1
|
Qi Z, Zhou L, Dai S, Zhang P, Zhong H, Zhou W, Zhao X, Xu H, Zhao G, Wu H, Ge J. Intermittent fasting inhibits platelet activation and thrombosis through the intestinal metabolite indole-3-propionate. LIFE METABOLISM 2025; 4:loaf002. [PMID: 40078933 PMCID: PMC11897983 DOI: 10.1093/lifemeta/loaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 03/14/2025]
Abstract
Platelet hyperreactivity contributes significantly to thrombosis in acute myocardial infarction and stroke. While antiplatelet drugs are used, residual ischemic risk remains. Intermittent fasting (IF), a dietary pattern characterized by alternating periods of eating and fasting, has shown cardiovascular benefits, but its effect on platelet activation is unclear. This study demonstrates that IF inhibits platelet activation and thrombosis in both patients with coronary artery disease and apolipoprotein E (ApoE) knockout (ApoE -/- ) mice, by enhancing intestinal flora production of indole-3-propionic acid (IPA). Mechanistically, elevated IPA in plasma directly attenuates platelet activation by binding to the platelet pregnane X receptor (PXR) and suppressing downstream signaling pathways, including Src/Lyn/Syk and LAT/PLCγ/PKC/Ca2+. Importantly, IF alleviates myocardial and cerebral ischemia/reperfusion injury in ApoE -/- mice. These findings suggest that IF mitigates platelet activation and thrombosis risk in coronary atherosclerosis by enhancing intestinal flora production of IPA, which subsequently activates the platelet PXR-related signaling pathways.
Collapse
Affiliation(s)
- Zhiyong Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Luning Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Shimo Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Wenxuan Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Huajie Xu
- Department of Infectious Disease, Zhongshan Hospital, Fudan University, 180 Fenglin Road , Shanghai 200032, China
| | - Gang Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, 131 Dong’an Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
2
|
Ma RX. A detective story of intermittent fasting effect on immunity. Immunology 2024; 173:227-247. [PMID: 38922825 DOI: 10.1111/imm.13829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Intermittent fasting (IF) refers to periodic fasting routines, that caloric intake is minimized not by meal portion size reduction but by intermittently eliminating ingestion of one or several consecutive meals. IF can instigate comprehensive and multifaceted alterations in energy metabolism, these metabolic channels may aboundingly function as primordial mechanisms that interface with the immune system, instigating intricate immune transformations. This review delivers a comprehensive understanding of IF, paying particular attention to its influence on the immune system, thus seeking to bridge these two research domains. We explore how IF effects lipid metabolism, hormonal levels, circadian rhythm, autophagy, oxidative stress, gut microbiota, and intestinal barrier integrity, and conjecture about the mechanisms orchestrating the intersect between these factors and the immune system. Moreover, the review includes research findings on the implications of IF on the immune system and patients burdened with autoimmune diseases.
Collapse
Affiliation(s)
- Ru-Xue Ma
- School of Medical, Qinghai University, Xining, China
| |
Collapse
|
3
|
Ozcan M, Abdellatif M, Javaheri A, Sedej S. Risks and Benefits of Intermittent Fasting for the Aging Cardiovascular System. Can J Cardiol 2024; 40:1445-1457. [PMID: 38354947 DOI: 10.1016/j.cjca.2024.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
Population aging and the associated increase in cardiovascular disease rates pose serious threats to global public health. Different forms of fasting have become an increasingly attractive strategy to directly address aging and potentially limit or delay the onset of cardiovascular diseases. A growing number of experimental studies and clinical trials indicate that the amount and timing of food intake as well as the daily time window during which food is consumed, are crucial determinants of cardiovascular health. Indeed, intermittent fasting counteracts the molecular hallmarks of cardiovascular aging and promotes different aspects of cardiometabolic health, including blood pressure and glycemic control, as well as body weight reduction. In this report, we summarize current evidence from randomized clinical trials of intermittent fasting on body weight and composition as well as cardiovascular and metabolic risk factors. Moreover, we critically discuss the preventive and therapeutic potential of intermittent fasting, but also possible detrimental effects in the context of cardiovascular aging and related disease. We delve into the physiological mechanisms through which intermittent fasting might improve cardiovascular health, and raise important factors to consider in the design of clinical trials on the efficacy of intermittent fasting to reduce major adverse cardiovascular events among aged individuals at high risk of cardiovascular disease. We conclude that despite growing evidence and interest among the lay and scientific communities in the cardiovascular health-improving effects of intermittent fasting, further research efforts and appropriate caution are warranted before broadly implementing intermittent fasting regimens, especially in elderly persons.
Collapse
Affiliation(s)
- Mualla Ozcan
- Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Ali Javaheri
- Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA; John J. Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria; Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia.
| |
Collapse
|
4
|
Gauvin DV, McComb M, Farero R. A Commentary on Fasting of Nonclinical Research Animals. Int J Toxicol 2024; 43:196-208. [PMID: 38151260 DOI: 10.1177/10915818231218975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
This commentary discusses the implementation of fasting in nonclinical animal experimental subjects. The short-term removal of food from cages of experimental animals is in all respects innocuous. The term "stress" is ill-defined and the statutes and regulations governing animal research laboratories that exert their authority in the performance of their operations do so without substantive grounds to base compliance. The legislative and administrative history of the implementation of the Animal Welfare Act (AWA) has evolved into the development of laboratory management strategies that focus on the reduction of the biological cost of stress to the animals and the determination of when subclinical stress (eustress) becomes distress. Animal welfare is based on the tenet that in laboratories conducting animal research in compliance with Good Laboratory Practices (Title 21 USC, Chapter 13,§58), it is the study protocol and the study director that establish procedures and processes that are approved by each Institutional Animal Care and Use Committee to ensure the humane care and use of animals in research, teaching, and testing and to ensure compliance with guidelines and regulations. This approval process establishes the justification of eustress in the environment that do not rise to the threshold of distress under the AWA.
Collapse
Affiliation(s)
- David V Gauvin
- Department of Neurobehavioral Sciences, Charles River Laboratories - MWN, Mattawan, MI, USA
| | - Margaret McComb
- Department of Neurobehavioral Sciences, Charles River Laboratories - MWN, Mattawan, MI, USA
| | - Ryan Farero
- Department of Neurobehavioral Sciences, Charles River Laboratories - MWN, Mattawan, MI, USA
| |
Collapse
|
5
|
Habiby M, Ezati P, Soltanian D, Rahehagh R, Hosseini F. Comparison of three methods of intermittent fasting in high-fat-diet-induced obese mice. Heliyon 2024; 10:e25708. [PMID: 38390147 PMCID: PMC10881537 DOI: 10.1016/j.heliyon.2024.e25708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
Purpose Intermittent fasting (IF), describes a variety of diets that the individual is exposed to intermittent periods of fasting and refeeding. The present study was designed to compare the three most popular intermittent fasting methods in high-fat-diet-induced obese mice. Methods 50 male C57BL/6 mice were divided into Normal Control (CN), High-Fat Control (CH), IF1 (Alternate Day Fasting), IF2 (Time-Restricted Feeding), IF3 (5:2 Diet) diets. In the first phase of the study, the mice were fed ad libitum either with a 54% Calorie high-fat (CH, IF1, IF2, IF3) or standard CHOW (CN) for 8 weeks. Then, in the intervention phase, the IF groups were fasted for four weeks based on their fasting protocol. At the end of the study, the mice fasting blood, liver and fat tissue samples were biochemically and pathologically assessed. Results The weight loss during the fasting period in IF1 and IF2 groups was significantly greater than CH. The epididymal fat pad weight was significantly lower in IF2 and IF3 compared to CH. The serum Triglyceride was significantly greater in CH than in the CN group. The tissue injury scores of the high-fat-diet groups were significantly greater than CN. Also, the tissue injury score was greater in IF1 group compared to the high-fat control group. Conclusion All of the fasting protocols can prompt acceptable energy restriction and the ADF and TRF protocols can significantly cause weight loss. Also, the TRF and 5:2 Diets can lower the visceral adiposity. However further human studies focusing on dietary adherence seem necessary.
Collapse
Affiliation(s)
- Mehrdad Habiby
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Pourya Ezati
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Danial Soltanian
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ramesh Rahehagh
- Department of Pathology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Foruzan Hosseini
- Department of Medical Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
6
|
Paoli A, Tinsley GM, Mattson MP, De Vivo I, Dhawan R, Moro T. Common and divergent molecular mechanisms of fasting and ketogenic diets. Trends Endocrinol Metab 2024; 35:125-141. [PMID: 38577754 DOI: 10.1016/j.tem.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 04/06/2024]
Abstract
Intermittent short-term fasting (ISTF) and ketogenic diets (KDs) exert overlapping but not identical effects on cell metabolism, function, and resilience. Whereas health benefits of KD are largely mediated by the ketone bodies (KBs), ISTF engages additional adaptive physiological responses. KDs act mainly through inhibition of histone deacetylases (HDACs), reduction of oxidative stress, improvement of mitochondria efficiency, and control of inflammation. Mechanisms of action of ISTF include stimulation of autophagy, increased insulin and leptin sensitivity, activation of AMP-activated protein kinase (AMPK), inhibition of the mechanistic target of rapamycin (mTOR) pathway, bolstering mitochondrial resilience, and suppression of oxidative stress and inflammation. Frequent switching between ketogenic and nonketogenic states may optimize health by increasing stress resistance, while also enhancing cell plasticity and functionality.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35127 Padua, Italy.
| | - Grant M Tinsley
- Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX 79409, USA
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ravi Dhawan
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padua, 35127 Padua, Italy
| |
Collapse
|
7
|
Schroor MM, Joris PJ, Plat J, Mensink RP. Effects of Intermittent Energy Restriction Compared with Those of Continuous Energy Restriction on Body Composition and Cardiometabolic Risk Markers - A Systematic Review and Meta-Analysis of Randomized Controlled Trials in Adults. Adv Nutr 2024; 15:100130. [PMID: 37827491 PMCID: PMC10831889 DOI: 10.1016/j.advnut.2023.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/21/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023] Open
Abstract
The interest in intermittent energy restriction (IER) diets as a weight-loss approach is increasing. Different IER protocols exist, including time-restricted eating (TRE), alternate-day fasting (ADF), and the 5:2 diet. This meta-analysis compared the effects of these IER diets with continuous energy restriction (CER) on anthropometrics and cardiometabolic risk markers in healthy adults. Twenty-eight trials were identified that studied TRE (k = 7), ADF (k = 10), or the 5:2 diet (k = 11) for 2-52 wk. Energy intakes between intervention groups within a study were comparable (17 trials), lower in IER (5 trials), or not reported (6 trials). Weighted mean differences (WMDs) were calculated using fixed- or random-effects models. Changes in body weight [WMD: -0.42 kg; 95% confidence interval (CI): -0.96 to 0.13; P = 0.132] and fat mass (FM) (WMD: -0.31 kg; 95% CI: -0.98 to 0.36; P = 0.362) were comparable when results of the 3 IER diets were combined and compared with those of CER. All IER diets combined reduced fat-free mass (WMD: -0.20 kg; 95% CI: -0.39 to -0.01; P = 0.044) and waist circumference (WMD: -0.91 cm; 95% CI: -1.76 to -0.06; P = 0.036) more than CER. Effects on body mass index [BMI (kg/m2)], glucose, insulin, homeostatic model assessment for insulin resistance (HOMA-IR), serum lipid and lipoprotein concentrations, and blood pressure did not differ. Further, TRE reduced body weight, FM, and fat-free mass more than CER, whereas ADF improved HOMA-IR more. BMI was reduced less in the 5:2 diet compared with CER. In conclusion, the 3 IER diets combined did not lead to superior improvements in anthropometrics and cardiometabolic risk markers compared with CER diets. Slightly greater reductions were, however, observed in fat-free mass and waist circumference. To what extent differences in energy intakes between groups within studies may have influenced these outcomes should be addressed in future studies.
