. On fasting days, the pro-aging amino-acid methionine, among others, was periodically depleted, while polyunsaturated fatty acids were elevated. We found reduced levels sICAM-1 (an age-associated inflammatory marker), low-density lipoprotein, and the metabolic regulator triiodothyronine after long-term ADF. These results shed light on the physiological impact of ADF and supports its safety. ADF could eventually become a clinically relevant intervention.
Collapse
Affiliation(s)
- Slaven Stekovic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria; BioTechMed Graz, Graz 8010, Austria
| | - Norbert Tripolt
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA; Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Philipp Royer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria
| | - Lukas Pein
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria; Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Julia T Stadler
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria; Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria
| | - Barbara Prietl
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Jasmin Url
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Sabrina Schroeder
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria; BioTechMed Graz, Graz 8010, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria; BioTechMed Graz, Graz 8010, Austria; NAWI Graz Central Lab Gracia, NAWI Graz, Graz, Austria
| | - Christoph Magnes
- HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz, Austria
| | - Michael Stumpe
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Elmar Zuegner
- HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz, Austria
| | - Natalie Bordag
- HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz, Austria
| | - Regina Riedl
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria
| | - Albrecht Schmidt
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Ewald Kolesnik
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Nicolas Verheyen
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Anna Springer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/, VI 8010 Graz, Austria
| | - Tobias Madl
- BioTechMed Graz, Graz 8010, Austria; Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/, VI 8010 Graz, Austria
| | - Frank Sinner
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria; HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz, Austria
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France; INSERM U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden; Center of Systems Medicine, Chinese Academy of Science Sciences, Suzhou, China
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland; Department of Dermatology, Medical Center, University of Freiburg, Hauptstr. 7, 79104 Freiburg, Germany
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Thomas R Pieber
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Neue Stiftingtalstraße 2, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Humboldtstraße 50, Graz 8010, Austria; BioTechMed Graz, Graz 8010, Austria.
| |
Collapse
|
181
|
Vázquez-Fonseca L, Schaefer J, Navas-Enamorado I, Santos-Ocaña C, Hernández-Camacho JD, Guerra I, Cascajo MV, Sánchez-Cuesta A, Horvath Z, Siendones E, Jou C, Casado M, Gutiérrez P, Brea-Calvo G, López-Lluch G, Fernández-Ayala DJM, Cortés-Rodríguez AB, Rodríguez-Aguilera JC, Matté C, Ribes A, Prieto-Soler SY, Dominguez-Del-Toro E, Francesco AD, Aon MA, Bernier M, Salviati L, Artuch R, Cabo RD, Jackson S, Navas P. ADCK2 Haploinsufficiency Reduces Mitochondrial Lipid Oxidation and Causes Myopathy Associated with CoQ Deficiency. J Clin Med 2019; 8:jcm8091374. [PMID: 31480808 PMCID: PMC6780728 DOI: 10.3390/jcm8091374] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 01/27/2023] Open
Abstract
Fatty acids and glucose are the main bioenergetic substrates in mammals. Impairment of mitochondrial fatty acid oxidation causes mitochondrial myopathy leading to decreased physical performance. Here, we report that haploinsufficiency of ADCK2, a member of the aarF domain-containing mitochondrial protein kinase family, in human is associated with liver dysfunction and severe mitochondrial myopathy with lipid droplets in skeletal muscle. In order to better understand the etiology of this rare disorder, we generated a heterozygous Adck2 knockout mouse model to perform in vivo and cellular studies using integrated analysis of physiological and omics data (transcriptomics–metabolomics). The data showed that Adck2+/− mice exhibited impaired fatty acid oxidation, liver dysfunction, and mitochondrial myopathy in skeletal muscle resulting in lower physical performance. Significant decrease in Coenzyme Q (CoQ) biosynthesis was observed and supplementation with CoQ partially rescued the phenotype both in the human subject and mouse model. These results indicate that ADCK2 is involved in organismal fatty acid metabolism and in CoQ biosynthesis in skeletal muscle. We propose that patients with isolated myopathies and myopathies involving lipid accumulation be tested for possible ADCK2 defect as they are likely to be responsive to CoQ supplementation.
