1
|
Mao L, Liu A, Zhang X. Effects of Intermittent Fasting on Female Reproductive Function: A Review of Animal and Human Studies. Curr Nutr Rep 2024; 13:786-799. [PMID: 39320714 DOI: 10.1007/s13668-024-00569-1] [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] [Accepted: 08/20/2024] [Indexed: 09/26/2024]
Abstract
PURPOSE OF REVIEW Intermittent fasting has gained significant attention, yet a comprehensive understanding of its impact on female reproductive health is lacking. This review aims to fill this gap by examining various intermittent fasting regimens and their effects on female reproductive function, along with potential mechanisms. RECENT FINDINGS In healthy non-overweight/obese or pregnant animal models, alternate-day fasting (ADF) and an 8-h time-restricted feeding (TRF) window may have adverse effects on reproductive function. However, these regimens show potential to mitigate negative consequences induced by a high-fat diet (HFD) or environmental exposure. A 10-h TRF demonstrates benefits in improving fertility in both normal-weight and HFD-fed animal models. In women with overweight/obesity or polycystic ovary syndrome (PCOS), the 5:2 diet and TRF significantly reduce the free androgen index while elevating sex hormone binding globulin, promising improvements in menstrual regulation. For pregnant Muslim women, available data do not strongly indicate adverse effects of Ramadan fasting on preterm delivery, but potential downsides to maternal weight gain, neonatal birthweight, and long-term offspring health need consideration. Factors linking intermittent fasting to female reproductive health include the circadian clock, gut microbiota, metabolic regulators, and modifiable lifestyles. Drawing definitive conclusions remains challenging in this evolving area. Nonetheless, our findings underscore the potential utility of intermittent fasting regimens as a therapeutic approach for addressing menstruation irregularities and infertility in women with obesity and PCOS. On the other hand, pregnant women should remain cognizant of potential risks associated with intermittent fasting practices.
Collapse
Affiliation(s)
- Lei Mao
- Department of Women's Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Aixia Liu
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| | - Xiaohui Zhang
- Department of Women's Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
2
|
Zhou Y, Guo X, Liu Z, Sun D, Liang Y, Shen H, Li X, Mu J, Liu J, Cao G, Chen M. 6-week time-restricted eating improves body composition, maintains exercise performance, without exacerbating eating disorder in female DanceSport dancers. J Int Soc Sports Nutr 2024; 21:2369613. [PMID: 38904148 PMCID: PMC11195454 DOI: 10.1080/15502783.2024.2369613] [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: 11/20/2023] [Accepted: 06/13/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Despite the high risk of eating disorder (ED)-related attitudes and behaviors among female dancers, targeted scientific dietary regimens are currently inadequate. Time-restricted eating (TRE), a popular intermittent fasting protocol, has been shown to be effective in enhancing body composition and exercise performance in athletes. In this study, TRE was employed as a dietary regimen to improve body composition and exercise performance and address ED attitudes and behaviors in DanceSport dancers. METHODS Twenty female DanceSport dancers were recruited and divided into two groups: TRE (n = 10) and normal diet (ND) (n = 10). The TRE group consumed their self-selected necessary energy intake exclusively between 11 a.m. and 7 p.m. (utilizing a 16-hour fasting and 8-hour eating window) for 6 weeks, while the ND group maintained their regular dieting patterns. The consumption of water, black tea, or coffee without added sugar or milk was not restricted. Physical activity and calorie intake were systematically recorded during the TRE intervention. Body composition, aerobic and anaerobic performance, and ED attitudes and behaviors were assessed before and after the TRE intervention. The trial was registered in the Chinese Clinical Trial Registry under the identifier ChiCTR2200063780. RESULTS The fixed effects tests (p < 0.0001) and estimates for the intercept (p < 0.0001) of hunger level indicated a noticeable effect on the initial state of hunger during TRE. No significant differences were observed in ED attitudes or behaviors (p > 0.05). TRE resulted in a reduction in hip circumference (p = 0.039), fat mass (kg) (p = 0.0004), and body fat percentage (p = 0.0005), with no significant decrease in fat-free mass (p > 0.05). No significant improvement was observed in aerobic performance (p > 0.05). The average power (AP) (p = 0.01) and AP/Body weight ratio (p = 0.003) significantly increased. Additionally, the power drop decreased significantly (p = 0.019). Group-by-time interactions were observed for fat mass (kg) (p = 0.01), body fat percentage (p = 0.035), and AP/Body weight (p = 0.020). CONCLUSION TRE can be considered a feasible nutritional strategy for DanceSport dancers, facilitating improvements in body composition without compromising aerobic and anaerobic exercise performance or exacerbating ED attitudes and behaviors. Moreover, TRE may facilitate more favorable physiological adaptations, potentially contributing to improved exercise performance.
Collapse
Affiliation(s)
- Yanbing Zhou
- Beijing Sport University, School of Art, Beijing, China
| | - Xian Guo
- Beijing Sport University, Sport Science School, Beijing, China
- Beijing Sport University, Beijing Sports Nutrition Engineering Research Center, Beijing, China
| | - Zeyao Liu
- Beijing Municipal Bureau of Sports, Beijing Lucheng Sports Technical School, Beijing, China
| | - Dan Sun
- Beijing Sport University, Competitive Sport School, Beijing, China
| | - Yujie Liang
- Beijing Sport University, School of Art, Beijing, China
| | - Hong Shen
- Beijing Sport University, School of Art, Beijing, China
| | - Xinxin Li
- Beijing Sport University, Sport Science School, Beijing, China
| | - Jinhao Mu
- Beijing Sport University, Sport Science School, Beijing, China
| | - Jingying Liu
- Beijing Sport University, Sport Science School, Beijing, China
| | - Guoxia Cao
- Beijing Sport University, Sport Science School, Beijing, China
| | - Mengmeng Chen
- Beijing Sport University, Sport Science School, Beijing, China
| |
Collapse
|
3
|
Selman C. The dietary exposome: a brief history of diet, longevity, and age-related health in rodents. Clin Sci (Lond) 2024; 138:1343-1356. [PMID: 39444221 DOI: 10.1042/cs20241248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
It has been recognized for over a century that feeding animals less food than they would normally eat increases lifespan and leads to broad-spectrum improvements in age-related health. A significant number of studies have subsequently shown that restricting total protein, branched chain amino acids or individual amino acids in the diet, as well as ketogenic diets, can elicit similar effects. In addition, it is becoming clear that fasting protocols, such as time-restricted-feeding or every-other-day feeding, without changes in overall energy intake can also profoundly affect rodent longevity and late-life health. In this review, I will provide a historical perspective on various dietary interventions that modulate ageing in rodents and discuss how this understanding of the dietary exposome may help identify future strategies to maintain late-life health and wellbeing in humans.
Collapse
Affiliation(s)
- Colin Selman
- School of Molecular Biosciences, University of Glasgow, Glasgow, United Kingdom, G12 8QQ
| |
Collapse
|
4
|
Benjamin JI, Pati P, Luong T, Liu X, De Miguel C, Pollock JS, Pollock DM. Chronic mistimed feeding results in renal fibrosis and disrupted circadian blood pressure rhythms. Am J Physiol Renal Physiol 2024; 327:F683-F696. [PMID: 39205662 DOI: 10.1152/ajprenal.00047.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Circadian disruption is a disturbance in biological timing, which can occur within or between different organizational levels, ranging from molecular rhythms within specific cells to the misalignment of behavioral and environmental cycles. Previous work from our group showed that less than 1 wk of food restriction to the light (inactive) period is sufficient to invert diurnal blood pressure rhythms in mice. However, kidney excretory rhythms and functions remained aligned with the light-dark cycle. Shift workers have an increased risk of cardiovascular disease that may different between sexes and often have irregular mealtimes, making the possibility of mistimed feeding as a potential contributor to the development of kidney disease. Thus, we hypothesized that chronic mistimed food intake would result in adverse cardiorenal effects, with sex differences in severity. Here, we show that chronic circadian disruption via mistimed feeding results in renal fibrosis and aortic stiffness in a sex-dependent manner. Our results indicate the importance of meal timing for the maintenance of blood pressure rhythms and kidney function, particularly in males. Our results also demonstrate that females are better able to acclimate to circadian-related behavioral change. NEW & NOTEWORTHY Circadian disruption through mistimed feeding resulted in nondipping blood pressure, renal fibrosis, and arterial stiffness that were less severe in females versus males. Mice fed exclusively during the daytime maintain their circadian rhythms of locomotor activity regardless of their loss of blood pressure rhythms. Although these mice ate less food, they maintained body weight, suggesting inefficiencies in overall metabolism. These findings demonstrate the importance of maintaining optimal food intake patterns to prevent cardiorenal pathophysiology.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Paramita Pati
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tha Luong
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Xiaofen Liu
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Liu T, Zhou L, Dong R, Qu Y, Liu Y, Song W, Lv J, Wu S, Peng W, Shi L. Isomalto-Oligosaccharide Potentiates Alleviating Effects of Intermittent Fasting on Obesity-Related Cognitive Impairment during Weight Loss and the Rebound Weight Gain. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23875-23892. [PMID: 39431286 DOI: 10.1021/acs.jafc.4c07351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Obesity-related cognitive dysfunction poses a significant threat to public health. The present study demonstrated mitigating effects of intermittent fasting (IF) and its combination with isomalto-oligosaccharides and IF (IF + IMO) on cognitive impairments induced by a high-fat-high-fructose (HFHF) diet in mice, with IF + IMO exhibiting superior effects. Transcriptomic analysis of the hippocampus revealed that the protective effects on cognition might be attributed to the suppression of toll-like receptor 4 (TLR4)/NFκB signaling, oxidative phosphorylation, and neuroinflammation. Moreover, both IF and IF + IMO modulated the gut microbiome and promoted the production of short-chain fatty acids, with IF + IMO displaying more pronounced effects. IF + IMO-modulated gut microbiota, metabolites, and molecular targets associated with cognitive impairments were further corroborated using human data from public databases Gmrepo and gutMgene. Furthermore, the fecal microbiome transplantation confirmed the direct impacts of IF + IMO-derived microbiota on improving cognition functions by suppressing TLR4/NFκB signaling and increasing BDNF levels. Notably, prior exposure to IF + IMO prevented weight-regain-induced cognitive decline, suppressed TLR4/NFκB signaling and inflammatory cytokines in the hippocampus, and mitigated weight regain-caused gut dysbacteriosis without altering body weight. Our study underscores that IMO-augmented alleviating effects of IF on obesity-related cognitive impairment particularly during weight-loss and weight-regain periods, presenting a novel nutritional strategy to tackle obesity-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Lanqi Zhou
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Rui Dong
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Yizhe Qu
- School of Physical Education, Shaanxi Normal University, Xi'an 710062, China
| | - Yuan Liu
- School of Physical Education, Shaanxi Normal University, Xi'an 710062, China
| | - Wei Song
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Jiayao Lv
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Shan Wu
- College of Food Science and Technology, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Wen Peng
- Nutrition and Health Promotion Center, Department of Public Health, Medical College, Qinghai University, Xining 810016, Qinghai, China
- Qinghai Provincial Key Laboratory of Traditional Chinese Medicine Research for Glucolipid Metabolic Diseases, Xining 810016, Qinghai, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| |
Collapse
|
6
|
Ginefra P, Hope HC, Lorusso G, D'Amelio P, Vannini N. The immunometabolic roots of aging. Curr Opin Immunol 2024; 91:102498. [PMID: 39461330 DOI: 10.1016/j.coi.2024.102498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 10/29/2024]
Abstract
Aging is one of the greatest risk factors for several chronic diseases and is accompanied by a progressive decline of cellular and organ function. Recent studies have highlighted the changes in metabolism as one of the main drivers of organism dysfunctions during aging and how that strongly deteriorate immune cell performance and function. Indeed, a dysfunctional immune system has been shown to have a pleiotropic impact on the organism, accelerating the overall aging process of an individual. Intrinsic and extrinsic factors are responsible for such metabolic alterations. Understanding the contribution, regulation, and connection of these different factors is fundamental to comprehend the process of aging and develop approaches to mitigate age-related immune decline. Here, we describe metabolic perturbations occurring at cellular and systemic levels. Particularly, we emphasize the interplay between metabolism and immunosenescence and describe novel interventions to protect immune function and promote health span.