Collapse
Affiliation(s)
- Maite M Schroor
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.
| | - Peter J Joris
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Ronald P Mensink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
8
|
Arumugam TV, Alli-Shaik A, Liehn EA, Selvaraji S, Poh L, Rajeev V, Cho Y, Cho Y, Kim J, Kim J, Swa HLF, Hao DTZ, Rattanasopa C, Fann DYW, Mayan DC, Ng GYQ, Baik SH, Mallilankaraman K, Gelderblom M, Drummond GR, Sobey CG, Kennedy BK, Singaraja RR, Mattson MP, Jo DG, Gunaratne J. Multiomics analyses reveal dynamic bioenergetic pathways and functional remodeling of the heart during intermittent fasting. eLife 2023; 12:RP89214. [PMID: 37769126 PMCID: PMC10538958 DOI: 10.7554/elife.89214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
Intermittent fasting (IF) has been shown to reduce cardiovascular risk factors in both animals and humans, and can protect the heart against ischemic injury in models of myocardial infarction. However, the underlying molecular mechanisms behind these effects remain unclear. To shed light on the molecular and cellular adaptations of the heart to IF, we conducted comprehensive system-wide analyses of the proteome, phosphoproteome, and transcriptome, followed by functional analysis. Using advanced mass spectrometry, we profiled the proteome and phosphoproteome of heart tissues obtained from mice that were maintained on daily 12- or 16 hr fasting, every-other-day fasting, or ad libitum control feeding regimens for 6 months. We also performed RNA sequencing to evaluate whether the observed molecular responses to IF occur at the transcriptional or post-transcriptional levels. Our analyses revealed that IF significantly affected pathways that regulate cyclic GMP signaling, lipid and amino acid metabolism, cell adhesion, cell death, and inflammation. Furthermore, we found that the impact of IF on different metabolic processes varied depending on the length of the fasting regimen. Short IF regimens showed a higher correlation of pathway alteration, while longer IF regimens had an inverse correlation of metabolic processes such as fatty acid oxidation and immune processes. Additionally, functional echocardiographic analyses demonstrated that IF enhances stress-induced cardiac performance. Our systematic multi-omics study provides a molecular framework for understanding how IF impacts the heart's function and its vulnerability to injury and disease.
Collapse
Affiliation(s)
- Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe UniversityMelbourneAustralia
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Asfa Alli-Shaik
- Translational Biomedical Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Elisa A Liehn
- National Heart Research Institute, National Heart Centre SingaporeSingaporeSingapore
- Institute for Molecular Medicine, University of Southern DenmarkOdenseDenmark
- National Institute of Pathology "Victor Babes"BucharestRomania
| | - Sharmelee Selvaraji
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of SingaporeSingaporeSingapore
| | - Luting Poh
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Vismitha Rajeev
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Yongeun Cho
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Jongho Kim
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Joonki Kim
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- Natural Products Research Center, Korea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Hannah LF Swa
- Translational Biomedical Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - David Tan Zhi Hao
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Chutima Rattanasopa
- Translational Laboratories in Genetic Medicine, Agency for Science, Technology and ResearchSingaporeSingapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of SingaporeSingaporeSingapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - David Castano Mayan
- Translational Laboratories in Genetic Medicine, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Sang-Ha Baik
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Karthik Mallilankaraman
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe UniversityMelbourneAustralia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe UniversityMelbourneAustralia
| | - Brian K Kennedy
- Department of Physiology, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
- Department of Biochemistry, Yong Loo Lin School Medicine, National University of SingaporeSingaporeSingapore
| | - Roshni R Singaraja
- Department of Medicine, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Jayantha Gunaratne
- Translational Biomedical Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| |
Collapse
|
9
|
Staffeld A, Gill S, Zimmermann A, Böge N, Schuster K, Lang S, Kipp M, Palme R, Frintrop L. Establishment of a Murine Chronic Anorexia Nervosa Model. Cells 2023; 12:1710. [PMID: 37443744 PMCID: PMC10340390 DOI: 10.3390/cells12131710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Anorexia nervosa (AN) is associated with hyperactivity, amenorrhea, and brain atrophy. The underlying pathophysiology is mostly unknown, and new targets for therapeutic interventions are needed. This study aimed to systematically establish a murine AN model with the parameter extent of starvation, animal age, and length of starvation for functional studies. The activity-based anorexia (ABA) model combines food restriction with running wheel access. Early adolescent and adolescent mice received 40% of their baseline food intake until a 20% or 25% weight reduction was reached (acute starvation). To mimic chronic starvation, body weight loss was maintained for another two weeks. Running activity was examined using wheel sensors, while amenorrhea was investigated by analysis of vaginal smears. Brain sections were used to analyze cerebral cortex volumes. Acute starvation did not lead to either AN-related symptoms, whereas chronic starvation led to hyperactivity and amenorrhea except in the adolescent cohort with 20% weight reduction. Only ABA mice with 25% weight reduction revealed a cortex volume reduction. The optimal parameters to mirror AN-related symptoms included a 25% weight reduction, early adolescent or adolescent mice, and chronic starvation. The ABA model enables functional analysis of the impact of chronic AN on the underlying hormonal, behavioral, and brain pathophysiology.
Collapse
Affiliation(s)
- Anna Staffeld
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Sadaf Gill
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Annelie Zimmermann
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Natalie Böge
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Katharina Schuster
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Stephan Lang
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Rupert Palme
- Unit of Physiology, Pathophysiology and Experimental Endocrinology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria
| | - Linda Frintrop
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
10
|
Lü W, Yu T, Kuang W. Effects of dietary restriction on cognitive function: a systematic review and meta-analysis. Nutr Neurosci 2023; 26:540-550. [PMID: 35469542 DOI: 10.1080/1028415x.2022.2068876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Potential positive link between cognitive function and dietary restriction has been widely concerned. The present study describes a systematic review and preliminary meta-analysis to assess the efficacy of dietary restriction on cognitive function. We classified dietary restrictions as Calories Restriction (CR) and Intermittent Fasting (IF). METHOD This systematic review and meta-analysis conducted following the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Statement (PRISMA) Checklist, Databases including PubMed, Embase, Web of Science, and the Cochrane Library were searched for randomized controlled trials (RCTs) assessing the cognition effectiveness of dietary restriction from database inception to Sep 2021. RESULT Eleven RCTs met the inclusion criteria in the systematic review and meta-analysis. The overall effect of dietary restriction on cognitive function is SMD = 0.22 (95% CI: 0.09-0.34, p < 0.01). The effect on global function SMD = 0.35 (95% CI: 0.04-0.65, p < 0.05) and memory SMD = 0.18 (95% CI: 0.00-0.35, p = 0.05) is significant. MCI showed the best effectiveness SMD = 0.36 (95% CI: 0.05-0.68, p < 0.05), followed by the normal weight population SMD = 0.28 (95% CI: 0.03-0.52, p < 0.05) and overweight population SMD = 0.20 (95% CI: 0.06-0.34, p < 0.01). No statistically significant difference showed between IF and CR (p > 0.05). CONCLUSION Our study demonstrated that dietary restriction has varying degrees of positive effect on cognitive function in overweight/normal-weight people and MCI. However, it should be cautious when generalizing to other populations. Additional high-quality, large-scale, cohort and intervention studies are needed to further assess the effectiveness of dietary restriction on cognition.
Collapse
Affiliation(s)
- Wenqi Lü
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tingting Yu
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Weihong Kuang
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
11
|
Yin W, Sun L, Liang Y, Luo C, Feng T, Zhang Y, Zhang W, Yin Y. Maternal intermittent fasting deteriorates offspring metabolism via suppression of hepatic mTORC1 signaling. FASEB J 2023; 37:e22831. [PMID: 36856728 DOI: 10.1096/fj.202201907r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 03/02/2023]
Abstract
The metabolic benefits of intermittent fasting (IF) have been well recognized. However, limited studies have examined the relationship between long-term maternal IF before pregnancy and offspring health. In this study, a C57BL/6J mouse model of long-term IF before pregnancy was established: 4-week-old female mice were subjected to alternate-day fasting for 12 weeks and resumed normal diet after mating. Female mice in the control group were fed ad libitum. Offspring mice were weaned at 6 weeks of age and fed a normal chow diet or a 60% high-fat diet. The effects of long-term pre-pregnancy IF on offspring metabolism and its underlying mechanism were examined. We found that neonatal IF offspring weighted significantly less relevant to control mice. This difference gradually disappeared as a result of catch-up growth. In the IF offspring, adipose tissue mass was significantly increased. This alteration was associated with a considerable deterioration in glucose tolerance. No significant difference in food intake was observed. Further, lipid deposition as well as triglyceride contents in the liver were greatly increased. Maternal IF significantly decreased levels of DNA methyltransferase in the liver of offspring. DNA methylation modifications of molecules associated with the mTORC1 signaling pathway were significantly altered, leading to the significant inhibition of mTORC1 signaling. Overexpression of S6K1 activated hepatic mTORC1 signaling and reversed the metabolic dysfunction in IF offspring. In conclusion, long-term pre-pregnancy IF increases hepatic steatosis and adiposity, as well as impairs glucose metabolism in adult offspring. This occurs through DNA methylation-dependent suppression of hepatic mTORC1 signaling activity.
Collapse
Affiliation(s)
- Wenzhen Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China.,Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yuan Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Chao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Tiange Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yunhua Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China.,Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| |
Collapse
|
12
|
Ambroselli D, Masciulli F, Romano E, Catanzaro G, Besharat ZM, Massari MC, Ferretti E, Migliaccio S, Izzo L, Ritieni A, Grosso M, Formichi C, Dotta F, Frigerio F, Barbiera E, Giusti AM, Ingallina C, Mannina L. New Advances in Metabolic Syndrome, from Prevention to Treatment: The Role of Diet and Food. Nutrients 2023; 15:640. [PMID: 36771347 PMCID: PMC9921449 DOI: 10.3390/nu15030640] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The definition of metabolic syndrome (MetS) has undergone several changes over the years due to the difficulty in establishing universal criteria for it. Underlying the disorders related to MetS is almost invariably a pro-inflammatory state related to altered glucose metabolism, which could lead to elevated cardiovascular risk. Indeed, the complications closely related to MetS are cardiovascular diseases (CVDs) and type 2 diabetes (T2D). It has been observed that the predisposition to metabolic syndrome is modulated by complex interactions between human microbiota, genetic factors, and diet. This review provides a summary of the last decade of literature related to three principal aspects of MetS: (i) the syndrome's definition and classification, pathophysiology, and treatment approaches; (ii) prediction and diagnosis underlying the biomarkers identified by means of advanced methodologies (NMR, LC/GC-MS, and LC, LC-MS); and (iii) the role of foods and food components in prevention and/or treatment of MetS, demonstrating a possible role of specific foods intake in the development of MetS.