Collapse
Affiliation(s)
- Luis Vázquez-Fonseca
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, and IRP Città della Speranza, 35100 Padova, Italy
| | - Jochen Schaefer
- Department of Neurology, Carl Gustav Carus University Dresden, 01307 Dresden, Germany
| | - Ignacio Navas-Enamorado
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- Boston University School of Medicine, Boston, MA 02118, USA
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 20201, USA
| | - Carlos Santos-Ocaña
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Juan D Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Ignacio Guerra
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
| | - María V Cascajo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Ana Sánchez-Cuesta
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Zoltan Horvath
- Department of Neurology, Carl Gustav Carus University Dresden, 01307 Dresden, Germany
| | - Emilio Siendones
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
| | - Cristina Jou
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
- Clinical Chemistry and Pathology Departments, Institut de Recerca Sant Joan de Déu, 08000 Barcelona, Spain
| | - Mercedes Casado
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
- Clinical Chemistry and Pathology Departments, Institut de Recerca Sant Joan de Déu, 08000 Barcelona, Spain
| | - Purificación Gutiérrez
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
| | - Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Daniel J M Fernández-Ayala
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Ana B Cortés-Rodríguez
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Juan C Rodríguez-Aguilera
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
| | - Cristiane Matté
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. CEP 90035-003, Porto Alegre, RS, Brazil
| | - Antonia Ribes
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
- Secciód'Errors Congènits del Metabolisme-IBC, Servei de Bioquímica I Genètica Molecular, Hospital Clinic, 08000 Barcelona, Spain
| | | | | | - Andrea di Francesco
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 20201, USA
| | - Miguel A Aon
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 20201, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 20201, USA
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, and IRP Città della Speranza, 35100 Padova, Italy
| | - Rafael Artuch
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain
- Clinical Chemistry and Pathology Departments, Institut de Recerca Sant Joan de Déu, 08000 Barcelona, Spain
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 20201, USA
| | - Sandra Jackson
- Department of Neurology, Carl Gustav Carus University Dresden, 01307 Dresden, Germany
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013 Sevilla, Spain.
- CIBERER, Instituto de Salud Carlos III, 28000 Madrid, Spain.
| |
Collapse
|
182
|
Zhang R, Wang X, Qu JH, Liu B, Zhang P, Zhang T, Fan PC, Wang XM, Xiao GY, Su Y, Xie Y, Liu Y, Pei JF, Zhang ZQ, Hao DL, Xu P, Chen HZ, Liu DP. Caloric Restriction Induces MicroRNAs to Improve Mitochondrial Proteostasis. iScience 2019; 17:155-166. [PMID: 31279933 PMCID: PMC6614116 DOI: 10.1016/j.isci.2019.06.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/07/2019] [Accepted: 06/17/2019] [Indexed: 01/09/2023] Open
Abstract
Both caloric restriction (CR) and mitochondrial proteostasis are linked to longevity, but how CR maintains mitochondrial proteostasis in mammals remains elusive. MicroRNAs (miRNAs) are well known for gene silencing in cytoplasm and have recently been identified in mitochondria, but knowledge regarding their influence on mitochondrial function is limited. Here, we report that CR increases miRNAs, which are required for the CR-induced activation of mitochondrial translation, in mouse liver. The ablation of miR-122, the most abundant miRNA induced by CR, or the retardation of miRNA biogenesis via Drosha knockdown significantly reduces the CR-induced activation of mitochondrial translation. Importantly, CR-induced miRNAs cause the overproduction of mtDNA-encoded proteins, which induces the mitochondrial unfolded protein response (UPRmt), and consequently improves mitochondrial proteostasis and function. These findings establish a physiological role of miRNA-enhanced mitochondrial function during CR and reveal miRNAs as critical mediators of CR in inducing UPRmt to improve mitochondrial proteostasis. CR increases miRNA biogenesis and the global expression of miRNAs in mitochondria miRNAs are critical for CR-induced activation of mitochondrial translation CR-induced miRNAs cause overproduction of mtDNA-encoded proteins and induce UPRmt