Collapse
Affiliation(s)
- Pierpaolo Ginefra
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Helen C Hope
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Girieca Lorusso
- Service of Geriatric Medicine, Department of Internal Medicine, CHUV University Hospital of Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Patrizia D'Amelio
- Service of Geriatric Medicine, Department of Internal Medicine, CHUV University Hospital of Lausanne, University of Lausanne, Lausanne, Switzerland.
| | - Nicola Vannini
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
7
|
Livelo C, Guo Y, Madhanagopal J, Morrow C, Melkani GC. Time-restricted feeding mediated modulation of microbiota leads to changes in muscle physiology in Drosophila obesity models. Aging Cell 2024:e14382. [PMID: 39446089 DOI: 10.1111/acel.14382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Recent research has highlighted the essential role of the microbiome in maintaining skeletal muscle physiology. The microbiota influences muscle health by regulating lipid metabolism, protein synthesis, and insulin sensitivity. However, metabolic disturbances such as obesity can lead to dysbiosis, impairing muscle function. Time-restricted feeding (TRF) has been shown to mitigate obesity-related muscle dysfunction, but its effects on restoring healthy microbiomes remain poorly understood. This study utilizes 16S microbiome analysis and bacterial supplementation to investigate the bacterial communities influenced by TRF that may benefit skeletal muscle physiology. In wild-type and obese Drosophila models (axenic models devoid of natural microbial communities), the absence of microbiota influence muscle performance and metabolism differently. Specifically, axenic wild-type Drosophila exhibited reduced muscle performance, higher glucose levels, insulin resistance, ectopic lipid accumulation, and decreased ATP levels. Interestingly, in obese Drosophila (induced by a high-fat diet or predisposed obesity mutant Sk2), the absence of microbiota improved muscle performance, lowered glucose levels, reduced insulin resistance, and increased ATP levels. TRF was found to modulate microbiota composition, notably increasing Acetobacter pasteurianus (AP) and decreasing Staphylococcus aureus (SA) in both obesity models. Supplementation with AP improved muscle performance and reduced glucose and insulin resistance, while SA supplementation had the opposite effect. This study provides novel insights into the complex interactions between TRF, microbiota, and skeletal muscle physiology in different Drosophila models.
Collapse
Affiliation(s)
- Christopher Livelo
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yiming Guo
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jagathnarayan Madhanagopal
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey Morrow
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Girish C Melkani
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- UAB Nathan Shock Center, 1300 University Boulevard Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
8
|
Johnson SL, Murray G, Manoogian ENC, Mason L, Allen JD, Berk M, Panda S, Rajgopal NA, Gibson JC, Bower CD, Berle EF, Joyner K, Villanueva R, Michalak EE, Kriegsfeld LJ. A pre-post trial to examine biological mechanisms of the effects of time-restricted eating on symptoms and quality of life in bipolar disorder. BMC Psychiatry 2024; 24:711. [PMID: 39434066 PMCID: PMC11492775 DOI: 10.1186/s12888-024-06157-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND The primary objective of this trial is to examine the mechanisms of time-restricted eating (TRE) as an adjunct to psychiatric care for people with bipolar disorder (BD) with sleep or circadian disruptions. This study builds on prior studies of circadian disruption in BD as well as growing evidence that TRE improves circadian functioning. METHODS One-hundred fifty participants diagnosed with BD 1 or II will be recruited via advertising in the local community. Main inclusion criteria include: obtaining medical treatment for BD; current sleep or circadian problems; self-reported eating period of ≥ 12 h; no eating disorder or other health conditions that would hinder or limit the safety of following TRE; and not currently experiencing a mood episode, acute suicidality, psychosis, alcohol or substance use disorder. Participants will be asked to complete a baseline period in which daily food intake is logged online for two weeks. After baseline, participants will be asked to follow TRE for 8 weeks and to continue to complete daily food logging during this time. Symptom severity interviews will be conducted by phone or videoconference at baseline, mid-intervention (6 weeks post-baseline), end of intervention (10 weeks post-baseline), and 6 months post-baseline. Self-rated symptom severity and quality of life data will be gathered online at the same time points as symptom severity interviews, and at 16 weeks post-baseline (6 weeks after the TRE period ends). To assess potential mechanisms of change, we will examine the change in diurnal amplitude of 'clock' gene expression as a primary mediator at 8 weeks compared to baseline. We will further test whether diurnal amplitude of clock gene expression is predictive above and beyond the role of two covariate potential mediators, glucose tolerance and inflammation at 8 weeks relative to baseline. To provide an index of whether TRE successfully decreases emotional lability, participants will be asked to complete 5 mood assessments per day for 7 days at baseline and at 10 weeks. These mood assessments will be optional. DISCUSSION The planned research will provide novel and important information on whether TRE improves sleep/circadian rhythm problems, along with reductions in mood symptoms and improvements in quality of life, for individuals with BD. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT06555406.
Collapse
Affiliation(s)
- Sheri L Johnson
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Greg Murray
- Centre for Mental Health, Swinburne University, Melbourne, VIC, 3122, Australia
| | | | - Liam Mason
- Research Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - J D Allen
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Michael Berk
- IMPACT Institute, School of Medicine, Deakin University, Geelong, VIC, Australia
| | | | | | - Jake C Gibson
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Carter D Bower
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Eline F Berle
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Keanan Joyner
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Robert Villanueva
- Department of Psychology, University of California, Berkeley, CA, USA
| | - Erin E Michalak
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Lance J Kriegsfeld
- Department of Psychology, University of California, Berkeley, CA, USA.
- Department of Neuroscience, University of California, Berkeley, CA, USA.
| |
Collapse
|
9
|
Ubaldo-Reyes LM, Espitia-Bautista E, Barajas-Martínez A, Martínez-Tapia R, Rodríguez-Mata V, Noriega-Navarro R, Escalona R, Castillo-Hernández J, Pérez-Torres A, Navarro L. High-Fat Diet-Induced Blood-Brain Barrier Dysfunction: Impact on Allodynia and Motor Coordination in Rats. Int J Mol Sci 2024; 25:11218. [PMID: 39457000 PMCID: PMC11508281 DOI: 10.3390/ijms252011218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The associations among increased pain sensitivity, obesity, and systemic inflammation have not been described as related to BBB dysfunctions. To analyze the metabolic, behavioral, and inflammatory effects of a high-fat diet (HFD) and ultrastructural modifications in brain regions, we used an in vivo experimental model. Adult male Wistar rats were randomly assigned to one of two conditions, an ad libitum control group or an HFD (60%)-fed group, for eight weeks. At the end of the protocol, glucose and insulin tolerance tests were performed. Additionally, we analyzed the response to a normally innocuous mechanical stimulus and changes in motor coordination. At the end of the protocol, HFD-fed rats presented increased HOMA-IR and metabolic syndrome (MetS) prevalence. HFD-fed rats also developed an increased nociceptive response to mechanical stimuli and neurological injury, resulting in impaired motor function. Hypothalamus and cerebellum neurons from HFD-fed rats presented with nuclear swelling, an absence of nucleoli, and karyolysis. These results reveal that HFD consumption affects vital brain structures such as the cerebellum, hippocampus, and hypothalamus. This, in turn, could be producing neuronal damage, impairing cellular communication, and consequently altering motricity and pain sensitivity. Although direct evidence of a causal link between BBB dysfunction and sensory-motor changes was not observed, understanding the association uncovered in this study could lead to targeted therapeutic strategies.