Collapse
Affiliation(s)
- Donatella Ambroselli
- Laboratory of Food Chemistry, Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, 00185 Rome, Italy
| | - Fabrizio Masciulli
- Laboratory of Food Chemistry, Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, 00185 Rome, Italy
| | - Enrico Romano
- Laboratory of Food Chemistry, Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, 00185 Rome, Italy
| | - Giuseppina Catanzaro
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | | | - Maria Chiara Massari
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Silvia Migliaccio
- Department of Movement, Human and Health Sciences, Health Sciences Section, University “Foro Italico”, 00135 Rome, Italy
| | - Luana Izzo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Alberto Ritieni
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
- UNESCO, Health Education and Sustainable Development, University of Naples Federico II, 80131 Naples, Italy
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Caterina Formichi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Francesco Frigerio
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Eleonora Barbiera
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Anna Maria Giusti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Cinzia Ingallina
- Laboratory of Food Chemistry, Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, 00185 Rome, Italy
| | - Luisa Mannina
- Laboratory of Food Chemistry, Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
13
|
Forte M, Rodolico D, Ameri P, Catalucci D, Chimenti C, Crotti L, Schirone L, Pingitore A, Torella D, Iacovone G, Valenti V, Schiattarella GG, Perrino C, Sciarretta S. Molecular mechanisms underlying the beneficial effects of exercise and dietary interventions in the prevention of cardiometabolic diseases. J Cardiovasc Med (Hagerstown) 2022; 24:e3-e14. [PMID: 36729582 DOI: 10.2459/jcm.0000000000001397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cardiometabolic diseases still represent a major cause of mortality worldwide. In addition to pharmacological approaches, lifestyle interventions can also be adopted for the prevention of these morbid conditions. Lifestyle changes include exercise and dietary restriction protocols, such as calorie restriction and intermittent fasting, which were shown to delay cardiovascular ageing and elicit health-promoting effects in preclinical models of cardiometabolic diseases. Beneficial effects are mediated by the restoration of multiple molecular mechanisms in heart and vessels that are compromised by metabolic stress. Exercise and dietary restriction rescue mitochondrial dysfunction, oxidative stress and inflammation. They also improve autophagy. The result of these effects is a marked improvement of vascular and heart function. In this review, we provide a comprehensive overview of the molecular mechanisms involved in the beneficial effects of exercise and dietary restriction in models of diabetes and obesity. We also discuss clinical studies and gap in animal-to-human translation.
Collapse
Affiliation(s)
- Maurizio Forte
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli
| | - Daniele Rodolico
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico.,Department of Internal Medicine, University of Genova, Genova
| | - Daniele Catalucci
- Humanitas Research Hospital, IRCCS, Rozzano.,National Research Council, Institute of Genetic and Biomedical Research - UOS, Milan
| | - Cristina Chimenti
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital.,Department of Medicine and Surgery, Università Milano-Bicocca, Milan
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina
| | - Annachiara Pingitore
- Department of General and Specialistic Surgery 'Paride Stefanini' Sapienza University of Rome
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro
| | | | | | - Gabriele G Schiattarella
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli.,Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina
| | | |
Collapse
|
14
|
Zhang W, Wang J, Wang L, Shi R, Chu C, Shi Z, Liu P, Li Y, Liu X, Liu Z. Alternate-day fasting prevents non-alcoholic fatty liver disease and working memory impairment in diet-induced obese mice. J Nutr Biochem 2022; 110:109146. [PMID: 36049672 DOI: 10.1016/j.jnutbio.2022.109146] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 04/28/2022] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Alternate-day fasting (ADF) regimen has been reported to alleviate obesity and insulin resistance. However, the effects of ADF on preventing diet-induced non-alcoholic fatty liver disease (NAFLD) and related cognitive deficits are still elusive. In the present study, a high-fat diet (HFD)-induced obese (DIO) C57BL/6 mouse model was established. Mice were treated with a 4-week long ADF regimen and/or switching the diet to a standard diet. ADF reduced lipid accumulation, activated AMPK/ULK1 signaling, and suppressed the phosphorylation of mTOR. Also, ADF inhibited lipid accumulation and inflammatory responses in the white adipose tissue and down-regulated expressions of PPAR-γ and cytokines. Moreover, ADF improved the working memory and synaptic structure in the DIO mice and upregulated PSD-95 and BDNF in the hippocampus. ADF reduced oxidative stress and microglial over-activation in the CNS. These results suggest that ADF attenuates NAFLD development in the liver of DIO mice, which is related to the mediating effects of ADF on autophagy and energy metabolism. ADF also enhanced cognitive function, which could be partly explained by the down-regulated peripheral inflammatory responses. This study indicates that ADF could be a promising intervention for alleviating NAFLD development and cognitive decline.
Collapse
Affiliation(s)
- Wentong Zhang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jun Wang
- Department of Digestive Diseases, Xijing Hospital, Xi'an, Shaanxi Province, China
| | - Luanfeng Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Renjie Shi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Chuanqi Chu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Zhiling Shi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Pujie Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yitong Li
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China; Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
15
|
Naz H, Haider R, Rashid H, Ul Haq Z, Malik J, Zaidi SMJ, Ishaq U, Trevisan R. Islamic fasting: cardiovascular disease perspective. Expert Rev Cardiovasc Ther 2022; 20:795-805. [PMID: 36260858 DOI: 10.1080/14779072.2022.2138344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
INTRODUCTION Ramadan is a month of obligatory fasting observed by the majority of 2 billion Muslims living around the globe. The guidelines for 'risk-free' fasting exist for chronic diseases, including diabetes mellitus, but recommendations for cardiovascular disease (CVD) patients are deficient due to the paucity of literature. AREAS COVERED Databases were screened to find relevant studies for an evidence-based consensus regarding the risk stratification and management of CVD. Using practical guidelines of the European Society of Cardiology (ESC), we categorized patients into low-, moderate-, and high-risk categories and proposed a pre-Ramadan checklist for the assessment of cardiac patients before fasting. Regular moderate-intensity exercise is recommended for most cardiac patients, which has been demonstrated to provide an anti-inflammatory and antioxidant effect that improves immune function. EXPERT OPINION In Ramadan, many physiological changes occur during fasting, which brings about a balanced metabolic homeostasis of the body. In addition, Ramadan fasting is a nonpharmacologic means of decreasing CV risk factors. As Islam exempts Muslims from fasting if they are unwell; therefore, patients with the acute coronary syndrome (ACS), advanced heart failure (HF), recent percutaneous coronary intervention (PCI), or cardiac surgery should avoid fasting.
Collapse
Affiliation(s)
- Hifza Naz
- Medical Student, School of Medicine and Surgery, University of Milan Bicocca, Bergamo, Italy
| | - Rakhshan Haider
- Department of Medicine, Shifa International Hospital, Islamabad, Pakistan
| | - Haroon Rashid
- Department of Intensive care, Wythenshawe Hospital, Manchester, UK
| | | | - Jahanzeb Malik
- Department of Cardiology, Rawalpindi Institute of Cardiology, Rawalpindi, Pakistan
| | | | - Uzma Ishaq
- Department of Hematology, Healthways Laboratories, Rawalpindi, Pakistan
| | - Roberto Trevisan
- Department of Endocrinology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
16
|
Ratiner K, Shapiro H, Goldenberg K, Elinav E. Time-limited diets and the gut microbiota in cardiometabolic disease. J Diabetes 2022; 14:377-393. [PMID: 35698246 PMCID: PMC9366560 DOI: 10.1111/1753-0407.13288] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, intermittent fasting (IF), including periodic fasting and time-restricted feeding (TRF), has been increasingly suggested to constitute a promising treatment for cardiometabolic diseases (CMD). A deliberate daily pause in food consumption influences the gut microbiome and the host circadian clock, resulting in improved cardiometabolic health. Understanding the molecular mechanisms by which circadian host-microbiome interactions affect host metabolism and immunity may add a potentially important dimension to effective implementation of IF diets. In this review, we discuss emerging evidence potentially linking compositional and functional alterations of the gut microbiome with IF impacts on mammalian metabolism and risk of development of hypertension, type 2 diabetes (T2D), obesity, and their long-term micro- and macrovascular complications. We highlight the challenges and unknowns in causally linking diurnal bacterial signals with dietary cues and downstream metabolic consequences and means of harnessing these signals toward future microbiome integration into precision medicine.
Collapse
Affiliation(s)
- Karina Ratiner
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Hagit Shapiro
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Kim Goldenberg
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Eran Elinav
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
- Microbiome & Cancer Division, DKFZHeidelbergGermany
| |
Collapse
|
17
|
Ali Q, Ma S, La S, Guo Z, Liu B, Gao Z, Farooq U, Wang Z, Zhu X, Cui Y, Li D, Shi Y. Microbial short-chain fatty acids: a bridge between dietary fibers and poultry gut health. Anim Biosci 2022; 35:1461-1478. [PMID: 35507857 PMCID: PMC9449382 DOI: 10.5713/ab.21.0562] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/18/2022] [Indexed: 11/27/2022] Open
Abstract
The maintenance of poultry gut health is complex depending on the intricate balance among diet, the commensal microbiota, and the mucosa, including the gut epithelium and the superimposing mucus layer. Changes in microflora composition and abundance can confer beneficial or detrimental effects on fowl. Antibiotics have devastating impacts on altering the landscape of gut microbiota, which further leads to antibiotic resistance or spread the pathogenic populations. By eliciting the landscape of gut microbiota, strategies should be made to break down the regulatory signals of pathogenic bacteria. The optional strategy of conferring dietary fibers (DFs) can be used to counterbalance the gut microbiota. DFs are the non-starch carbohydrates indigestible by host endogenous enzymes but can be fermented by symbiotic microbiota to produce short-chain fatty acids (SCFAs). This is one of the primary modes through which the gut microbiota interacts and communicate with the host. The majority of SCFAs are produced in the large intestine (particularly in the caecum), where they are taken up by the enterocytes or transported through portal vein circulation into the bloodstream. Recent shreds of evidence have elucidated that SCFAs affect the gut and modulate the tissues and organs either by activating G-protein-coupled receptors or affecting epigenetic modifications in the genome through inducing histone acetylase activities and inhibiting histone deacetylases. Thus, in this way, SCFAs vastly influence poultry health by promoting energy regulation, mucosal integrity, immune homeostasis, and immune maturation. In this review article, we will focus on DFs, which directly interact with gut microbes and lead to the production of SCFAs. Further, we will discuss the current molecular mechanisms of how SCFAs are generated, transported, and modulated the pro-and anti-inflammatory immune responses against pathogens and host physiology and gut health.
Collapse
|
18
|
El Agaty SM, Nassef NA, Abou-Bakr DA, Hanafy AA. Chronic activation of cardiac Atg-5 and pancreatic Atg-7 by intermittent fasting alleviates acute myocardial infarction in old rats. Egypt Heart J 2022; 74:31. [PMID: 35416562 PMCID: PMC9008107 DOI: 10.1186/s43044-022-00268-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aging is associated with cardiovascular and metabolic changes, increasing the susceptibility to acute myocardial infarction (AMI). Intermittent fasting (IF) has a beneficial effect on the age-associated cardiovascular diseases. The present study was planned to investigate the possible protective effect of IF against acute AMI induced by isoproterenol (ISO) in old rats and its possible underlying mechanisms mediated by heart and pancreatic autophagy. Thirty Male Wistar rats were divided into four groups: adult; old; Old-ISO (rats subjected to AMI by ISO) and Old-F-ISO groups (rats were subjected to IF for 4 weeks and AMI by ISO). RESULTS IF significantly increased the mRNA expression of cardiac Atg-5 and pancreatic Atg-7 in Old-F-ISO versus old and adult groups. This was associated with a significant decrease in serum troponin-I, serum creatine kinase (CK-MB), cardiac malondialdehyde and cardiac TNF-α, fasting plasma glucose, and HOMA-IR in Old-F-ISO compared to Old-ISO group. Also, IF significantly decreased the age-related overall and visceral obesity in Old-F-ISO versus old and Old-ISO groups. Histological studies revealed attenuation of the local inflammatory response in Old-F-ISO versus Old-ISO group. Pancreatic Atg-7 and heart Atg-5 were significantly increased in Old-ISO versus old rats. CONCLUSIONS IF protects against acute AMI in old rats, possibly, via chronic activation of heart Atg-5 and pancreatic Atg-7, and alleviation of age-related overall and visceral obesity. Thus, IF could be a dietary lifestyle modification for attenuation of the susceptibility to acute AMI in aged population. On the other hand, acute activation of heart and pancreatic autophagy by ISO might augment cardiac injury.