Collapse
Affiliation(s)
- Ran Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Xu Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Jia-Hua Qu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Bing Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Peng Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Tao Zhang
- State Key Laboratory of Proteomics, National Centre for Protein Sciences Beijing, Beijing Proteome Research Centre, National Engineering Research Centre for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing 102206, P.R. China
| | - Peng-Cheng Fan
- State Key Laboratory of Proteomics, National Centre for Protein Sciences Beijing, Beijing Proteome Research Centre, National Engineering Research Centre for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing 102206, P.R. China
| | - Xiao-Man Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Guang-Yuan Xiao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Ye Su
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Yan Xie
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Yue Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Jian-Fei Pei
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Zhu-Qin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - De-Long Hao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Ping Xu
- State Key Laboratory of Proteomics, National Centre for Protein Sciences Beijing, Beijing Proteome Research Centre, National Engineering Research Centre for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing 102206, P.R. China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China.
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China.
| |
Collapse
|
183
|
Villanueva JE, Livelo C, Trujillo AS, Chandran S, Woodworth B, Andrade L, Le HD, Manor U, Panda S, Melkani GC. Time-restricted feeding restores muscle function in Drosophila models of obesity and circadian-rhythm disruption. Nat Commun 2019; 10:2700. [PMID: 31221967 PMCID: PMC6586848 DOI: 10.1038/s41467-019-10563-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 05/16/2019] [Indexed: 12/19/2022] Open
Abstract
Pathological obesity can result from genetic predisposition, obesogenic diet, and circadian rhythm disruption. Obesity compromises function of muscle, which accounts for a majority of body mass. Behavioral intervention that can counteract obesity arising from genetic, diet or circadian disruption and can improve muscle function holds untapped potential to combat the obesity epidemic. Here we show that Drosophila melanogaster (fruit fly) subject to obesogenic challenges exhibits metabolic disease phenotypes in skeletal muscle; sarcomere disorganization, mitochondrial deformation, upregulation of Phospho-AKT level, aberrant intramuscular lipid infiltration, and insulin resistance. Imposing time-restricted feeding (TRF) paradigm in which flies were fed for 12 h during the day counteracts obesity-induced dysmetabolism and improves muscle performance by suppressing intramuscular fat deposits, Phospho-AKT level, mitochondrial aberrations, and markers of insulin resistance. Importantly, TRF was effective even in an irregular lighting schedule mimicking shiftwork. Hence, TRF is an effective dietary intervention for combating metabolic dysfunction arising from multiple causes.
Collapse
Affiliation(s)
- Jesús E Villanueva
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Christopher Livelo
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Adriana S Trujillo
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Sahaana Chandran
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Brendon Woodworth
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Leo Andrade
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Hiep D Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Girish C Melkani
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA.