Collapse
Affiliation(s)
- Laura M. Ubaldo-Reyes
- Department of Anatomy, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Estefania Espitia-Bautista
- Laboratory of Molecular Neurophysiology, National Institute of Psychiatry Ramón de la Fuente, Mexico City 14370, Mexico;
| | - Antonio Barajas-Martínez
- Center for Complexity Science, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Ricardo Martínez-Tapia
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| | - Verónica Rodríguez-Mata
- Department of Histology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (V.R.-M.); (A.P.-T.)
| | - Roxana Noriega-Navarro
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| | - Rene Escalona
- Laboratory of Embryology and Genetics, Departamento de Embriología y Genética, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Jesús Castillo-Hernández
- Multidisciplinary Academic Unit Middle Zone, Autonomous University of San Luis Potosí, San Luis Potosí 79615, Mexico;
| | - Armando Pérez-Torres
- Department of Histology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (V.R.-M.); (A.P.-T.)
| | - Luz Navarro
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| |
Collapse
|
10
|
Klingbeil EA, Schade R, Lee SH, Kirkland R, de La Serre CB. Manipulation of feeding patterns in high fat diet fed rats improves microbiota composition dynamics, inflammation and gut-brain signaling. Physiol Behav 2024; 285:114643. [PMID: 39059597 DOI: 10.1016/j.physbeh.2024.114643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Chronic consumption of high fat (HF) diets has been shown to increase meal size and meal frequency in rodents, resulting in overeating. Reducing meal frequency and establishing periods of fasting, independently of caloric intake, may improve obesity-associated metabolic disorders. Additionally, diet-driven changes in microbiota composition have been shown to play a critical role in the development and maintenance of metabolic disorders. In this study, we used a pair-feeding paradigm to reduce meal frequency and snacking episodes while maintaining overall intake and body weight in HF fed rats. We hypothesized that manipulation of feeding patterns would improve microbiota composition and metabolic outcomes. Male Wistar rats were placed in three groups consuming either a HF, low fat diet (LF, matched for sugar), or pair-fed HF diet for 7 weeks (n = 11-12/group). Pair-fed animals received the same amount of food consumed by the HF fed group once daily before dark onset (HF-PF). Rats underwent oral glucose tolerance and gut peptide cholecystokinin sensitivity tests. Bacterial DNA was extracted from the feces collected during both dark and light cycles and sequenced via Illumina MiSeq sequencing of the 16S V4 region. Our pair-feeding paradigm reduced meal numbers, especially small meals in the inactive phase, without changing total caloric intake. This shift in feeding patterns reduced relative abundances of obesity-associated bacteria and maintained circadian fluctuations in microbial abundances. These changes were associated with improved gastrointestinal (GI) function, reduced inflammation, and improved glucose tolerance and gut to brain signaling. We concluded from these data that targeting snacking may help improve metabolic outcomes, independently of energy content of the diet and hyperphagia.
Collapse
Affiliation(s)
- E A Klingbeil
- Department of Nutritional Sciences, The University of Texas at Austin, United States
| | - R Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, United States
| | - S H Lee
- Department of Food Sciences, Sun Moon University, South Korea
| | - R Kirkland
- Office of Research, University of Georgia, United States
| | - C B de La Serre
- Department of Nutritional Sciences, University of Georgia, United States; Department of Biomedical Sciences, Colorado State University, United States.
| |
Collapse
|
11
|
Malakmahmoudi N, Pisu R, Laconi E, Marongiu F. Dietary Rhythms and MASLD-Related Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:3481. [PMID: 39456575 PMCID: PMC11505995 DOI: 10.3390/cancers16203481] [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: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Dietary rhythms have emerged as a relevant variable in the equation relating nutrition and health. Both experimental and epidemiological studies point to potential beneficial effects of adequate fasting intervals between meals on the evolution of chronic diseases associated with aging. Metabolic dysfunction-associated steatotic liver disease (MASLD) is eminently related to diet and unsurprisingly, diet-based approaches are a mainstay in countering its long-term clinical evolution, including the emergence of hepatocellular carcinoma (HCC). We briefly discuss current evidence linking fasting intervals, MASLD, and HCC and propose a working hypothesis to reconcile some of the apparently conflicting results. This hypothesis relates the beneficial effects of time-restricted eating schedules to the quantity and quality of food, and it is easily amenable to testing.
Collapse
Affiliation(s)
| | | | - Ezio Laconi
- Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy; (N.M.); (R.P.); (F.M.)
| | | |
Collapse
|
12
|
Suyoto PS, Pamungkas NP, de Vries JH, Feskens EJ. Associations between Variability in Between- and Within-Day Dietary Intake with Adiposity and Glucose Homeostasis in Adults: A Systematic Review. Adv Nutr 2024:100310. [PMID: 39389469 DOI: 10.1016/j.advnut.2024.100310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
This systematic review aims to comprehensively evaluate the literature regarding the impact of variations in dietary intake, both between- and within-day, on adiposity and glucose metabolism. We included observational and experimental articles obtained from PubMed, Scopus, Cochrane Library, and gray literature until 9 October, 2023, evaluating the impact of between- or within-day variations in meal, energy, or macronutrient intake on these outcomes. Our focus was on adults aged ≥18 y, spanning both healthy individuals and those with type 2 diabetes mellitus (T2DM). Given the diverse range of exposures, treatments, and outcomes among the selected articles, we chose a qualitative synthesis approach to effectively analyze the data. Eighty articles from 43 observational and 37 experimental studies were included, involving 89,178 participants. Patterns of dietary intake variation were identified and systematically organized into distinct categories based on similarities. Between-day variations in dietary intake consisted of between-day variations in both the quantity consumed and meal timing. Meanwhile, within-day variations encompassed factors such as eating window, meal omission, within-day meal timing, within-day variation in dietary intake quantity, and temporal distribution. Despite mixed results, time-restricted eating was generally associated with lower adiposity. However, limited control for total daily energy intake (TDEI) suggests that the contribution of lower energy intake cannot be conclusively excluded. Conversely, the adverse effect of meal omission on glucose parameters was consistently supported by randomized trials. Interestingly, the results showed that consuming a substantial portion of TDEI in the morning may increase the likelihood of observing improvements in adiposity. Furthermore, inconsistencies in outcomes across articles examining the effects in healthy compared with T2DM populations, or in energy-sufficient compared with deficient individuals, indicate potential condition-specific effects. These findings support the need for further investigation into the effects of between- and within-day variations in dietary intake to better understand their impact on adiposity and glucose homeostasis. This review was registered in PROSPERO as CRD42020214307.
Collapse
Affiliation(s)
- Perdana St Suyoto
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands; Department of Nutrition and Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Indonesia
| | - Nindya P Pamungkas
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - Jeanne Hm de Vries
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - Edith Jm Feskens
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands.
| |
Collapse
|
13
|
Rebello CJ, Zhang D, Anderson JC, Bowman RF, Peeke PM, Greenway FL. From starvation to time-restricted eating: a review of fasting physiology. Int J Obes (Lond) 2024:10.1038/s41366-024-01641-0. [PMID: 39369112 DOI: 10.1038/s41366-024-01641-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024]
Abstract
We have long known that subjects with obesity who fast for several weeks survive. Calculations that assume the brain can only use glucose indicated that all carbohydrate and protein sources would be consumed by the brain within several weeks yet subjects with obesity who fasted for several weeks survived. This anomaly led to the determination of the metabolic role of ketone bodies. Subsequent studies transformed our understanding of ketone bodies and illustrated the value of challenging the norm and adapting theory to evidence. Although prolonged fasting is no longer a treatment for obesity, the early studies of starvation provided valuable insights about macronutrient metabolism and ketone body adaptations that fasting elicits. Intermittent fasting and its variants such as time-restricted eating are fasting models that are far less regimented than starvation and severe calorie restriction; yet they produce metabolic benefits. The mechanisms that produce the metabolic changes that intermittent fasting elicits are relatively unknown. In this article, we review the physiology of starvation, starvation adaptation diets, diet-induced ketosis, and intermittent fasting. Understanding the premise and physiology that these regimens induce is necessary to draw parallels and provoke thoughts on the mechanisms underlying the metabolic benefits of intermittent fasting and its variants.
Collapse
Affiliation(s)
- Candida J Rebello
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Dachuan Zhang
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Joseph C Anderson
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | | | - Frank L Greenway
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
14
|
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
|
15
|
Wang L, Wang Q, Wang X, Yang C, Wang X, Liu H, Wang H. Intermittent fasting alleviates postoperative cognitive dysfunction by reducing neuroinflammation in aged mice. Brain Res Bull 2024; 216:111034. [PMID: 39053649 DOI: 10.1016/j.brainresbull.2024.111034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Elderly individuals undergoing surgical procedures are often confronted with the peril of experiencing postoperative cognitive dysfunction (POCD). Prior research has demonstrated the exacerbating effect of sevoflurane anesthesia on neuroinflammation, which can further deteriorate the condition of POCD in elderly patients. Intermittent fasting (IF) restricts food consumption to a specific time window and has been demonstrated to ameliorate cognitive dysfunction induced by neuropathic inflammation. We subjected 18-month-old male mice to 16 hours of fasting and 8 hours of unrestricted eating over a 24-hour period for 0, 1, 2, and 4 weeks, followed by abdominal exploration under sevoflurane anesthesia. In this study, we aim to explore the potential impact of IF on postoperative cognitive function in aged mice undergoing sevoflurane surgery through the preoperative implementation of IF measures. The findings indicate two weeks of IF leads to a significant enhancement of learning and memory capabilities in mice following surgery. The cognitive performance, as determined by the novel object recognition and Morris water maze tests, as well as the synaptic plasticity, as measured by in vivo electrophysiological recordings, has demonstrated marked improvements. Furthermore, the administration of IF markedly enhances the expression of synaptic-associated proteins in hippocampal neurons, concomitant with a decreasing expression of pro-inflammatory factors and a reduced density of microglial cells within the hippocampal brain region. To summarize, the results of this study indicate that IF may mitigate inflammation in the hippocampal area of the brain. Furthermore, IF appears to provide a safeguard against cognitive impairment and synaptic plasticity impairment brought on by sevoflurane anesthesia.