Collapse
Affiliation(s)
- Sahar Mohamed El Agaty
- Department of Physiology, Medical Research Center, Faculty of Medicine, Ain Shams University, 24 Mohamed El Makaref Street, Nasr City, Cairo, Egypt.
| | - Noha A Nassef
- Department of Physiology, Medical Research Center, Faculty of Medicine, Ain Shams University, 24 Mohamed El Makaref Street, Nasr City, Cairo, Egypt
| | - Doaa A Abou-Bakr
- Department of Physiology, Medical Research Center, Faculty of Medicine, Ain Shams University, 24 Mohamed El Makaref Street, Nasr City, Cairo, Egypt
| | - Aya A Hanafy
- Department of Physiology, Medical Research Center, Faculty of Medicine, Ain Shams University, 24 Mohamed El Makaref Street, Nasr City, Cairo, Egypt
| |
Collapse
|
19
|
Dutzmann J, Knöpp K, Kefalianakis Z, Daniel JM, Gufler H, Wohlgemuth W, Kahles F, Sedding DG. Effect of intermittent fasting after ST-elevation myocardial infarction on left ventricular function: study protocol of a pilot randomised controlled trial (INTERFAST-MI). BMJ Open 2022; 12:e050067. [PMID: 35393305 PMCID: PMC8990699 DOI: 10.1136/bmjopen-2021-050067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Preclinical studies consistently show robust disease-modifying effects of intermittent fasting in animal models of cardiovascular disease. However, the impact of intermittent fasting on cardiovascular endpoints after myocardial infarction has not been investigated in a clinical trial so far. METHODS AND ANALYSIS The INTERmittent FASTing after Myocardial Infarction (INTERFAST-MI) trial is a monocentric prospective randomised controlled non-confirmatory pilot study including 48 patients with ST-segment elevation myocardial infarction. They will be randomised in a 1:1 ratio to either intermittent fasting (daily time-restricted eating; consuming food for not more than 8 hours/day, fasting for at least 16 hours/day) or to a control group without a particular diet. The follow-up time is 6 months. The prespecified primary outcome is change in left ventricular systolic function at 4 weeks from baseline to estimate effect size required to establishing sample size and power calculation for a future full-scale trial. Secondary outcomes include protocol adherence, recruitment, major adverse cardiac events, revascularisation, changes in left ventricular systolic function at 3 and 6 months, patient weight, blood pressure, and serum markers of inflammation and cardiovascular disease. Enrolment began on 1 November 2020 and is expected to conclude in December 2021. ETHICS AND DISSEMINATION The trial has received ethics approval from the Medical Ethics Committee of the Martin-Luther-University Halle-Wittenberg. Results of the study will be submitted for publication in a peer-reviewed journal and presented at scientific conferences. TRIAL REGISTRATION NUMBER DRKS00021784.
Collapse
Affiliation(s)
- Jochen Dutzmann
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Kai Knöpp
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Zoe Kefalianakis
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Jan-Marcus Daniel
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Hubert Gufler
- Department of Radiology, University Hospital Halle, Halle, Germany
| | | | - Florian Kahles
- Department of Internal Medicine I, University Hospital Aachen, Aachen, Germany
| | - Daniel G Sedding
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| |
Collapse
|
20
|
The Role of Intermittent Energy Restriction Diet on Metabolic Profile and Weight Loss among Obese Adults. Nutrients 2022; 14:nu14071509. [PMID: 35406122 PMCID: PMC9002823 DOI: 10.3390/nu14071509] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a disease defined by an elevated body mass index (BMI), which is the result of excessive or abnormal accumulation of fat. Dietary intervention is fundamental and essential as the first-line treatment for obese patients, and the main rule of every dietary modification is calorie restriction and consequent weight loss. Intermittent energy restriction (IER) is a special type of diet consisting of intermittent pauses in eating. There are many variations of IER diets such as alternate-day fasting (ADF) and time-restricted feeding (TRF). In the literature, the IER diet is known as an effective method for bodyweight reduction. Furthermore, IER diets have a beneficial effect on systolic or diastolic pressure, lipid profile, and glucose homeostasis. In addition, IER diets are presented as being as efficient as a continuous energy restriction diet (CER) in losing weight and improving metabolic parameters. Thus, the IER diet could present an alternative option for those who cannot accept a constant food regimen.
Collapse
|
21
|
He J, Tuo W, Zhang X, Dai Y, Fang M, Zhou T, Xiu M, Liu Y. Olfactory Senses Modulate Food Consumption and Physiology in Drosophila melanogaster. Front Behav Neurosci 2022; 16:788633. [PMID: 35431829 PMCID: PMC9011337 DOI: 10.3389/fnbeh.2022.788633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Both sensory and metabolic processes guide food intake. Olfactory inputs help coordinate food appreciation and selection, but their role in food consumption and post-feeding physiology remains poorly understood. In this study, using Drosophila melanogaster as a model system, we investigated the effects of olfactory sensory neurons (OSNs) on food consumption, metabolism, and stress responses. We found that dysfunction of OSNs affects diverse processes, including decreased food consumption, increased triacylglycerol level, enhanced stress resistance to starvation or desiccation, and decreased cold resistance. Decreased neuropeptide F receptor (NPFR) level or increased insulin activity in OSNs inhibited food consumption, while impaired NPF signaling or insulin signaling in OSNs increased resistance to starvation and desiccation. These studies provide insights into the function of the olfactory system in control of feeding behaviors and physiology.
Collapse
Affiliation(s)
- Jianzheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Wenjuan Tuo
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xueyan Zhang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuting Dai
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ming Fang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Minghui Xiu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
- *Correspondence: Minghui Xiu,
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Yongqi Liu,
| |
Collapse
|
22
|
Bartholomew CL, Muhlestein JB, Anderson JL, May HT, Knowlton KU, Bair TL, Le VT, Bailey BW, Horne BD. Association of periodic fasting lifestyles with survival and incident major adverse cardiovascular events in patients undergoing cardiac catheterization. Eur J Prev Cardiol 2022; 28:1774-1781. [PMID: 33624026 DOI: 10.1093/eurjpc/zwaa050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
Abstract
AIMS Animal models repeatedly show fasting increases longevity. Human data, though, are limited to anecdotal claims. This study evaluated the association of routine fasting with survival and, secondarily, with incident major adverse cardiovascular events. METHODS AND RESULTS Cardiac catheterization patients enrolled in the Intermountain INSPIRE longitudinal cohort (n = 2785) during 2013-2015 were followed through March 2019. A fasting survey was completed in n = 2025 (73%) of this cohort and 1957 were included in the final data analysis after 68 participants were removed (24 for data issues and 44 for fasting less than 5 years). Self-reported routine fasting behaviour, years of participation in fasting, and other fasting characteristics were surveyed. Mortality was the primary outcome and incident myocardial infarction (MI), stroke, and heart failure (HF) were secondary. Routine fasters (n = 389, mean age 64 ± 14 years, 34% female) averaged 42 ± 18 years of routine fasting (minimum 5 years). Non-fasters (n = 1568, aged 63 ± 14 years, 36% female) included never fasters (n = 1120 with 0 years of fasting) and previous fasters (n = 448 who averaged 32 ± 21 years of prior fasting but had stopped prior to enrolment). Routine fasters had greater survival vs. non-fasters [adjusted hazard ratio (HR) = 0.54, 95% confidence interval (CI) = 0.36-0.80; P = 0.002] and lower incidence of HF (adjusted HR = 0.31, CI = 0.12-0.78; P = 0.013), but not MI or stroke after adjustment. CONCLUSIONS Routine fasting followed during two-thirds of the lifespan was associated with higher survival after cardiac catheterization. This may in part be explained by an association of routine fasting with a lower incidence of HF. CLINICAL STUDY REGISTRATION The Intermountain INSPIRE registry https://clinicaltrials.gov/, NCT02450006.
Collapse
Affiliation(s)
- Ciera L Bartholomew
- Department of Exercise Sciences, 106 SFH, Brigham Young University, Provo, UT 84602, USA
| | - Joseph B Muhlestein
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA.,Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Jeffrey L Anderson
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA.,Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Heidi T May
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA
| | - Kirk U Knowlton
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA.,Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, 9500 Gilman Drive, Mail Code 7411, La Jolla, CA 92037-7411 USA
| | - Tami L Bair
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA
| | - Viet T Le
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA.,Principle PA Faculty, Rocky Mountain University of Health Professions, 122 E 1700 S building 3, Provo, UT 84606, USA
| | - Bruce W Bailey
- Department of Exercise Sciences, 106 SFH, Brigham Young University, Provo, UT 84602, USA
| | - Benjamin D Horne
- Intermountain Medical Center Heart Institute, 5121 S. Cottonwood St., Salt Lake City, UT 84107, USA.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University, 870 Quarry Road, Stanford, CA 94305, USA
| |
Collapse
|
23
|
Maeda N, Fujiki J, Hasegawa Y, Ieko T, Miyasho T, Iwasaki T, Yokota H. Testicular induced corticosterone synthesis in male rats under fasting stress. Steroids 2022; 177:108947. [PMID: 34843801 DOI: 10.1016/j.steroids.2021.108947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 11/28/2022]
Abstract
Testicular steroidogenesis is depressed by adrenal-secreted corticosterone (CORT) under stress. However, the mechanisms are not well understood. This study investigated the details of testicular steroidogenesis depression during fasting. Blood levels of adrenocorticotropic hormone secreted from the pituitary glands increased, but blood CORT was not changed in rats that fasted for 96 h, in spite of the rats being severely stressed. CORT in fasting adult male rats increased more than three times in the testis, but reduced testicular testosterone (T) and blood T levels to 5% and 2% of the control, respectively, was observed. The contents of T precursor (except PGN) were drastically reduced in the fasted-rat testes. Testicular CORT levels were elevated, but the enzymatic activity of cytochrome P45011β, which produces CORT, remained unchanged. The enzymatic activities of 3β-hydroxysteroid dehydrogenase (3β-HSD), mediating the conversion of pregnenolone to progesterone, decreased in the fasted-rat testes. Thus, fasting suppressed testicular steroidogenesis by affecting the enzyme activity of 3β-HSD in the testes and drastically reduced T and increased CORT synthesis. It can be considered that T synthesis involved in cell proliferation is suppressed due to lack of energy during fasting. Conversely, 11β-hydroxylase enzyme activity was induced and CORT synthesis is increased to cope with the fasting stress. Hence, it can be concluded that CORT synthesis in the testes plays a role in the local defense response.