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
184
|
Chaix A, Manoogian ENC, Melkani GC, Panda S. Time-Restricted Eating to Prevent and Manage Chronic Metabolic Diseases. Annu Rev Nutr 2019; 39:291-315. [PMID: 31180809 DOI: 10.1146/annurev-nutr-082018-124320] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Molecular clocks are present in almost every cell to anticipate daily recurring and predictable changes, such as rhythmic nutrient availability, and to adapt cellular functions accordingly. At the same time, nutrient-sensing pathways can respond to acute nutrient imbalance and modulate and orient metabolism so cells can adapt optimally to a declining or increasing availability of nutrients. Organismal circadian rhythms are coordinated by behavioral rhythms such as activity-rest and feeding-fasting cycles to temporally orchestrate a sequence of physiological processes to optimize metabolism. Basic research in circadian rhythms has largely focused on the functioning of the self-sustaining molecular circadian oscillator, while research in nutrition science has yielded insights into physiological responses to caloric deprivation or to specific macronutrients. Integration of these two fields into actionable new concepts in the timing of food intake has led to the emerging practice of time-restricted eating. In this paradigm, daily caloric intake is restricted to a consistent window of 8-12 h. This paradigm has pervasive benefits on multiple organ systems.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Emily N C Manoogian
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Girish C Melkani
- Molecular Biology Program and Heart Institute, Department of Biology, San Diego State University, San Diego, California 92182, USA
| | - Satchidananda Panda
- Regulatory Biology Lab, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| |
Collapse
|
185
|
Jamshed H, Beyl RA, Della Manna DL, Yang ES, Ravussin E, Peterson CM. Early Time-Restricted Feeding Improves 24-Hour Glucose Levels and Affects Markers of the Circadian Clock, Aging, and Autophagy in Humans. Nutrients 2019; 11:E1234. [PMID: 31151228 PMCID: PMC6627766 DOI: 10.3390/nu11061234] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Time-restricted feeding (TRF) is a form of intermittent fasting that involves having a longer daily fasting period. Preliminary studies report that TRF improves cardiometabolic health in rodents and humans. Here, we performed the first study to determine how TRF affects gene expression, circulating hormones, and diurnal patterns in cardiometabolic risk factors in humans. Eleven overweight adults participated in a 4-day randomized crossover study where they ate between 8 am and 2 pm (early TRF (eTRF)) and between 8 am and 8 pm (control schedule). Participants underwent continuous glucose monitoring, and blood was drawn to assess cardiometabolic risk factors, hormones, and gene expression in whole blood cells. Relative to the control schedule, eTRF decreased mean 24-hour glucose levels by 4 ± 1 mg/dl (p = 0.0003) and glycemic excursions by 12 ± 3 mg/dl (p = 0.001). In the morning before breakfast, eTRF increased ketones, cholesterol, and the expression of the stress response and aging gene SIRT1 and the autophagy gene LC3A (all p < 0.04), while in the evening, it tended to increase brain-derived neurotropic factor (BNDF; p = 0.10) and also increased the expression of MTOR (p = 0.007), a major nutrient-sensing protein that regulates cell growth. eTRF also altered the diurnal patterns in cortisol and the expression of several circadian clock genes (p < 0.05). eTRF improves 24-hour glucose levels, alters lipid metabolism and circadian clock gene expression, and may also increase autophagy and have anti-aging effects in humans.
Collapse
Affiliation(s)
- Humaira Jamshed
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Robbie A Beyl
- Biostatistics and Analysis Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Deborah L Della Manna
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Eric Ravussin
- Translational Physiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Courtney M Peterson
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
186
|
Bland JS. Fasting Physiology and Therapeutic Diets: A Look Back to the Future. Integr Med (Encinitas) 2019; 18:16-21. [PMID: 31341428 PMCID: PMC6601432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The evidence presented at this event demonstrated the multiple clinical benefits of fasting physiology and points toward a future in which the clinical applications of dietary approaches will be well understood and successfully utilized. The conference reflected the scope and breadth of current research efforts in this important clinical area. Clearly, the application of the important new concepts related to fasting physiology that are emerging will require the advocacy and participation of professionals who are well trained in the fields of clinical nutrition and personalized lifestyle medicine.