Collapse
Affiliation(s)
- Lei Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Qiang Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xiaoqing Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Chenyi Yang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China
| | - Xinyi Wang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China
| | - Huan Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Haiyun Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China.
| |
Collapse
|
16
|
Ye Z, Huang K, Dai X, Gao D, Gu Y, Qian J, Zhang F, Zhai Q. Light-phase time-restricted feeding disrupts the muscle clock and insulin sensitivity yet potentially induces muscle fiber remodeling in mice. Heliyon 2024; 10:e37475. [PMID: 39328525 PMCID: PMC11425116 DOI: 10.1016/j.heliyon.2024.e37475] [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: 05/08/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Skeletal muscle plays a critical role in regulating systemic metabolic homeostasis. It has been demonstrated that time-restricted feeding (TRF) during the rest phase can desynchronize the suprachiasmatic nucleus (SCN) and peripheral clocks, thereby increasing the risk of metabolic diseases. However, the impact of dietary timing on the muscle clock and health remains poorly understood. Here, through the analysis of cycling genes and differentially expressed genes in the skeletal muscle transcriptome, we identified disruptions in muscle diurnal rhythms by 2 weeks of light-phase TRF. Furthermore, compared with ad libitum (AL) feeding mice, 2 weeks of light-phase TRF was found to induce insulin resistance, muscle fiber type remodeling, and changes in the expression of muscle growth-related genes, while both light-phase and dark-phase TRF having a limited impact on bone quality relative to AL mice. In summary, our research reveals that the disruption of the skeletal muscle clock may contribute to the abnormal metabolic phenotype resulting from feeding restricted to the inactive period. Additionally, our study provides a comprehensive omics atlas of the diurnal rhythms in skeletal muscle regulated by dietary timing.
Collapse
Affiliation(s)
- Zhou Ye
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Kai Huang
- Orthopaedic Institute, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, China
| | - Xueqin Dai
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Dandan Gao
- Wenzhou Medical University, Wenzhou, China
| | - Yue Gu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, China
| | - Jun Qian
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Feng Zhang
- The Joint Innovation Center for Engineering in Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Qiaocheng Zhai
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- The Joint Innovation Center for Engineering in Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Nakajima S, Hanzawa F, Ikeda S, Oda H. Physical inactivity and breakfast skipping caused visceral fat accumulation in rats. Sci Rep 2024; 14:22644. [PMID: 39349485 PMCID: PMC11442645 DOI: 10.1038/s41598-024-68058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/19/2024] [Indexed: 10/02/2024] Open
Abstract
Physical inactivity as well as breakfast skipping is known as risk factor for various metabolic diseases, such as obesity and type 2 diabetes. We have previously reported that a breakfast skipping model, in which the timing of feeding is delayed, induces abnormal lipid metabolism by altering the circadian rhythm of lipid metabolism-related genes in rats. The purpose of this study was to elucidate the synergistic effect of physical inactivity and breakfast skipping on lipid metabolism. We adopted sciatic neurectomized rats as physically inactive models, because we confirmed that the rats mildly decreased their spontaneous locomotor activity compared to sham-operated rats. And then the physically inactive model rats were fed a mild high-fat diet during zeitgeber time (ZT) 12-0 in the control group and ZT16-0 in the breakfast skipping group for 11 days. Body weight gain and total food intake were similar in both groups. Breakfast skipping induced a significant visceral fat accumulation, which was not observed in our previous breakfast skipping or physically inactive studies. The mRNA levels of clock and lipogenesis-related genes were altered by breakfast skipping in the liver and epididymal adipose tissue, and serum insulin level was altered by breakfast skipping. These results suggest that physical inactivity and breakfast skipping synergistically induces drastic visceral fat accumulation due to the alteration of circadian clock and lipid metabolism in the liver and adipose tissue. Therefore, regular feeding timing plays an important role in the health of a sedentary modern society.
Collapse
Affiliation(s)
- Syunsuke Nakajima
- Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8601, Japan
| | - Fumiaki Hanzawa
- Department of Nutritional Science, Nagoya University of Arts and Sciences, Nisshin, 470-0196, Japan
| | - Saiko Ikeda
- Department of Nutritional Science, Nagoya University of Arts and Sciences, Nisshin, 470-0196, Japan
| | - Hiroaki Oda
- Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8601, Japan.
| |
Collapse
|
18
|
Kędzierska-Kapuza K, Łopuszyńska I, Niewiński G, Franek E, Szczuko M. The Influence of Non-Pharmacological and Pharmacological Interventions on the Course of Autosomal Dominant Polycystic Kidney Disease. Nutrients 2024; 16:3216. [PMID: 39339816 PMCID: PMC11434835 DOI: 10.3390/nu16183216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Polycystic kidney disease (PKD) includes autosomal dominant (ADPKD) and autosomal recessive (ARPKD) forms, both of which are primary genetic causes of kidney disease in adults and children. ADPKD is the most common hereditary kidney disease, with a prevalence of 329 cases per million in Europe. This condition accounts for 5-15% of end-stage chronic kidney disease (ESKD) cases, and in developed countries such as Poland, 8-10% of all dialysis patients have ESKD due to ADPKD. The disease is caused by mutations in the PKD1 and PKD2 genes, with PKD1 mutations responsible for 85% of cases, leading to a more aggressive disease course. Recent research suggests that ADPKD involves a metabolic defect contributing to cystic epithelial proliferation and cyst growth. Aim: This review explores the interplay between metabolism, obesity, and ADPKD, discussing dietary and pharmacological strategies that target these metabolic abnormalities to slow disease progression. Conclusion: Metabolic reprogramming therapies, including GLP-1 analogs and dual agonists of GIP/GLP-1 or glucagon/GLP-1 receptors, show promise, though further research is needed to understand their potential in ADPKD treatment fully.
Collapse
Affiliation(s)
- Karolina Kędzierska-Kapuza
- Department of Internal Diseases, Endocrinology and Diabetology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Inga Łopuszyńska
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Grzegorz Niewiński
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Edward Franek
- Department of Internal Diseases, Endocrinology and Diabetology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Małgorzata Szczuko
- Department of Human Nutrition and Metabolomic, Pomeranian Medical University, 24 W. Broniewskiego St., 71-460 Szczecin, Poland
| |
Collapse
|
19
|
Yang L, Wang XZ, Wang CZ, Wang DH, Wang ZS, Zhang XY. Time-restricted feeding modulates gene expression related with rhythm and inflammation in Mongolian gerbils. Comp Biochem Physiol C Toxicol Pharmacol 2024; 287:110038. [PMID: 39260783 DOI: 10.1016/j.cbpc.2024.110038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/11/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
Time-restricted feeding (TRF) has the potential to modulate circadian rhythm and widely studied in humans and laboratory mice. However, less is known about the physiological responses to TRF in wild mammals. Here, we used Mongolian gerbils, Meriones unguiculatus, to explore the effect of 6-week TRF on gene expression related with circadian rhythm and inflammation. The TRF gerbils had higher cumulative food intake than the ad libitum (AL) group, but body mass, feeding frequency/time and metabolic rate did not differ between groups. In the hypothalamus, downregulation of rhythm-related genes Per3, Cry1 and Dbp was detected in the daytime-restricted feeding (DRF) group and Cry1 was downregulated in the nighttime-restricted feeding (NRF) group. In the liver, the expression of Per1/3, Rev-erbα/β and Dbp was lower, and Bmal1 was higher in the DRF than in AL group, while NRF gerbils showed no changes. In the colon, the expression of Bmal1 and Cry1 was higher but Per3, Rev-erbα/β and Dbp were lower in the DRF than in AL group. Further, the expression of inflammation-related genes such as NF-κB, IL-1β, IL-18 and Nlrp3 was lower in the liver of DRF gerbils, and IL-1β was lower both in the hypothalamus and liver of NRF gerbils. Moreover, the genes related with inflammation such as NF-κB, Nlrp3, IL-10/18/1β and Tnf-α were positively or negatively correlated with multiple rhythm-related genes in the central and peripheral organs. In conclusion, TRF, particularly DRF, could modulate rhythm-related genes in the central and peripheral tissues and reduce hepatic expression of inflammation-related genes in gerbils.
Collapse
Affiliation(s)
- Lin Yang
- School of Life Sciences, Hebei University, Baoding 071002, China; State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xi-Zhi Wang
- School of Life Sciences, Hebei University, Baoding 071002, China; State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chen-Zhu Wang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - De-Hua Wang
- School of Life Sciences, Shandong University, Qingdao 266237, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen-Shan Wang
- School of Life Sciences, Hebei University, Baoding 071002, China.