Collapse
Affiliation(s)
- Naoyuki Maeda
- Laboratory of Meat Science and Technology, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Jumpei Fujiki
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Yasuhiro Hasegawa
- Laboratory of Applied Biochemistry, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Takahiro Ieko
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Taku Miyasho
- Laboratory of Animal Biological Responses, Department of Veterinary Science, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Tomohito Iwasaki
- Laboratory of Applied Biochemistry, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido 069-8501, Japan
| | - Hiroshi Yokota
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido 069-8501, Japan.
| |
Collapse
|
24
|
Tesic V, Ciric J, Jovanovic Macura I, Zogovic N, Milanovic D, Kanazir S, Perovic M. Corticosterone and Glucocorticoid Receptor in the Cortex of Rats during Aging-The Effects of Long-Term Food Restriction. Nutrients 2021; 13:nu13124526. [PMID: 34960078 PMCID: PMC8703853 DOI: 10.3390/nu13124526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Numerous beneficial effects of food restriction on aging and age-related pathologies are well documented. It is also well-established that both short- and long-term food restriction regimens induce elevated circulating levels of glucocorticoids, stress-induced hormones produced by adrenal glands that can also exert deleterious effects on the brain. In the present study, we examined the effect of long-term food restriction on the glucocorticoid hormone/glucocorticoid receptor (GR) system in the cortex during aging, in 18- and 24-month-old rats. Corticosterone level was increased in the cortex of aged ad libitum-fed rats. Food restriction induced its further increase, accompanied with an increase in the level of 11β-hydroxysteroid dehydrogenase type 1. However, alterations in the level of GR phosphorylated at Ser232 were not detected in animals on food restriction, in line with unaltered CDK5 level, the decrease of Hsp90, and an increase in a negative regulator of GR function, FKBP51. Moreover, our data revealed that reduced food intake prevented age-related increase in the levels of NFκB, gfap, and bax, confirming its anti-inflammatory and anti-apoptotic effects. Along with an increase in the levels of c-fos, our study provides additional evidences that food restriction affects cortical responsiveness to glucocorticoids during aging.
Collapse
Affiliation(s)
- Vesna Tesic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Jelena Ciric
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Irena Jovanovic Macura
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Nevena Zogovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia;
| | - Desanka Milanovic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Selma Kanazir
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
- Correspondence:
| | - Milka Perovic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| |
Collapse
|
25
|
Jahrami HA, Faris ME, I Janahi A, I Janahi M, Abdelrahim DN, Madkour MI, Sater MS, Hassan AB, Bahammam AS. Does four-week consecutive, dawn-to-sunset intermittent fasting during Ramadan affect cardiometabolic risk factors in healthy adults? A systematic review, meta-analysis, and meta-regression. Nutr Metab Cardiovasc Dis 2021; 31:2273-2301. [PMID: 34167865 DOI: 10.1016/j.numecd.2021.05.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/07/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023]
Abstract
AIMS This study aimed to evaluate the effects of Ramadan diurnal intermittent fasting (RDIF; 29-30 days) on cardiometabolic risk factors (CMRF) in healthy adults, and examine the effect of various cofactors on the outcomes using sub-group meta-regression. DATA SYNTHESIS We conducted a systematic review and meta-analysis to measure the effect sizes of changes in CMRF in healthy adult Muslims observing RDIF. Ten scientific databases (EBSCOhost, CINAHL, Cochrane, EMBASE, PubMed/MEDLINE, Scopus, Google Scholar, ProQuest Medical, ScienceDirect, and Web of Science) were searched from the date of inception (1950) to the end of November 2020. The CMRF searched and analyzed were total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), diastolic blood pressure (DBP), and heart rate (HR). We identified 91 studies (4431 adults aged 18-85 years) conducted between 1982 and 2020 in 23 countries distributed over four continents. RDIF-induced effect sizes for CMRF were: TC (no. of studies K = 77, number of subjects N = 3705, Hedge's g = -0.092, 95% confidence interval (CI): -0.168, 0.016); TG (K = 74, N = 3591, Hedge's g = -0.127, 95% CI: -0.203, 0.051); HDL-C (K = 68, N = 3528, Hedge's g = 0.138, 95% CI: 0.051, 0.224); LDL-C (K = 65, N = 3354, Hedge's g = -0.115, 95% CI: -0.197, -0.034); VLDL-C (K = 13, N = 648, Hedge's g = -0.252, 95% CI: -0.431, 0.073), DBP (K = 32, N = 1716, Hedge's g = -0.255, 95% CI: -0.363, 0.147), and HR (K = 12, N = 674, Hedge's g = -0.082, 95% CI: -0.300, 0.136). Meta-regression revealed that the age of fasting people was a significant moderator of changes in both HDL-C (P = 0.02) and VLDL-C (P = 0.01). Male sex was the only significant moderator of changes in LDL-C (P = 0.055). Fasting time duration was the only significant moderator of HDL-C (P = 0.001) at the end of Ramadan. CONCLUSIONS RDIF positively impacts CMRF, which may confer short-term transient protection against cardiovascular disease among healthy people.
Collapse
Affiliation(s)
- Haitham A Jahrami
- Ministry of Health, Bahrain; College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - MoezAlIslam E Faris
- Department of Clinical Nutrition and Dietetics, College of Health Sciences/Research Institute for Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, United Arab Emirates.
| | - Abdulrahman I Janahi
- College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Mohamed I Janahi
- College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Dana N Abdelrahim
- Department of Nutrition and Food Technology, School of Agriculture, The University of Jordan, Amman, Jordan
| | - Mohamed I Madkour
- Department of Medical Laboratory Sciences, College of Health Sciences/Research Institute for Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, United Arab Emirates
| | - Mai S Sater
- College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Adla B Hassan
- College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Ahmed S Bahammam
- Department of Medicine, College of Medicine, University Sleep Disorders Center, King Saud University, Saudi Arabia; The Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Choi A, Hallett M, Ehrlich D. Nutritional Ketosis in Parkinson's Disease - a Review of Remaining Questions and Insights. Neurotherapeutics 2021; 18:1637-1649. [PMID: 34235637 PMCID: PMC8608995 DOI: 10.1007/s13311-021-01067-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/04/2023] Open
Abstract
Nutritional ketosis has promise for treating Parkinson's disease. Three previous studies explored the use of a ketogenic diet in cohorts with Parkinson's disease, and, while not conclusive, the data suggest non-motor symptom benefit. Before the ketogenic diet can be considered as a therapeutic option, it is important to establish with greater certainty that there is a reliable symptomatic benefit: which symptoms or groups of symptoms are impacted (if non-motor symptoms, which ones, and by which mechanism), what timescale is needed to obtain benefit, and how large an effect size can be achieved? To accomplish this, further investigation into the disease mechanisms based on pre-clinical data and hints from the clinical outcomes to date is useful to understand target engagement and gauge which mechanism could lead to a testable hypothesis. We review research pertaining to ketogenic diet, exogenous ketones, fasting, clinical studies, and theoretical review papers regarding therapeutic mechanisms from direct ketone body signaling and indirect metabolic effects. Through discussion of these findings and consideration of whether the ketogenic diet can be regarded as therapeutically useful for adjunctive therapy for Parkinson's disease, we identify remaining questions for the clinician to consider prior to recommending this diet.
Collapse
Affiliation(s)
- Alexander Choi
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, USA.
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, USA
| | - Debra Ehrlich
- Office of the Clinical Director, Parkinson Disease Clinic, National Institute of Neurological Disorders and Stroke, Bethesda, USA
| |
Collapse
|
27
|
Late-life intermittent fasting decreases aging-related frailty and increases renal hydrogen sulfide production in a sexually dimorphic manner. GeroScience 2021; 43:1527-1554. [PMID: 33675469 PMCID: PMC8492807 DOI: 10.1007/s11357-021-00330-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/25/2021] [Indexed: 12/19/2022] Open
Abstract
Global average life expectancy continues to rise. As aging increases the likelihood of frailty, which encompasses metabolic, musculoskeletal, and cognitive deficits, there is a need for effective anti-aging treatments. It is well established in model organisms that dietary restriction (DR), such as caloric restriction or protein restriction, enhances health and lifespan. However, DR is not widely implemented in the clinic due to patient compliance and its lack of mechanistic underpinnings. Thus, the present study tested the effects of a somewhat more clinically applicable and adoptable DR regimen, every-other-day (EOD) intermittent fasting, on frailty in 20-month-old male and female C57BL/6 mice. Frailty was determined by a series of metabolic, musculoskeletal, and cognitive tasks performed prior to and toward the end of the 2.5-month dietary intervention. Late-life EOD fasting attenuated overall energy intake, hypothalamic inflammatory gene expression, and frailty in males. However, it failed to reduce overall caloric intake and had a little positive effect in females. Given that the selected benefits of DR are dependent on augmented production of the gasotransmitter hydrogen sulfide (H2S) and that renal H2S production declines with age, we tested the effects of EOD fasting on renal H2S production capacity and its connection to frailty in males. EOD fasting boosted renal H2S production, which positively correlated with improvements in multiple components of frailty tasks. Therefore, late-life initiated EOD fasting is sufficient to reduce aging-related frailty, at least in males, and suggests that renal H2S production capacity may modulate the effects of late-life EOD fasting on frailty.
Collapse
|
28
|
Elesawy BH, Raafat BM, Muqbali AA, Abbas AM, Sakr HF. The Impact of Intermittent Fasting on Brain-Derived Neurotrophic Factor, Neurotrophin 3, and Rat Behavior in a Rat Model of Type 2 Diabetes Mellitus. Brain Sci 2021; 11:brainsci11020242. [PMID: 33671898 PMCID: PMC7918995 DOI: 10.3390/brainsci11020242] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 01/17/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is known to be associated with an increased risk of dementia, specifically Alzheimer’s disease and vascular dementia. Intermittent fasting (IF) has been proposed to produce neuroprotective effects through the activation of several signaling pathways. In this study, we investigated the effect of IF on rat behavior in type 2 diabetic rats. Forty male Wistar Kyoto rats were divided into four groups (n = 10 for each): the ad libitum (Ad) group, the intermittent fasting group (IF), the streptozotocin-induced diabetic 2 group (T2DM) fed a high-fat diet for 4 weeks followed by a single intraperitoneal (i.p.) injection of streptozotocin (STZ) 25 mg kg−1, and the diabetic group with intermittent fasting (T2DM+IF). We evaluated the impact of 3 months of IF (16 h of food deprivation daily) on the levels of brain-derived neurotrophic factor (BDNF), neurotrophin 3 (NT3), serotonin, dopamine, and glutamate in the hippocampus, and rat behavior was assessed by the forced swim test and elevated plus maze. IF for 12 weeks significantly increased (p < 0.05) the levels of NT3 and BDNF in both control and T2DM rats. Additionally, it increased serotonin, dopamine, and glutamic acid in diabetic rats. Moreover, IF modulated glucose homeostasis parameters, with a significant decrease (p < 0.05) in insulin resistance and downregulation of serum corticosterone level. Interestingly, T2DM rats showed a significant increase in anxiety and depression behaviors, which were ameliorated by IF. These findings suggest that IF could produce a potentially protective effect by increasing the levels of BDNF and NT3 in both control and T2DM rats. IF could be considered as an additional therapy for depression, anxiety, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Basem H. Elesawy
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Bassem M. Raafat
- Radiological Sciences Department, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Aya Al Muqbali
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Koudh, Muscat PC 123, Oman;
| | - Amr M. Abbas
- Department of Physiology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia;
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Hussein F. Sakr
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Koudh, Muscat PC 123, Oman;
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence:
| |
Collapse
|
29
|
Sayadi JJ, Sayadi L, Satteson E, Chopan M. Nerve injury and repair in a ketogenic milieu: A systematic review of traumatic injuries to the spinal cord and peripheral nervous tissue. PLoS One 2021; 16:e0244244. [PMID: 33395427 PMCID: PMC7781473 DOI: 10.1371/journal.pone.0244244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/04/2020] [Indexed: 12/26/2022] Open
Abstract
Dietary interventions such as intermittent fasting and the ketogenic diet have demonstrated neuroprotective effects in various models of neurological insult. However, there has been a lack of evaluation of these interventions from a surgical perspective despite their potential to augment reparative processes that occur following nerve injury. Thus, we sought to analyze the effects of these dietary regimens on nerve regeneration and repair by critical appraisal of the literature. Following PRISMA guidelines, a systematic review was performed to identify studies published between 1950 and 2020 that examined the impact of either the ketogenic diet or intermittent fasting on traumatic injuries to the spinal cord or peripheral nerves. Study characteristics and outcomes were analyzed for each included article. A total of 1,890 articles were reviewed, of which 11 studies met inclusion criteria. Each of these articles was then assessed based on a variety of qualitative parameters, including type of injury, diet composition, timing, duration, and outcome. In total, seven articles examined the ketogenic diet, while four examined intermittent fasting. Only three studies examined peripheral nerves. Neuroprotective effects manifested as either improved histological or functional benefits in most of the included studies. Overall, we conclude that intermittent fasting and the ketogenic diet may promote neuroprotection and facilitate the regeneration and repair of nerve fibers following injury; however, lack of consistency between the studies in terms of animal models, diet compositions, and timing of dietary interventions preclude synthesis of their outcomes as a whole.