Collapse
|
187
|
Zhou L, Kang L, Xiao X, Jia L, Zhang Q, Deng M. "Gut Microbiota-Circadian Clock Axis" in Deciphering the Mechanism Linking Early-Life Nutritional Environment and Abnormal Glucose Metabolism. Int J Endocrinol 2019; 2019:5893028. [PMID: 31534453 PMCID: PMC6732598 DOI: 10.1155/2019/5893028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
The prevalence of diabetes mellitus (DM) has been increasing dramatically worldwide, but the pathogenesis is still unknown. A growing amount of evidence suggests that an abnormal developmental environment in early life increases the risk of developing metabolic diseases in adult life, which is referred to as the "metabolic memory" and the Developmental Origins of Health and Disease (DOHaD) hypothesis. The mechanism of "metabolic memory" has become a hot topic in the field of DM worldwide and could be a key to understanding the pathogenesis of DM. In recent years, several large cohort studies have shown that shift workers have a higher risk of developing type 2 diabetes mellitus (T2DM) and worse control of blood glucose levels. Furthermore, a maternal high-fat diet could lead to metabolic disorders and abnormal expression of clock genes and clock-controlled genes in offspring. Thus, disorders of circadian rhythm might play a pivotal role in glucose metabolic disturbances, especially in terms of early adverse nutritional environments and the development of metabolic diseases in later life. In addition, as a peripheral clock, the gut microbiota has its own circadian rhythm that fluctuates with periodic feeding and has been widely recognized for its significant role in metabolism. In light of the important roles of the gut microbiota and circadian clock in metabolic health and their interconnected regulatory relationship, we propose that the "gut microbiota-circadian clock axis" might be a novel and crucial mechanism to decipher "metabolic memory." The "gut microbiota-circadian clock axis" is expected to facilitate the future development of a novel target for the prevention and intervention of diabetes during the early stage of life.
Collapse
Affiliation(s)
- Liyuan Zhou
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lin Kang
- Department of Endocrinology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lijing Jia
- Department of Endocrinology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingqun Deng
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
188
|
Yabluchanskiy A, Ungvari Z, Csiszar A, Tarantini S. Advances and challenges in geroscience research: An update. Physiol Int 2018; 105:298-308. [PMID: 30587027 PMCID: PMC9341286 DOI: 10.1556/2060.105.2018.4.32] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Aging remains the most pervasive risk factor for a wide range of chronic diseases that afflict modern societies. In the United States alone, incidence of age-related diseases (e.g., cardiovascular disease, stroke, Alzheimer's disease, vascular cognitive impairment and dementia, cancer, hypertension, type-2 diabetes, chronic obstructive pulmonary disease, and osteoarthritis) is on the rise, posing an unsustainable socioeconomic burden even for the most developed countries. Tackling each and every age-related disease alone is proving to be costly and ineffective. The emerging field of geroscience has posed itself as an interdisciplinary approach that aims to understand the relationship between the biology of aging and the pathophysiology of chronic age-related diseases. According to the geroscience concept, aging is the single major risk factor that underlies several age-related chronic diseases, and manipulation of cellular and systemic aging processes can delay the manifestation and/or severity of these age-related chronic pathologies. The goal of this endeavor is to achieve health improvements by preventing/delaying the pathogenesis of several age-related diseases simultaneously in the elderly population by targeting key cellular and molecular processes of aging instead of managing diseases of aging as they arise individually. In this review, we discuss recent advances in the field of geroscience, highlighting their implications for potential future therapeutic targets and the associated scientific challenges and opportunities that lay ahead.
Collapse
Affiliation(s)
- A Yabluchanskiy
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
| | - Z Ungvari
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
- 3 Department of Medical Physics and Informatics, University of Szeged , Szeged, Hungary
- 4 Department of Pulmonology, Semmelweis University , Budapest, Hungary
| | - A Csiszar
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
- 3 Department of Medical Physics and Informatics, University of Szeged , Szeged, Hungary
| | - S Tarantini
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
| |
Collapse
|
189
|
Morris A. Exploring the effect of diet composition in calorie restriction interventions. Nat Rev Endocrinol 2018; 14:625. [PMID: 30258090 DOI: 10.1038/s41574-018-0103-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
190
|
Di Germanio C, Di Francesco A, Bernier M, de Cabo R. Yo-Yo Dieting is Better than None. Obesity (Silver Spring) 2018; 26:1673. [PMID: 30358152 PMCID: PMC6208150 DOI: 10.1002/oby.22335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 09/14/2018] [Indexed: 11/11/2022]
|