| | - Xue-Ying Zhang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
20
|
Chen Y, Jing Y, Hu L, Xi Z, Lu Z, Loor JJ, Wang M. Overexpression of PER2 Promotes De Novo Fatty Acid Synthesis, Fatty Acid Desaturation, and Triglyceride Accumulation in Bovine Mammary Epithelial Cells. Int J Mol Sci 2024; 25:9785. [PMID: 39337271 PMCID: PMC11431620 DOI: 10.3390/ijms25189785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
The core clock gene Period2 (PER2) is associated with mammary gland development and lipid synthesis in rodents and has recently been found to have a diurnal variation in the process of lactation, but has not yet been demonstrated in bovine mammary epithelial cells (BMECs). To explore the regulatory function of PER2 on milk fat synthesis in bovine mammary epithelial cells, we initially assessed the expression of clock genes and milk fat metabolism genes for 24 h using real-time quantitative PCR and fitted the data to a cosine function curve. Subsequently, we overexpressed the PER2 in BMECs using plasmid vector (pcDNA3.1-PER2), with empty vector pcDNA3.1-myc as the control. After transfecting BMECs for 48 h, we assessed the protein abundance related to milk fat synthesis by Western blot, the expression of genes coding for these proteins using real time-quantitative PCR, the production of triacylglycerol, and the fatty acid profile. The findings indicated that a total of nine clock genes (PER1/2, CRY1/2, REV-ERBα, BMAL1, NCOR1, NR2F2, FBXW11), seven fatty acid metabolism genes (CD36, ACSS2, ACACA, SCD, FADS1, DGAT1, ADFP), and six nuclear receptor-related genes (INSIG1, SCAP, SREBF1, C/EBP, PPARG, LXR) exhibited oscillation with a period close to 24 h in non-transfected BMECs (R2 ≥ 0.7). Compared to the control group (transfected with empty pcDNA3.1-myc), the triglyceride content significantly increased in the PER2 overexpression group (p < 0.05). The lipogenic genes for fatty acid transport and triglyceride synthesis (ACACA, SCD, LPIN1, DGAT1, and SREBF1) were upregulated after PER2 overexpression, along with the upregulation of related protein abundance (p < 0.05). The contents and ratios of palmitic acid (C16:0), oleic acid (C18:1n9c), and trans-oleic acid (C18:1n9t) were significantly increased in the overexpression group (p < 0.05). Overall, the data supported that PER2 participated in the process of milk fat metabolism and is potentially involved in the de novo synthesis and desaturation of fatty acid in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Yifei Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yujia Jing
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Liangyu Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Zanna Xi
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhiqi Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Juan J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Mengzhi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
21
|
Morris MJ, Hasebe K, Shinde AL, Leong MKH, Billah MM, Hesam-Shariati S, Kendig MD. Time-restricted feeding does not prevent adverse effects of palatable cafeteria diet on adiposity, cognition and gut microbiota in rats. J Nutr Biochem 2024; 134:109761. [PMID: 39251144 DOI: 10.1016/j.jnutbio.2024.109761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Time-restricted feeding (TRF) is a popular dietary strategy whereby daily food intake is limited to a <12h window. As little is known about the effects of TRF on cognitive and behavioral measures, the present study examined the effects of time-restricted (8h/day; zeitgeber time [ZT]12-20) or continuous access to a high-fat, high-sugar cafeteria-style diet (Caf; Caf and Caf-TRF groups; n=12 adult male Sprague-Dawley rats) or standard chow (Chow and Chow-TRF groups) on short-term memory, anxiety-like behavior, adiposity and gut microbiota composition over 13-weeks with daily food intake measures. TRF significantly reduced daily energy intake in Caf- but not chow-fed groups. In Caf-fed groups, TRF reduced the proportion of energy derived from sugar while increasing that derived from protein. Caf diet significantly increased weight gain, adiposity and fasting glucose within 4 weeks; TRF partially reduced these effects. Caf diet increased anxiety-like behavior in the Elevated Plus Maze in week 3 but not week 12, and impaired hippocampal-dependent place recognition memory in week 11; neither measure was affected by TRF. Global microbiota composition differed markedly between chow and Caf groups, with a small effect of TRF in rats fed chow. In both chow and Caf diet groups, TRF reduced microbiota alpha diversity measures of Shannon diversity and evenness relative to continuous access. Results indicate only limited benefits of TRF access to an obesogenic diet under these conditions, suggesting that more severe time restriction may be required to offset adverse metabolic and cognitive effects when using highly palatable diets.
Collapse
Affiliation(s)
| | - Kyoko Hasebe
- School of Biomedical Sciences, UNSW Sydney, Kensington, Australia
| | - Arya L Shinde
- School of Biomedical Sciences, UNSW Sydney, Kensington, Australia
| | | | | | | | - Michael D Kendig
- School of Biomedical Sciences, UNSW Sydney, Kensington, Australia; School of Life Sciences, University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
22
|
Zambrano C, González E, Salmeron D, Ruiz-Ojeda FJ, Luján J, Scheer FA, Garaulet M. Time-restricted eating affects human adipose tissue fat mobilization. Obesity (Silver Spring) 2024; 32:1680-1688. [PMID: 39073251 PMCID: PMC11357894 DOI: 10.1002/oby.24057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/11/2024] [Accepted: 04/16/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Time-restricted eating (TRE), a dietary approach that confines food intake to specific time windows, has shown metabolic benefits. However, its impact on body weight loss remains inconclusive. The objective of this study was to investigate the influence of early TRE (eTRE) and delayed TRE (dTRE) on fat mobilization using human adipose tissue (AT) cultures. METHODS Subcutaneous AT was collected from 21 participants with severe obesity. We assessed fat mobilization by measuring glycerol release in AT culture across four treatment conditions: control, eTRE, dTRE, and 24-h fasting. RESULTS TRE had a significant impact on lipolysis (glycerol release [mean (SD)] in micromoles per hour per gram: control, 0.05 [0.003]; eTRE, 0.10 [0.006]; dTRE, 0.08 [0.005]; and fasting, 0.17 [0.008]; p < 0.0001). Both eTRE and dTRE increased lipolysis compared with the control group, with eTRE showing higher glycerol mobilization than dTRE during the overall 24-h time window, especially at the nighttime/habitual sleep episode (p < 0.0001). Further analysis of TRE based on fasting duration revealed that, independently of the time window, glycerol release increased with fasting duration (in micromoles per hour per gram: 8 h = 0.08 [0.001]; 12 h = 0.09 [0.008]; and 16 h of fasting = 0.12 [0.011]; p < 0.0001). CONCLUSIONS This study provides insights into the potential benefits of TRE on fat mobilization and may guide the design of future dietary strategies for weight management and metabolic health.
Collapse
Affiliation(s)
- Carolina Zambrano
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, 30100 Murcia, Spain
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital 30120, Murcia, Spain
| | - Elena González
- Department of Nutrition and integrative physiology, University of Utah, Salt Lake City, Utah, United States
| | - Diego Salmeron
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital 30120, Murcia, Spain
- Health and Social Sciences Department, University of Murcia, Murcia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Francisco Javier Ruiz-Ojeda
- Institute of Nutrition and Food Technology “José Mataix,” Center of Biomedical Research, University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - Juan Luján
- General Surgery Service, Hospital Quiron salud Murcia, Spain
| | - Frank A.J.L Scheer
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (M.I.T.) and Harvard, Cambridge, MA, United States
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Marta Garaulet
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, 30100 Murcia, Spain
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital 30120, Murcia, Spain
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
23
|
Chaix A, Lin T, Ramms B, Cutler RG, Le T, Lopez C, Miu P, Pinto AFM, Saghatelian A, Playford MP, Mehta NN, Mattson MP, Gordts P, Witztum JL, Panda S. Time-Restricted Feeding Reduces Atherosclerosis in LDLR KO Mice but Not in ApoE Knockout Mice. Arterioscler Thromb Vasc Biol 2024; 44:2069-2087. [PMID: 39087348 PMCID: PMC11409897 DOI: 10.1161/atvbaha.124.320998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Dyslipidemia increases cardiovascular disease risk, the leading cause of death worldwide. Under time-restricted feeding (TRF), wherein food intake is restricted to a consistent window of <12 hours, weight gain, glucose intolerance, inflammation, dyslipidemia, and hypercholesterolemia are all reduced in mice fed an obesogenic diet. LDLR (low-density lipoprotein receptor) mutations are a major cause of familial hypercholesterolemia and early-onset cardiovascular disease. METHODS We subjected benchmark preclinical models, mice lacking LDLR-knockout or ApoE knockout to ad libitum feeding of an isocaloric atherogenic diet either ad libitum or 9 hours TRF for up to 13 weeks and assessed disease development, mechanism, and global changes in hepatic gene expression and plasma lipids. In a regression model, a subset of LDLR-knockout mice were ad libitum fed and then subject to TRF. RESULTS TRF could significantly attenuate weight gain, hypercholesterolemia, and atherosclerosis in mice lacking the LDLR-knockout mice under experimental conditions of both prevention and regression. In LDLR-knockout mice, increased hepatic expression of genes mediating β-oxidation during fasting is associated with reduced VLDL (very-low-density lipoprotein) secretion and lipid accumulation. Additionally, increased sterol catabolism coupled with fecal loss of cholesterol and bile acids contributes to the atheroprotective effect of TRF. Finally, TRF alone or combined with a cholesterol-free diet can reduce atherosclerosis in LDLR-knockout mice. However, mice lacking ApoE, which is an important protein for hepatic lipoprotein reuptake do not respond to TRF. CONCLUSIONS In a preclinical animal model, TRF is effective in both the prevention and regression of atherosclerosis in LDLR knockout mice. The results suggest TRF alone or in combination with a low-cholesterol diet can be a lifestyle intervention for reducing cardiovascular disease risk in humans.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Terry Lin
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Bastian Ramms
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Roy G. Cutler
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. 21224
| | - Tiffani Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Catherine Lopez
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Phuong Miu
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Antonio F. M. Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States. 21205
| | - Philip Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Joseph L. Witztum
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Lead contact
| |
Collapse
|
24
|
Marko DM, Conn MO, Schertzer JD. Intermittent fasting influences immunity and metabolism. Trends Endocrinol Metab 2024; 35:821-833. [PMID: 38719726 DOI: 10.1016/j.tem.2024.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 09/12/2024]
Abstract
Intermittent fasting (IF) modifies cell- and tissue-specific immunometabolic responses that dictate metabolic flexibility and inflammation during obesity and type 2 diabetes (T2D). Fasting forces periods of metabolic flexibility and necessitates increased use of different substrates. IF can lower metabolic inflammation and improve glucose metabolism without lowering obesity and can influence time-dependent, compartmentalized changes in immunity. Liver, adipose tissue, skeletal muscle, and immune cells communicate to relay metabolic and immune signals during fasting. Here we review the connections between metabolic and immune cells to explain the divergent effects of IF compared with classic caloric restriction (CR) strategies. We also explore how the immunometabolism of metabolic diseases dictates certain IF outcomes, where the gut microbiota triggers changes in immunity and metabolism during fasting.