Collapse
Affiliation(s)
- Jamasb Joshua Sayadi
- Stanford University School of Medicine, Stanford, California, United States of America
| | - Lohrasb Sayadi
- Department of Plastic Surgery, University of California, Irvine, California, United States of America
| | - Ellen Satteson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida Health, Gainesville, Florida, United States of America
| | - Mustafa Chopan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Florida Health, Gainesville, Florida, United States of America
| |
Collapse
|
30
|
Grundler F, Mesnage R, Michalsen A, Wilhelmi de Toledo F. Blood Pressure Changes in 1610 Subjects With and Without Antihypertensive Medication During Long-Term Fasting. J Am Heart Assoc 2020; 9:e018649. [PMID: 33222606 PMCID: PMC7763762 DOI: 10.1161/jaha.120.018649] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background We investigated daily blood pressure (BP) changes during fasting periods ranging from 4 to 41 (10.0±3.8) days in a cohort of 1610 subjects, including 920 normotensive, 313 hypertensive nonmedicated, and 377 hypertensive medicated individuals. Methods and Results Subjects underwent a multidisciplinary fasting program with a daily intake of ≈250 kcal. Weight and stress scores decreased during fasting, and the well‐being index increased, documenting a good tolerability. BP mean values decreased from 126.2±18.6/81.4±11.0 to 119.7±15.9/77.6±9.8 mm Hg (mean change, −6.5/3.8 mm Hg). BP changes were larger for hypertensive nonmedicated subjects (>140/90 mm Hg) and reduced by 16.7/8.8 mm Hg. This reduction reached 24.7/13.1 mm Hg for hypertensive nonmedicated subjects (n=76) with the highest BP (>160/100 mm Hg). In the normotensive group, BP decreased moderately by 3.0/1.9 mm Hg. Interestingly, we documented an increase of 6.3/2.2 mm Hg in a subgroup of 69 female subjects with BP <100/60 mm Hg. In the hypertensive medicated group, although BP decreased from 134.6/86.0 to 127.3/81.3 mm Hg, medication was stopped in 23.6% of the subjects, whereas dosage was reduced in 43.5% and remained unchanged in 19.4%. The decrease in BP was larger in subjects fasting longer. Baseline metabolic parameters, such as body mass index and glucose levels, as well as age, can be used to predict the amplitude of the BP decrease during fasting with a machine learning model. Conclusions Long‐term fasting tends to decrease BP in subjects with elevated BP values. This effect persisted during the 4 days of stepwise food reintroduction, even when subjects stopped their antihypertensive medication. Registration URL: https://www.drks.de/drks_web/; Unique identifier: DRKS00010111.
Collapse
Affiliation(s)
- Franziska Grundler
- Buchinger Wilhelmi Clinic Überlingen Germany.,Charité-Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| | - Robin Mesnage
- Gene Expression and Therapy Group Department of Medical and Molecular Genetics Faculty of Life Sciences and Medicine King's College LondonGuy's Hospital London United Kingdom
| | - Andreas Michalsen
- Institute of Social Medicine, Epidemiology and Health Economics Charité- Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt-Universität zu Berlin, and Berlin Institute of Health Berlin Germany.,Department of Internal and Integrative Medicine Immanuel Krankenhaus Berlin Berlin Germany
| | | |
Collapse
|
31
|
Mattson MP. Applying available knowledge and resources to alleviate familial and sporadic neurodegenerative disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:91-107. [PMID: 33453944 DOI: 10.1016/bs.pmbts.2020.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Here I present the scientific rationale and implementation strategies for elimination of early-onset neurodegenerative disorders (EONDD) from future generations, and for risk reduction and treatments for the more common late-onset neurodegenerative disorders (LONDD). Young adults with a family history of an EONDD should be educated on the genetics and familial burden of EONDD. They can then be genotyped and, if positive for the mutation, counseled as to how they can ensure that none of their children will be affected by choosing either adoption or in vitro fertilization and preimplantation genetic testing. LONDD risk reduction will require education of physicians and patients on the benefits of regular intermittent bioenergetic and cognitive challenges (exercise, intermittent fasting, intellectual challenges and social engagement) for brain health, and on specific risk-reduction regimens. Regulations will be required to counteract the disease-promoting mercenary practices of the processed food and pharmaceutical industries. Clinical trials of pharmacological interventions should shift to small trials of agents that substantially mimic mechanisms of action of exercise and intermittent fasting to bolster neuronal bioenergetics and stress resistance.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
32
|
Castro-Barquero S, Ruiz-León AM, Sierra-Pérez M, Estruch R, Casas R. Dietary Strategies for Metabolic Syndrome: A Comprehensive Review. Nutrients 2020; 12:nu12102983. [PMID: 33003472 PMCID: PMC7600579 DOI: 10.3390/nu12102983] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome is a cluster of metabolic risk factors, characterized by abdominal obesity, dyslipidemia, low levels of high-density lipoprotein cholesterol (HDL-c), hypertension, and insulin resistance. Lifestyle modifications, especially dietary habits, are the main therapeutic strategy for the treatment and management of metabolic syndrome, but the most effective dietary pattern for its management has not been established. Specific dietary modifications, such as improving the quality of the foods or changing macronutrient distribution, showed beneficial effects on metabolic syndrome conditions and individual parameters. On comparing low-fat and restricted diets, the scientific evidence supports the use of the Mediterranean Dietary Approaches to Stop Hypertension (DASH) diet intervention as the new paradigm for metabolic syndrome prevention and treatment. The nutritional distribution and quality of these healthy diets allows health professionals to provide easy-to-follow dietary advice without the need for restricted diets. Nonetheless, energy-restricted dietary patterns and improvements in physical activity are crucial to improve the metabolic disturbances observed in metabolic syndrome patients.
Collapse
Affiliation(s)
- Sara Castro-Barquero
- Department of Medicine, Faculty of Medicine and Life Sciences, University of Barcelona, 08036 Barcelona, Spain; (S.C.-B.); (M.S.-P.); (R.E.)
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana María Ruiz-León
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria Sierra-Pérez
- Department of Medicine, Faculty of Medicine and Life Sciences, University of Barcelona, 08036 Barcelona, Spain; (S.C.-B.); (M.S.-P.); (R.E.)
| | - Ramon Estruch
- Department of Medicine, Faculty of Medicine and Life Sciences, University of Barcelona, 08036 Barcelona, Spain; (S.C.-B.); (M.S.-P.); (R.E.)
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Casas
- Department of Medicine, Faculty of Medicine and Life Sciences, University of Barcelona, 08036 Barcelona, Spain; (S.C.-B.); (M.S.-P.); (R.E.)
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red, Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-932275400; Fax: +34-932272907
| |
Collapse
|
33
|
Yang F, Chen H, Gao Y, An N, Li X, Pan X, Yang X, Tian L, Sun J, Xiong X, Xing Y. Gut microbiota-derived short-chain fatty acids and hypertension: Mechanism and treatment. Biomed Pharmacother 2020; 130:110503. [PMID: 34321175 DOI: 10.1016/j.biopha.2020.110503] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Hypertension (HTN) is an growing emerging health issue around across the world. In recent years, increasing attention has been paid to the role of dysbacteriosis in HTN and its underlying mechanism. Short-chain fatty acids (SCFAs), which are novel metabolites of intestinal flora, exert substantial regulatory effects on HTN, providing an exciting avenue for novel therapies for this disease. They function primarily by activating transmembrane G protein-coupled receptors and inhibiting histone acetylation. In this review, we discuss the mechanisms underlying the complex interaction between SCFAs and gut microbiota composition to lower blood pressure by regulating the brain-gut and kidney-gut axes, and the role of high-salt diet, immune system, oxidative stress, and inflammatory mechanism in the development of HTN. Furthermore, we also discuss the various treatment strategies for HTN, including diet, antibiotics, probiotics, fecal microflora transplantation, and traditional Chinese medicine. In conclusion, manipulation of SCFAs opens new avenues to improve treatment of HTN.
Collapse
Affiliation(s)
- Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hengwen Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Na An
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing, China
| | - Xinye Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing, China
| | - Xiandu Pan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Yang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Li Tian
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing, China
| | - Jiahao Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing, China
| | - Xingjiang Xiong
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
34
|
Jung J, Kim AS, Ko HJ, Choi HI, Hong HE. Association between Breakfast Skipping and the Metabolic Syndrome: The Korea National Health and Nutrition Examination Survey, 2017. ACTA ACUST UNITED AC 2020; 56:medicina56080396. [PMID: 32784810 PMCID: PMC7466351 DOI: 10.3390/medicina56080396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 01/22/2023]
Abstract
Background and Objectives: Recently, the prevalence of metabolic syndrome in Korea has increased rapidly. Current knowledge reflects the importance of dietary control in relation to the metabolic syndrome. The objective of this study was to evaluate the influence of skipping breakfast on the metabolic syndrome. Materials and Methods: We conducted a cross-sectional study using data from the Korea National Health and Nutrition Examination Survey 2017 for the second year. A total of 3864 adults aged 20 to 64 were included in the study. We stratified the study population into three groups, based on breakfast patterns: the regular group, irregular group, and skipping group. Multiple logistic regression analysis was used to analyze the association between skipping breakfast and the presence of metabolic syndrome. Results: We noted an increase in the proportion of metabolic syndrome cases as follows: skipping group (3.3%), irregular group (5.4%), and regular group (8.5%) (p < 0.001). The multivariate-adjusted odds ratios of metabolic syndrome in the skipping and irregular groups compared with the regular group were 0.68 (95% CI; 0.35 to 1.35) and 0.81 (95% CI; 0.51 to 1.28), respectively. In the 40-65-year-old age group, which had a high prevalence of metabolic syndrome, the multivariate-adjusted odds ratios of metabolic syndrome in the skipping group compared with regular group were 0.78 (95%CI, 0.39 to 1.62). Conclusions: There was no significant correlation between skipping breakfast and risk factors of metabolic syndrome (after adjusting for risk factors), but a tendency of skipping breakfast to lower the risk of metabolic syndrome was observed. A rationale for these results is proposed through the association between skipping breakfast and intermittent fasting.