Collapse
Affiliation(s)
- Daniel M Marko
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Meghan O Conn
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
25
|
Sukkriang N, Buranapin S. Effect of intermittent fasting 16:8 and 14:10 compared with control-group on weight reduction and metabolic outcomes in obesity with type 2 diabetes patients: A randomized controlled trial. J Diabetes Investig 2024; 15:1297-1305. [PMID: 38932663 PMCID: PMC11363092 DOI: 10.1111/jdi.14186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/08/2024] [Accepted: 03/04/2024] [Indexed: 06/28/2024] Open
Abstract
AIMS/INTRODUCTION To compare the percent weight change and metabolic outcomes among diabetic participants with obesity on intermittent fasting (IF) 16:8, IF 14:10, or normal controlled diets. MATERIALS AND METHODS A randomized controlled trial was conducted to randomize participants into three groups. Each group followed IF 16:8, IF 14:10, according to the protocol 3 days/week for 3 months or a control group. RESULTS A total of 99 participants completed the study. The percentage weight change from baseline was -4.02% (95% CI, -4.40 to -3.64) in IF 16:8, -3.15% (95% CI, -3.41 to -2.89) in IF 14:10, and -0.55% (95% CI, -1.05 to -0.05) in the control group. The percentage weight loss from baseline was significantly more in both IF groups (P < 0.001, both) when compared with the control group. Weight loss was significantly more in the IF 16:8 group than in that of the IF 14:10 group (P < 0.001). Metabolic outcomes (decrease in FBS and HbA1C, and improvement in lipid profiles) were significantly improved from baseline in both IF groups in comparison with the control group. CONCLUSIONS Either IF 16:8 or 14:10 had a benefit in the percentage weight change, glucose and lipid profiles in obese diabetic patients compared with the control group when consumed for 3 days a week for 3 months.
Collapse
Affiliation(s)
| | - Supawan Buranapin
- Division of Endocrinology, Department of Internal Medicine, Faculty of MedicineChiang Mai UniversityChiang MaiThailand
| |
Collapse
|
26
|
Surugiu R, Iancu MA, Vintilescu ȘB, Stepan MD, Burdusel D, Genunche-Dumitrescu AV, Dogaru CA, Dumitra GG. Molecular Mechanisms of Healthy Aging: The Role of Caloric Restriction, Intermittent Fasting, Mediterranean Diet, and Ketogenic Diet-A Scoping Review. Nutrients 2024; 16:2878. [PMID: 39275194 PMCID: PMC11397047 DOI: 10.3390/nu16172878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
As the population ages, promoting healthy aging through targeted interventions becomes increasingly crucial. Growing evidence suggests that dietary interventions can significantly impact this process by modulating fundamental molecular pathways. This review focuses on the potential of targeted dietary strategies in promoting healthy aging and the mechanisms by which specific nutrients and dietary patterns influence key pathways involved in cellular repair, inflammation, and metabolic regulation. Caloric restriction, intermittent fasting, the Mediterranean diet, as well as the ketogenic diet showed promising effects on promoting healthy aging, possibly by modulating mTORC1 AMPK, an insulin signaling pathway. By understanding the intricate interplay between diet and molecular pathways, we can develop personalized dietary strategies that not only prevent age-related diseases, but also promote overall health and well-being throughout the aging process.
Collapse
Affiliation(s)
- Roxana Surugiu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania
| | - Mihaela Adela Iancu
- Department of Internal Medicine (Cardiology, Gastroenterology, Hepatology, Rheumatology, Geriatrics), Family Medicine, Labor Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ștefănița Bianca Vintilescu
- Department of Infant Care-Pediatrics-Neonatology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania
| | - Mioara Desdemona Stepan
- Department of Infant Care-Pediatrics-Neonatology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania
| | - Daiana Burdusel
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania
| | | | - Carmen-Adriana Dogaru
- Department of Family Medicine, University of Medicine and Pharmacy of Craiova, St. Petru Rareș, No. 2-4, 200349 Craiova, Romania
| | - Gheorghe Gindrovel Dumitra
- Department of Family Medicine, University of Medicine and Pharmacy of Craiova, St. Petru Rareș, No. 2-4, 200349 Craiova, Romania
| |
Collapse
|
27
|
Acosta-Rodríguez VA, Rijo-Ferreira F, van Rosmalen L, Izumo M, Park N, Joseph C, Hepler C, Thorne AK, Stubblefield J, Bass J, Green CB, Takahashi JS. Misaligned feeding uncouples daily rhythms within brown adipose tissue and between peripheral clocks. Cell Rep 2024; 43:114523. [PMID: 39046875 DOI: 10.1016/j.celrep.2024.114523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
Extended food consumption during the rest period perturbs the phase relationship between circadian clocks in the periphery and the brain, leading to adverse health effects. Beyond the liver, how metabolic organs respond to a timed hypocaloric diet is largely unexplored. We investigated how feeding schedules impacted circadian gene expression in epididymal white and brown adipose tissue (eWAT and BAT) compared to the liver and hypothalamus. We restricted food to either daytime or nighttime in C57BL/6J male mice, with or without caloric restriction. Unlike the liver and eWAT, rhythmic clock genes in the BAT remained insensitive to feeding time, similar to the hypothalamus. We uncovered an internal split within the BAT in response to conflicting environmental cues, displaying inverted oscillations on a subset of metabolic genes without modifying its local core circadian machinery. Integrating tissue-specific responses on circadian transcriptional networks with metabolic outcomes may help elucidate the mechanism underlying the health burden of eating at unusual times.
Collapse
Affiliation(s)
- Victoria A Acosta-Rodríguez
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Berkeley Public Health, Molecular Cell Biology Department, University of California, Berkeley, Berkeley, CA, USA
| | - Laura van Rosmalen
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mariko Izumo
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Noheon Park
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Chryshanthi Joseph
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Chelsea Hepler
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anneke K Thorne
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeremy Stubblefield
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Benedictine College, Atchison, KS, USA
| | - Joseph Bass
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Carla B Green
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
28
|
Zou M, Johnson L, Leary S, Ibacache Fuentes F, Northstone K. Later eating rhythm measured in children at 7 years of age in the ALSPAC cohort. Wellcome Open Res 2024; 9:77. [PMID: 39280725 PMCID: PMC11393536 DOI: 10.12688/wellcomeopenres.20605.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Later eating rhythm (LER) refers to later timing, greater energy intake (EI), and higher frequency of eating occasions (meal/snack) in the evening. The significance of LER in child health is becoming increasingly recognised. However, the lack of consensus regarding definitions of LER make it challenging to fully comprehend its role. This data note describes LER variables derived in the Avon Longitudinal Study of Parents and Children (ALSPAC), an ongoing birth cohort which enrolled 14,541 pregnant women living in Avon, UK, with an expected date of delivery between April 1991 - December 1992. When children were 7 years, parents completed a structured 3-day food diary, recording all foods/drinks consumed over 3 days (preferably 1 weekend day and 2 weekdays). Data was available for 7,285 children (50.1% response rate). A subsample of 4,869 children had exact time of eating occasions added to the existing database, which only included broad indications of eating timing based on 2-7 hour long meal slots. 13 LER variables were derived for the entire week and weekdays/weekend days separately. These comprise: 1) eating around individual bedtime (number days); 2) eating around average bedtime (number days); 3) time of evening main meal (hrs:mins); 4) time of last eating occasion (hrs:mins); 5) EI in the evening (percentage of total daily energy intake, %TDEI); 6) EI within 2hrs before bedtime (%TDEI); 7) EI for evening main meal (%TDEI); 8) EI for evening snacks (%TDEI); 9) Night eating1 (NE1): eating over 30% of total daily energy intake after 18:00 (number days); 10) NE2: eating over 25% of total daily energy intake within 2hrs before bedtime (number days); 11) eating frequency after 17:00 (number of eating occasions); 12) regularity of dinner (number of days); 13) frequency of evening snacks (number days). We describe the derivation, prevalence and inter-corelations between LER variables.
Collapse
Affiliation(s)
- Mengxuan Zou
- University of Bristol Medical School, Bristol, England, BS8 2BN, UK
| | - Laura Johnson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England, BS8 2PS, UK
| | - Sam Leary
- Bristol Dental School, University of Bristol, Bristol, England, BS2 0PT, UK
| | | | - Kate Northstone
- Bristol Medical School, University of Bristol, Bristol, England, BS8 2BN, UK
| |
Collapse
|
29
|
Malik DM, Rhoades SD, Zhang SL, Sengupta A, Barber A, Haynes P, Arnadottir ES, Pack A, Kibbey RG, Kain P, Sehgal A, Weljie AM. Glucose Challenge Uncovers Temporal Fungibility of Metabolic Homeostasis over a day:night cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.30.564837. [PMID: 37961230 PMCID: PMC10634956 DOI: 10.1101/2023.10.30.564837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Rhythmicity is a cornerstone of behavioral and biological processes, especially metabolism, yet the mechanisms behind metabolite cycling remain elusive. This study uncovers a robust oscillation in key metabolite pathways downstream of glucose in humans. A purpose-built 13C6-glucose isotope tracing platform was used to sample Drosophila every 4h and probe these pathways, revealing a striking peak in biosynthesis shortly after lights-on in wild-type flies. A hyperactive mutant (fumin) demonstrates increased Krebs cycle labelling and dawn-specific glycolysis labelling. Surprisingly, neither underlying feeding rhythms nor the presence of food availability explain the rhythmicity of glucose processing across genotypes, suggesting a robust internal mechanism for metabolic control of glucose processing. These results align with clinical data highlighting detrimental effects of mistimed energy intake. Our approach offers a unique insight into the dynamic range of daily metabolic processing and provides a mechanistic foundation for exploring circadian metabolic homeostasis in disease contexts.
Collapse
Affiliation(s)
- Dania M. Malik
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- These authors contributed equally
| | - Seth D. Rhoades
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Fulgens Consulting, LLC, Cambridge, Massachusetts 02142, USA
- These authors contributed equally
| | - Shirley L. Zhang
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Annika Barber
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08854, USA
| | - Paula Haynes
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Erna Sif Arnadottir
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Allan Pack
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Richard G. Kibbey
- Department of Internal Medicine, Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Pinky Kain
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
30
|
Xie X, Zhang M, Luo H. Regulation of metabolism by circadian rhythms: Support from time-restricted eating, intestinal microbiota & omics analysis. Life Sci 2024; 351:122814. [PMID: 38857654 DOI: 10.1016/j.lfs.2024.122814] [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: 02/18/2024] [Revised: 05/05/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Circadian oscillatory system plays a key role in coordinating the metabolism of most organisms. Perturbation of genetic effects and misalignment of circadian rhythms result in circadian dysfunction and signs of metabolic disorders. The eating-fasting cycle can act on the peripheral circadian clocks, bypassing the photoperiod. Therefore, time-restricted eating (TRE) can improve metabolic health by adjusting eating rhythms, a process achieved through reprogramming of circadian genomes and metabolic programs at different tissue levels or remodeling of the intestinal microbiota, with omics technology allowing visualization of the regulatory processes. Here, we review recent advances in circadian regulation of metabolism, focus on the potential application of TRE for rescuing circadian dysfunction and metabolic disorders with the contribution of intestinal microbiota in between, and summarize the significance of omics technology.