Collapse
Affiliation(s)
- JaeHun Jung
- Department of Family Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; (J.J.); (H.-J.K.); (H.-I.C.); (H.-E.H.)
| | - A-Sol Kim
- Department of Family Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Family Medicine, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
- Correspondence:
| | - Hae-Jin Ko
- Department of Family Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; (J.J.); (H.-J.K.); (H.-I.C.); (H.-E.H.)
- Department of Family Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hye-In Choi
- Department of Family Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; (J.J.); (H.-J.K.); (H.-I.C.); (H.-E.H.)
| | - Hee-Eun Hong
- Department of Family Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; (J.J.); (H.-J.K.); (H.-I.C.); (H.-E.H.)
| |
Collapse
|
35
|
Wei W, Tang L, Chen M, Chen S, Zhou H, Ying Z. Intermittent fasting ameliorates PM 2.5 exposure-induced abnormalities in glycaemic control. Toxicol Appl Pharmacol 2020; 404:115181. [PMID: 32758488 DOI: 10.1016/j.taap.2020.115181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 11/25/2022]
Abstract
Exposure to ambient fine particulate matter (PM2.5) elicits various abnormalities in glycaemic control and thus correlates with type 2 diabetes. Intermittent fasting is an emerging treatment for type 2 diabetes. This study, therefore, tested whether intermittent fasting ameliorates PM2.5 exposure-induced abnormalities in glycaemic control. To this end, C57Bl/6 J mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) for 16 weeks and concurrently subject to ad libitum feeding or intermittent fasting. The food intake assessment showed that CAP exposure transiently reduced food intake in ad libitum fed mice, but persistently reduced food intake in intermittently fasted mice. In contrast, CAP exposure persistently promoted mouse weight gain in ad libitum fed mice, while intermittent fasting blocked this CAP exposure-induced weight gain. The glucose homeostasis assessments revealed that CAP exposure elicited insulin resistance and glucose intolerance and meanwhile increased glucose-induced insulin secretion (GIIS). The insulin resistance and glucose intolerance, but not the increase in GIIS, induced by CAP exposure were blocked by intermittent fasting. Analysis of Akt phosphorylation, the indicator of local insulin signaling, showed that CAP exposure reduced insulin signaling in the liver and adipose tissues but not in the skeletal muscle. Intermittent fasting blocked CAP exposure-induced insulin resistance in the liver but not in the adipose tissues. The present study demonstrates that intermittent fasting ameliorates PM2.5 exposure-induced insulin resistance and glucose intolerance, strongly supporting that it may be used to prevent type 2 diabetes due to exposure to PM2.5.
Collapse
Affiliation(s)
- Wei Wei
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Bile Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Tang
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Bile Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minjie Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sufang Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Endocrinology, the People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou, Henan 450003, China
| | - Huifen Zhou
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pathology, Hubei University of Science and Technology, Xianning, Hubei 437100, China
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Wilhelmi de Toledo F, Grundler F, Sirtori CR, Ruscica M. Unravelling the health effects of fasting: a long road from obesity treatment to healthy life span increase and improved cognition. Ann Med 2020; 52:147-161. [PMID: 32519900 PMCID: PMC7877980 DOI: 10.1080/07853890.2020.1770849] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years a revival of interest has emerged in the health benefits of intermittent fasting and long-term fasting, as well as of other related nutritional strategies. In addition to meal size and composition a new focus on time and frequency of meals has gained attention. The present review will investigate the effects of the main forms of fasting, activating the metabolic switch from glucose to fat and ketones (G-to-K), starting 12-16 h after cessation or strong reduction of food intake. During fasting the deactivation of mTOR regulated nutrient signalling pathways and activation of the AMP protein kinase trigger cell repair and inhibit anabolic processes. Clinical and animal studies have clearly indicated that modulating diet and meal frequency, as well as application of fasting patterns, e.g. intermittent fasting, periodic fasting, or long-term fasting are part of a new lifestyle approach leading to increased life and health span, enhanced intrinsic defences against oxidative and metabolic stresses, improved cognition, as well as a decrease in cardiovascular risk in both obese and non-obese subjects. Finally, in order to better understand the mechanisms beyond fasting-related changes, human studies as well as non-human models closer to human physiology may offer useful clues.KEY-MESSAGESBiochemical changes during fasting are characterised by a glucose to ketone switch, leading to a rise of ketones, advantageously used for brain energy, with consequent improved cognition.Ketones reduce appetite and help maintain effective fasting.Application of fasting patterns increases healthy life span and defences against oxidative and metabolic stresses.Today's strategies for the use of therapeutic fasting are based on different protocols, generally relying on intermittent fasting, of different duration and calorie intake.Long-term fasting, with durations between 5 and 21 days can be successfully repeated in the course of a year.
Collapse
Affiliation(s)
| | - Franziska Grundler
- Buchinger Wilhelmi Clinic, Wilhelm-Beck-Straße 27, Überlingen, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Cesare R Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
37
|
Gut Microbiota during Dietary Restrictions: New Insights in Non-Communicable Diseases. Microorganisms 2020; 8:microorganisms8081140. [PMID: 32731505 PMCID: PMC7465033 DOI: 10.3390/microorganisms8081140] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
In recent decades, there has been a growing interest in dietary restrictions for their promising effects on longevity and health span. Indeed, these strategies are supposed to delay the onset and burden of non-communicable diseases (NCDs) such as obesity, diabetes, cancer and neurological and gastrointestinal inflammatory diseases. At the same time, the gut microbiota has been shown to play a crucial role in NCDs since it is actively involved in maintaining gut homeostasis through its impact on nutrients metabolism, gut barrier, and immune system. There is evidence that dietary restrictions could slow down age-related changes in the types and numbers of gut bacteria, which may counteract gut dysbiosis. The beneficial effects on gut microbiota may positively influence host metabolism, gut barrier permeability, and brain functions, and subsequently, postpone the onset of NCDs prolonging the health span. These new insights could lead to the development of novel strategies for modulating gut microbiota with the end goal of treating/preventing NCDs. This review provides an overview of animal and human studies focusing on gut microbiota variations during different types of dietary restriction, in order to highlight the close relationship between gut microbiota balance and the host's health benefits induced by these nutritional regimens.
Collapse
|
38
|
Abstract
A healthy gut microbiota is essential in maintaining the human body in a homeostatic state by its functions in digestion and immune tolerance. Under states of aberrant microbial composition or function (dysbiosis), the gut microbiota induces systemic inflammation that can lead to the onset of many diseases. In this review, we describe some evidence, largely from rodent studies, that supports the possible role of a dysbiotic gut microbiota in the onset and exacerbation of ocular diseases, primarily diabetic retinopathy, age-related macular degeneration, choroidal neovascularization, and uveitis. Furthermore, we examine several potential therapeutic measures that show promise in restoring the gut microbiota to a eubiotic state, preventing the aforementioned disease pathologies.
Collapse
Affiliation(s)
- Jason L Floyd
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maria B Grant
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
39
|
Badreh F, Joukar S, Badavi M, Rashno M, Dehesh T. The Effects of Age and Fasting Models on Blood Pressure, Insulin/Glucose Profile, and Expression of Longevity Proteins in Male Rats. Rejuvenation Res 2020; 23:224-236. [DOI: 10.1089/rej.2019.2205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Firuzeh Badreh
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Siyavash Joukar
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Science, Kerman, Iran
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Badavi
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tania Dehesh
- Department of Epidemiology and Biostatistics, School of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
40
|
Lee JH, Verma N, Thakkar N, Yeung C, Sung HK. Intermittent Fasting: Physiological Implications on Outcomes in Mice and Men. Physiology (Bethesda) 2020; 35:185-195. [PMID: 32293230 DOI: 10.1152/physiol.00030.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Intermittent fasting (IF) is a widely practiced dietary method that encompasses periodic restriction of food consumption. Due to its protective benefits against metabolic diseases, aging, and cardiovascular and neurodegenerative diseases, IF continues to gain attention as a preventative and therapeutic intervention to counteract these chronic diseases. Although numerous animal studies have reported positive health benefits of IF, its feasibility and efficacy in clinical settings remain controversial. Importantly, since dietary interventions such as IF have systemic effects, thoroughly investigating the tissue-specific changes in animal models is crucial to identify IF's mechanism and evaluate its potential adverse effects in humans. As such, we will review and compare the outcomes and underlying mechanisms of IF in both animal and human studies. Moreover, the limitations of IF and inconsistencies between preclinical and clinical studies will be discussed to provide insight into the gaps between translating research from bench to bedside.
Collapse
Affiliation(s)
- Ju Hee Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Navkiran Verma
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Nikita Thakkar
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Christy Yeung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Affiliation(s)
- Ali Tootee
- 1Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- 2Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Chakraborty A, Banerjee S, Mukherjee B, Poddar MK. Calorie restriction improves aging-induced impairment of cognitive function in relation to deregulation of corticosterone status and brain regional GABA system. Mech Ageing Dev 2020; 189:111248. [PMID: 32339520 DOI: 10.1016/j.mad.2020.111248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/16/2020] [Accepted: 04/05/2020] [Indexed: 12/21/2022]
Abstract
Aging is known to affect adversely the corticosterone status and the brain function including cognition. Calorie restricted (CR) diet has been found to improve brain aging. The objective of the present investigation is to study the effect of short-term CR diet without any food deprivation on aging-induced impairment of cognitive function in relation to the corticosterone status and the brain regional GABA system. The result showed that aging-induced deregulation of the brain regional GABA system, increase in plasma and adrenal corticosterone levels and cognitive impairment were attenuated with short-term CR diet supplementation for consecutive 1 and 2 months to the aged (18 and 24 months) rats. But in young rats (4 months) consumption of the same CR diet under similar conditions reversibly affected those above-mentioned parameters. These results, thus suggest that (a) aging down-regulates brain regional GABA system with an up-regulation of corticosterone status and impairment of cognitive function, (b) CR diet consumption improves this aging-induced deregulation of brain regional GABA system, corticosterone status, and cognitive function, (c) these attenuating effects of CR diet are greater with a longer period of consumption but (d) CR diet consumption is harmful to young rats as observed in those parameters.
Collapse
Affiliation(s)
- Apala Chakraborty
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S.C. Mallick Road, Kolkata, 700032, India
| | - Soumyabrata Banerjee
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S.C. Mallick Road, Kolkata, 700032, India
| | - Biswajit Mukherjee
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S.C. Mallick Road, Kolkata, 700032, India
| | - Mrinal Kanti Poddar
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S.C. Mallick Road, Kolkata, 700032, India.
| |
Collapse
|
43
|
Koutroumpakis E, Jozwik B, Aguilar D, Taegtmeyer H. Strategies of Unloading the Failing Heart from Metabolic Stress. Am J Med 2020; 133:290-296. [PMID: 31520618 PMCID: PMC7054139 DOI: 10.1016/j.amjmed.2019.08.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
Abstract
We propose a unifying perspective of heart failure in patients with type 2 diabetes mellitus. The reasoning is as follows: cellular responses to fuel overload include dysregulated insulin signaling, impaired mitochondrial respiration, reactive oxygen species formation, and the accumulation of certain metabolites, collectively termed glucolipotoxicity. As a consequence, cardiac function is impaired, with intracellular calcium cycling and diastolic dysfunction as an early manifestation. In this setting, increasing glucose uptake by insulin or insulin sensitizing agents only worsens the disrupted fuel homeostasis of the heart. Conversely, restricting fuel supply by means of caloric restriction, surgical intervention, or certain pharmacologic agents will improve cardiac function by restoring metabolic homeostasis. The concept is borne out by clinical interventions, all of which unload the heart from metabolic stress.