Collapse
Affiliation(s)
- Ximei Xie
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China
| | - Mengjie Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China.
| |
Collapse
|
31
|
Levkovich G, Shmulevitch R, Almagor D, Reshef L, Shiklov G, Rusal M, Halachmi I, Sagi D. Synchronizing food availability with the natural rhythm substantially improves reproduction and extends healthspan in laying hens. Sci Rep 2024; 14:18780. [PMID: 39138285 PMCID: PMC11322325 DOI: 10.1038/s41598-024-69703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
Disrupted sleep due to nighttime eating can raise various concerns, impacting both physical health and overall well-being. Nevertheless, there is a lack of direct evidence linking nighttime eating with the female reproduction system, possibly due to the absence of suitable models. Here, we use the laying hen, a diurnal animal maintained under ad libitum feeding, as a vertebrate model to quantify the impact of nighttime eating on reproduction and aging. To do this, we have built an Arduino-based setup that regulates food availability and exclusively tracks the eating events of individuals. Our data indicate that synchronizing food availability with the natural sleep-wake rhythm substantially improves reproduction and extends healthspan in hens. With reproductive aging becoming progressively more prevalent in contemporary society due to the trend of delayed childbearing, synchronizing eating with the natural rhythm could help mitigate reproductive aging in vertebrates and potentially in humans as well.
Collapse
Affiliation(s)
- Guy Levkovich
- Department of Poultry and Aquaculture, Institute of Animal Science, Agricultural Research Organization, Volcani Institute, Rishon LeZion, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, The Sagol Center for Healthy Human Longevity, Bar-Ilan University, Ramat Gan, Israel
| | - Ran Shmulevitch
- Lab For Precision Livestock Farming (PLF), Agricultural Engineering Institute, Volcani Institute, Rishon LeZion, Israel
- Industrial Engineering & Management (IEM) Department, Faculty of Engineering Sciences, Ben-Gurion University, Be'er Sheva, Israel
| | - Dana Almagor
- Hebrew University of Jerusalem Koret School of Veterinary Medicine, Rehovot, Israel
| | - Liad Reshef
- Lab For Precision Livestock Farming (PLF), Agricultural Engineering Institute, Volcani Institute, Rishon LeZion, Israel
| | - German Shiklov
- Department of Poultry and Aquaculture, Institute of Animal Science, Agricultural Research Organization, Volcani Institute, Rishon LeZion, Israel
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Mark Rusal
- Department of Poultry and Aquaculture, Institute of Animal Science, Agricultural Research Organization, Volcani Institute, Rishon LeZion, Israel
| | - Ilan Halachmi
- Lab For Precision Livestock Farming (PLF), Agricultural Engineering Institute, Volcani Institute, Rishon LeZion, Israel
| | - Dror Sagi
- Department of Poultry and Aquaculture, Institute of Animal Science, Agricultural Research Organization, Volcani Institute, Rishon LeZion, Israel.
| |
Collapse
|
32
|
Mirasierra M, Fernández-Pérez A, Lizarbe B, Keiran N, Ruiz-Cañas L, Casarejos MJ, Cerdán S, Vendrell J, Fernández-Veledo S, Vallejo M. Alx3 deficiency disrupts energy homeostasis, alters body composition, and impairs hypothalamic regulation of food intake. Cell Mol Life Sci 2024; 81:343. [PMID: 39129011 PMCID: PMC11335267 DOI: 10.1007/s00018-024-05384-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/03/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024]
Abstract
The coordination of food intake, energy storage, and expenditure involves complex interactions between hypothalamic neurons and peripheral tissues including pancreatic islets, adipocytes, muscle, and liver. Previous research shows that deficiency of the transcription factor Alx3 alters pancreatic islet-dependent glucose homeostasis. In this study we carried out a comprehensive assessment of metabolic alterations in Alx3 deficiency. We report that Alx3-deficient mice exhibit decreased food intake without changes in body weight, along with reduced energy expenditure and altered respiratory exchange ratio. Magnetic resonance imaging reveals increased adiposity and decreased muscle mass, which was associated with markers of motor and sympathetic denervation. By contrast, Alx3-deficient mice on a high-fat diet show attenuated weight gain and improved insulin sensitivity, compared to control mice. Gene expression analysis demonstrates altered lipogenic and lipolytic gene profiles. In wild type mice Alx3 is expressed in hypothalamic arcuate nucleus neurons, but not in major peripheral metabolic organs. Functional diffusion-weighted magnetic resonance imaging reveals selective hypothalamic responses to fasting in the arcuate nucleus of Alx3-deficient mice. Additionally, altered expression of proopiomelanocortin and melanocortin-3 receptor mRNA in the hypothalamus suggests impaired regulation of feeding behavior. This study highlights the crucial role for Alx3 in governing food intake, energy homeostasis, and metabolic nutrient partitioning, thereby influencing body mass composition.
Collapse
Affiliation(s)
- Mercedes Mirasierra
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Fernández-Pérez
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Centro para el Desarrollo Tecnológico e Industrial (CDTI), Madrid, Spain
| | - Blanca Lizarbe
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain
| | - Noelia Keiran
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV) - Hospital Universitari de Tarragona Joan XXIII, Universitat Rovira i Virgili, Tarragona, Spain
| | - Laura Ruiz-Cañas
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
- Chronic Diseases and Cancer Area 3, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain
| | - María José Casarejos
- Neuropharmacology Laboratory, Neurobiology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain
| | - Joan Vendrell
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV) - Hospital Universitari de Tarragona Joan XXIII, Universitat Rovira i Virgili, Tarragona, Spain
| | - Sonia Fernández-Veledo
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV) - Hospital Universitari de Tarragona Joan XXIII, Universitat Rovira i Virgili, Tarragona, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
33
|
Ribas-Latre A, Fernández-Veledo S, Vendrell J. Time-restricted eating, the clock ticking behind the scenes. Front Pharmacol 2024; 15:1428601. [PMID: 39175542 PMCID: PMC11338815 DOI: 10.3389/fphar.2024.1428601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Maintaining metabolic balance relies on accumulating nutrients during feeding periods and their subsequent release during fasting. In obesity and metabolic disorders, strategies aimed at reducing food intake while simulating fasting have garnered significant attention for weight loss. Caloric restriction (CR) diets and intermittent fasting (IF) interventions have emerged as effective approaches to improving cardiometabolic health. Although the comparative metabolic benefits of CR versus IF remain inconclusive, this review focuses on various forms of IF, particularly time-restricted eating (TRE). Methods This study employs a narrative review methodology, systematically collecting, synthesizing, and interpreting the existing literature on TRE and its metabolic effects. A comprehensive and unbiased search of relevant databases was conducted to identify pertinent studies, including pre-clinical animal studies and clinical trials in humans. Keywords such as "Obesity," "Intermittent Fasting," "Time-restricted eating," "Chronotype," and "Circadian rhythms" guided the search. The selected studies were critically appraised based on predefined inclusion and exclusion criteria, allowing for a thorough exploration and synthesis of current knowledge. Results This article synthesizes pre-clinical and clinical studies on TRE and its metabolic effects, providing a comprehensive overview of the current knowledge and identifying gaps for future research. It explores the metabolic outcomes of recent clinical trials employing different TRE protocols in individuals with overweight, obesity, or type II diabetes, emphasizing the significance of individual chronotype, which is often overlooked in practice. In contrast to human studies, animal models underscore the role of the circadian clock in mitigating metabolic disturbances induced by obesity through time-restricted feeding (TRF) interventions. Consequently, we examine pre-clinical evidence supporting the interplay between the circadian clock and TRF interventions. Additionally, we provide insights into the role of the microbiota, which TRE can modulate and its influence on circadian rhythms.
Collapse
Affiliation(s)
- Aleix Ribas-Latre
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joan Vendrell
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| |
Collapse
|
34
|
Dell’Angelica D, Singh K, Colwell CS, Ghiani CA. Circadian Interventions in Preclinical Models of Huntington's Disease: A Narrative Review. Biomedicines 2024; 12:1777. [PMID: 39200241 PMCID: PMC11351982 DOI: 10.3390/biomedicines12081777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 09/02/2024] Open
Abstract
Huntington's Disease (HD) is a neurodegenerative disorder caused by an autosomal-dominant mutation in the huntingtin gene, which manifests with a triad of motor, cognitive and psychiatric declines. Individuals with HD often present with disturbed sleep/wake cycles, but it is still debated whether altered circadian rhythms are intrinsic to its aetiopathology or a consequence. Conversely, it is well established that sleep/wake disturbances, perhaps acting in concert with other pathophysiological mechanisms, worsen the impact of the disease on cognitive and motor functions and are a burden to the patients and their caretakers. Currently, there is no cure to stop the progression of HD, however, preclinical research is providing cementing evidence that restoring the fluctuation of the circadian rhythms can assist in delaying the onset and slowing progression of HD. Here we highlight the application of circadian-based interventions in preclinical models and provide insights into their potential translation in clinical practice. Interventions aimed at improving sleep/wake cycles' synchronization have shown to improve motor and cognitive deficits in HD models. Therefore, a strong support for their suitability to ameliorate HD symptoms in humans emerges from the literature, albeit with gaps in our knowledge on the underlying mechanisms and possible risks associated with their implementation.