Collapse
Affiliation(s)
- Efstratios Koutroumpakis
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Bartosz Jozwik
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - David Aguilar
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston.
| |
Collapse
|
44
|
Affiliation(s)
- Rafael de Cabo
- From the Translational Gerontology Branch (R.C.) and the Laboratory of Neurosciences (M.P.M.), Intramural Research Program, National Institute on Aging, National Institutes of Health, and the Department of Neuroscience, Johns Hopkins University School of Medicine (M.P.M.) - both in Baltimore
| | - Mark P Mattson
- From the Translational Gerontology Branch (R.C.) and the Laboratory of Neurosciences (M.P.M.), Intramural Research Program, National Institute on Aging, National Institutes of Health, and the Department of Neuroscience, Johns Hopkins University School of Medicine (M.P.M.) - both in Baltimore
| |
Collapse
|
45
|
Allaf M, Elghazaly H, Mohamed OG, Fareen MFK, Zaman S, Salmasi AM, Tsilidis K, Dehghan A. Intermittent fasting for the prevention of cardiovascular disease. Cochrane Database Syst Rev 2019. [DOI: 10.1002/14651858.cd013496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | | | | | | | - Sadia Zaman
- Imperial College London; School of Medicine; London UK
| | - Abdul-Majeed Salmasi
- London North West University Healthcare NHS Trust; Department of Cardiology; London UK
| | - Kostas Tsilidis
- Imperial College London; Department of Epidemiology and Biostatistics, School of Public Health; London UK
- University of Ioannina School of Medicine; Department of Hygiene and Epidemiology; Ioannina Greece
| | - Abbas Dehghan
- Imperial College London; School of Public Health; London UK
| |
Collapse
|
46
|
Fasting as a Therapy in Neurological Disease. Nutrients 2019; 11:nu11102501. [PMID: 31627405 PMCID: PMC6836141 DOI: 10.3390/nu11102501] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Fasting is deeply entrenched in evolution, yet its potential applications to today’s most common, disabling neurological diseases remain relatively unexplored. Fasting induces an altered metabolic state that optimizes neuron bioenergetics, plasticity, and resilience in a way that may counteract a broad array of neurological disorders. In both animals and humans, fasting prevents and treats the metabolic syndrome, a major risk factor for many neurological diseases. In animals, fasting probably prevents the formation of tumors, possibly treats established tumors, and improves tumor responses to chemotherapy. In human cancers, including cancers that involve the brain, fasting ameliorates chemotherapy-related adverse effects and may protect normal cells from chemotherapy. Fasting improves cognition, stalls age-related cognitive decline, usually slows neurodegeneration, reduces brain damage and enhances functional recovery after stroke, and mitigates the pathological and clinical features of epilepsy and multiple sclerosis in animal models. Primarily due to a lack of research, the evidence supporting fasting as a treatment in human neurological disorders, including neurodegeneration, stroke, epilepsy, and multiple sclerosis, is indirect or non-existent. Given the strength of the animal evidence, many exciting discoveries may lie ahead, awaiting future investigations into the viability of fasting as a therapy in neurological disease.
Collapse
|
47
|
Rowland NE, Toth LA. Analytic and Interpretational Pitfalls to Measuring Fecal Corticosterone Metabolites in Laboratory Rats and Mice. Comp Med 2019; 69:337-349. [PMID: 31578162 DOI: 10.30802/aalas-cm-18-000119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Minimization and alleviation of stress are generally viewed as desirable aspects of laboratory animal management and use. However, achieving that goal requires an unambiguous and valid measure of stress. Glucocorticoid concentrations are commonly used as a physiologic index of stress. Measurement of glucocorticoids in blood, serum or plasma clearly reflects many types of both acute and chronic stress. However, the rapid rise in concentrations of circulating glucocorticoids that occurs even with relatively simple manipulations such as handling has led to the increased use of fecal glucocorticoid metabolite (FCM) assays, which provide a temporally integrated measure that may allow a more accurate interpretation of chronic stressors. In this review, we consider 3 aspects of glucocorticoids as a measure of stress. First, we discuss the analytic and interpretational pitfalls of using FCM concentrations as an index of stress in mice and rats. Second, we consider evidence that some degree of stress may benefit animals by priming physiologic and behavioral adaptations that render the animals more resilient in the face of stress. Finally, we use 2 situations-social housing and food restriction-to illustrate the concept of hormesis-a biologic phenomenon in which a low dose or intensity of a challenge has a beneficial effect, whereas exposure to high doses or intensities is detrimental.
Collapse
Affiliation(s)
- Neil E Rowland
- Department of Psychology, University of Florida, Gainesville, Florida;,
| | - Linda A Toth
- Department of Pharmacology, School of Medicine, Southern Illinois University, Springfield, Illinois
| |
Collapse
|
48
|
Harney DJ, Hutchison AT, Hatchwell L, Humphrey SJ, James DE, Hocking S, Heilbronn LK, Larance M. Proteomic Analysis of Human Plasma during Intermittent Fasting. J Proteome Res 2019; 18:2228-2240. [PMID: 30892045 PMCID: PMC6503536 DOI: 10.1021/acs.jproteome.9b00090] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intermittent fasting (IF) increases lifespan and decreases metabolic disease phenotypes and cancer risk in model organisms, but the health benefits of IF in humans are less clear. Human plasma derived from clinical trials is one of the most difficult sample sets to analyze using mass spectrometry-based proteomics due to the extensive sample preparation required and the need to process many samples to achieve statistical significance. Here, we describe an optimized and accessible device (Spin96) to accommodate up to 96 StageTips, a widely used sample preparation medium enabling efficient and consistent processing of samples prior to LC-MS/MS. We have applied this device to the analysis of human plasma from a clinical trial of IF. In this longitudinal study employing 8-weeks IF, we identified significant abundance differences induced by the IF intervention, including increased apolipoprotein A4 (APOA4) and decreased apolipoprotein C2 (APOC2) and C3 (APOC3). These changes correlated with a significant decrease in plasma triglycerides after the IF intervention. Given that these proteins have a role in regulating apolipoprotein particle metabolism, we propose that IF had a positive effect on lipid metabolism through modulation of HDL particle size and function. In addition, we applied a novel human protein variant database to detect common protein variants across the participants. We show that consistent detection of clinically relevant peptides derived from both alleles of many proteins is possible, including some that are associated with human metabolic phenotypes. Together, these findings illustrate the power of accessible workflows for proteomics analysis of clinical samples to yield significant biological insight.
Collapse
Affiliation(s)
- Dylan J Harney
- Charles Perkins Centre, School of Life and Environmental Sciences , University of Sydney , Sydney , NSW 2006 , Australia
| | - Amy T Hutchison
- Discipline of Medicine , University of Adelaide , Adelaide , SA 5005 , Australia
| | - Luke Hatchwell
- Charles Perkins Centre, School of Life and Environmental Sciences , University of Sydney , Sydney , NSW 2006 , Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences , University of Sydney , Sydney , NSW 2006 , Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences , University of Sydney , Sydney , NSW 2006 , Australia
| | - Samantha Hocking
- Central Clinical School, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Leonie K Heilbronn
- Discipline of Medicine , University of Adelaide , Adelaide , SA 5005 , Australia
| | - Mark Larance
- Charles Perkins Centre, School of Life and Environmental Sciences , University of Sydney , Sydney , NSW 2006 , Australia
| |
Collapse
|
49
|
de Bruijn R, Romero LM. The role of glucocorticoids in the vertebrate response to weather. Gen Comp Endocrinol 2018; 269:11-32. [PMID: 30012539 DOI: 10.1016/j.ygcen.2018.07.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022]
Abstract
Changes in the environment related to inclement weather can threaten survival and reproductive success both through direct adverse exposure and indirectly by decreasing food availability. Glucocorticoids, released during activation of the hypothalamic-pituitary-adrenal axis as part of the stress response, are an important candidate for linking vertebrate coping mechanisms to weather. This review attempts to determine if there is a consensus response of glucocorticoids to exposure to weather-related stimuli, including food availability, precipitation, temperature and barometric pressure. The included studies cover field and laboratory studies for all vertebrate taxa, and are separated into four exposure periods, e.g., hours, days, weeks and months. Each reported result was assigned a score based on the glucocorticoid response, e.g., increased, no change, or decreased. Short-term exposure to weather-related stimuli, of up to 24 h, is generally associated with increased glucocorticoids (79% of studies), suggesting that these stimuli are perceived as stressors by most animals. In contrast, the pattern for exposures longer than 24 h shows more variation, even though a majority of studies still report an increase (64%). Lack of glucocorticoid increases appeared to result from instances where: (1) prolonged exposure was a predictable part of the life history of an animal; (2) environmental context was important for the ultimate effect of a stimulus (e.g., precipitation limited food availability in one environment, but increased food in another); (3) prolonged exposure induced chronic stress; and (4) long-term responses appeared to reflect adaptations to seasonal shifts, instead of to short-term weather. However, there is a strong bias towards studies in domesticated laboratory species and wild animals held in captivity, indicating a need for field studies, especially in reptiles and amphibians. In conclusion, the accumulated literature supports the hypothesis that glucocorticoids can serve as the physiological mechanism promoting fitness during inclement weather.
Collapse
Affiliation(s)
- Robert de Bruijn
- Department of Biology, Tufts University, Medford, MA 02155, USA.
| | - L Michael Romero
- Department of Biology, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
50
|
Chadda KR, Ajijola OA, Vaseghi M, Shivkumar K, Huang CLH, Jeevaratnam K. Ageing, the autonomic nervous system and arrhythmia: From brain to heart. Ageing Res Rev 2018; 48:40-50. [PMID: 30300712 DOI: 10.1016/j.arr.2018.09.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/21/2018] [Accepted: 09/30/2018] [Indexed: 02/08/2023]
Abstract
An ageing myocardium possesses significant electrophysiological alterations that predisposes the elderly patient to arrhythmic risk. Whilst these alterations are intrinsic to the cardiac myocytes, they are modulated by the cardiac autonomic nervous system (ANS) and consequently, ageing of the cardiac ANS is fundamental to the development of arrhythmias. A systems-based approach that incorporates the influence of the cardiac ANS could lead to better mechanistic understanding of how arrhythmogenic triggers and substrates interact spatially and temporally to produce sustained arrhythmia and why its incidence increases with age. Despite the existence of physiological oscillations of ANS activity on the heart, pathological oscillations can lead to defective activation and recovery properties of the myocardium. Such changes can be attributable to the decrease in functionality and structural alterations to ANS specific receptors in the myocardium with age. These altered ANS adaptive responses can occur either as a normal ageing process or accelerated in the presence of specific cardiac pathologies, such as genetic mutations or neurodegenerative conditions. Targeted intervention that seek to manipulate the ageing ANS influence on the myocardium may prove to be an efficacious approach for the management of arrhythmia in the ageing population.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center, UCLA Health System/David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Marmar Vaseghi
- UCLA Cardiac Arrhythmia Center, UCLA Health System/David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center, UCLA Health System/David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, CB2 1QW, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom.
| |
Collapse
|