Collapse
Affiliation(s)
- Derek Dell’Angelica
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
| | - Karan Singh
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
| | - Christopher S. Colwell
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
| | - Cristina A. Ghiani
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA
| |
Collapse
|
35
|
Tang S, Wu H, Chen Q, Tang T, Li J, An H, Zhu S, Han L, Sun H, Ge J, Qian X, Wang X, Wang Q. Maternal Obesity Induces the Meiotic Defects and Epigenetic Alterations During Fetal Oocyte Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309184. [PMID: 38868907 PMCID: PMC11321662 DOI: 10.1002/advs.202309184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/23/2024] [Indexed: 06/14/2024]
Abstract
It has been widely reported that obesity adversely impacts reproductive performance of females. However, the effects of maternal obesity on fetal germ cells remain poorly understood. In the present study, by employing a high-fat diet (HFD)-based mouse model, it is discovered that maternal obesity disrupts the chromosomal synapsis and homologous recombination during fetal oogenesis. Moreover, transcriptomic profiling reveales the potential molecular network controlling this process. Of note, the global hypermethylation of genomic DNA in fetal oocytes from obese mouse is detected. Importantly, time-restricted feeding (TRF) of obese mice not only ameliorate the meiotic defects, but also partly restore the epigenetic remodeling in fetal oocytes. In sum, the evidence are provided showing the deficit fetal oogenesis in obese mother, implicating a mechanism underlying the intergenerational effects of environmental insults. TRF may represent a potentially effective approach for mitigating fertility issues in obese patients.
Collapse
Affiliation(s)
- Shoubin Tang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
- Department of Nutrition and Food HygieneSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Huihua Wu
- Suzhou Municipal HospitalNanjing Medical UniversityNanjing211166China
| | - Qiuzhen Chen
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Tao Tang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Jiashuo Li
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Huiqing An
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Hongzheng Sun
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Xu Qian
- Department of Nutrition and Food HygieneSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| |
Collapse
|
36
|
Farahmand F, Sidikpramana M, Gomez AR, Rivera LJ, Trzeciak JR, Sharif S, Tang Q, Leinninger GM, Güler AD, Steele AD. Dopamine production in neurotensin receptor 1 neurons is required for diet-induced obesity and increased day eating on a high-fat diet. Obesity (Silver Spring) 2024; 32:1448-1452. [PMID: 38979671 PMCID: PMC11269025 DOI: 10.1002/oby.24066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVE This study aimed to determine a dopaminergic circuit required for diet-induced obesity in mice. METHODS We created conditional deletion mutants for tyrosine hydroxylase (TH) using neurotensin receptor 1 (Ntsr1) Cre and other Cre drivers and measured feeding and body weight on standard and high-fat diets. We then used an adeno-associated virus to selectively restore TH to the ventral tegmental area (VTA) Ntsr1 neurons in conditional knockout (cKO) mice. RESULTS Mice with cKO of Th using Vglut2-Cre, Cck-Cre, Calb1-Cre, and Bdnf-Cre were susceptible to obesity on a high-fat diet; however, Ntsr1-Cre Th cKO mice resisted weight gain on a high-fat diet and did not experience an increase in day eating unlike their wild-type littermate controls. Restoration of TH to the VTA Ntsr1 neurons of the Ntsr1-Cre Th cKO mice using an adeno-associated virus resulted in an increase in weight gain and day eating on a high-fat diet. CONCLUSIONS Ntsr1-Cre Th cKO mice failed to increase day eating on a high-fat diet, offering a possible explanation for their resistance to diet-induced obesity. These results implicate VTA Ntsr1 dopamine neurons as promoting out-of-phase feeding behavior on a high-fat diet that could be an important contributor to diet-induced obesity in humans.
Collapse
Affiliation(s)
- Firozeh Farahmand
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| | - Michael Sidikpramana
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| | - Alyssa R. Gomez
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| | - Luis J. Rivera
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| | - Jacqueline R. Trzeciak
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| | - Sarah Sharif
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| | - Qijun Tang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Gina M. Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI 48114, United States
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Andrew D. Steele
- Department of Biological Sciences, California State Polytechnic University Pomona; Pomona, CA; USA
| |
Collapse
|
37
|
McHill AW, Butler MP. Eating Around the Clock: Circadian Rhythms of Eating and Metabolism. Annu Rev Nutr 2024; 44:25-50. [PMID: 38848598 DOI: 10.1146/annurev-nutr-062122-014528] [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: 06/09/2024]
Abstract
The time of day that we eat is increasingly recognized as contributing as importantly to overall health as the amount or quality of the food we eat. The endogenous circadian clock has evolved to promote intake at optimal times when an organism is intended to be awake and active, but electric lights and abundant food allow eating around the clock with deleterious health outcomes. In this review, we highlight literature pertaining to the effects of food timing on health, beginning with animal models and then translation into human experiments. We emphasize the pitfalls and opportunities that technological advances bring in bettering understanding of eating behaviors and their association with health and disease. There is great promise for restricting the timing of food intake both in clinical interventions and in public health campaigns for improving health via nonpharmacological therapies.
Collapse
Affiliation(s)
- Andrew W McHill
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, Portland, Oregon, USA
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Matthew P Butler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA;
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
38
|
Campbell KJ, Jiang P, Olker C, Lin X, Kim SY, Lee CJ, Song EJ, Turek FW, Vitaterna MH. The impacts of sex and the 5xFAD model of Alzheimer's disease on the sleep and spatial learning responses to feeding time. Front Neurol 2024; 15:1430989. [PMID: 39144714 PMCID: PMC11322461 DOI: 10.3389/fneur.2024.1430989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction The relationships between the feeding rhythm, sleep and cognition in Alzheimer's disease (AD) are incompletely understood, but meal time could provide an easy-to-implement method of curtailing disease-associated disruptions in sleep and cognition. Furthermore, known sex differences in AD incidence could relate to sex differences in circadian rhythm/sleep/cognition interactions. Methods The 5xFAD transgenic mouse model of AD and non-transgenic wild-type controls were studied. Both female and male mice were used. Food access was restricted each day to either the 12-h light phase (light-fed groups) or the 12-h dark phase (dark-fed groups). Sleep (electroencephalographic/electromyographic) recording and cognitive behavior measures were collected. Results The 5xFAD genotype reduces NREM and REM as well as the number of sleep spindles. In wild-type mice, light-fed groups had disrupted vigilance state amounts, characteristics, and rhythms relative to dark-fed groups. These feeding time differences were reduced in 5xFAD mice. Sex modulates these effects. 5xFAD mice display poorer spatial memory that, in female mice, is curtailed by dark phase feeding. Similarly, female 5xFAD mice have decreased anxiety-associated behavior. These emotional and cognitive measures are correlated with REM amount. Discussion Our study demonstrates that the timing of feeding can alter many aspects of wake, NREM and REM. Unexpectedly, 5xFAD mice are less sensitive to these feeding time effects. 5xFAD mice demonstrate deficits in cognition which are correlated with REM, suggesting that this circadian-timed aspect of sleep may link feeding time and cognition. Sex plays an important role in regulating the impact of feeding time on sleep and cognition in both wild-type and 5xFAD mice, with females showing a greater cognitive response to feeding time than males.
Collapse
Affiliation(s)
- Katrina J. Campbell
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Christopher Olker
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Xuanyi Lin
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Sarah Y. Kim
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Christopher J. Lee
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Eun Joo Song
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Fred W. Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Martha Hotz Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| |
Collapse
|
39
|
Jin X, Deng Y, Zhang W, Xu X, Rong S. Counting hours or calories? Metabolic regulatory role of time-restricted eating in adults with overweight and obesity: a systematic review and meta-analysis. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39069716 DOI: 10.1080/10408398.2024.2382344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Time-restricted eating (TRE) effectively improves healthspan, including controlling obesity and improving metabolic health. To date, few meta-analyses have been conducted to explore the effects of various protocols of TRE in participants with overweight/obesity. PubMed, Embase and the Cochrane Central Register of Controlled Trials were searched up until October 15, 2022. Randomized and non-randomized clinical trials that investigated the effect of TRE on body weight, body composition and cardiometabolic parameters in participants with overweight/obesity were included. Mean differences of changes from the baseline were used for all analyses between the two groups. Prespecified subgroup analyses based on different protocols of TRE were performed. Twenty-three studies were included in the meta-analysis with 1867 participants. TRE interventions led to significant changes in body weight. When energy restriction strategies were conducted in both the TRE and control groups, the weight-loss effect of TRE remained significant. TRE with 4 ∼ 8h feeding window, morning or late eating strategies, led to reduction in body weight and fat mass for at least 8 wk. Hence TRE is a potential and effective approach for weight loss for participants with overweight/obesity. An 8h-TRE intervention with a morning eating strategy for at least eight weeks might be the optimum TRE intervention mode.
Collapse
Affiliation(s)
- Xin Jin
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Deng
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Wenxue Zhang
- Department of Nutrition, School of Public Health, Wuhan University; Research Center of Public Health, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xintian Xu
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Rong
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
- Department of Nutrition, School of Public Health, Wuhan University; Research Center of Public Health, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
40
|
Van Drunen R, Dai Y, Wei H, Fekry B, Noori S, Shivshankar S, Bravo R, Zhao Z, Yoo SH, Justice N, Wu JQ, Tong Q, Eckel-Mahan K. Cell-specific regulation of the circadian clock by BMAL1 in the paraventricular nucleus: Implications for regulation of systemic biological rhythms. Cell Rep 2024; 43:114380. [PMID: 38935503 PMCID: PMC11446153 DOI: 10.1016/j.celrep.2024.114380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/28/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Circadian rhythms are internal biological rhythms driving temporal tissue-specific, metabolic programs. Loss of the circadian transcription factor BMAL1 in the paraventricular nucleus (PVN) of the hypothalamus reveals its importance in metabolic rhythms, but its functions in individual PVN cells are poorly understood. Here, loss of BMAL1 in the PVN results in arrhythmicity of processes controlling energy balance and alters peripheral diurnal gene expression. BMAL1 chromatin immunoprecipitation sequencing (ChIP-seq) and single-nucleus RNA sequencing (snRNA-seq) reveal its temporal regulation of target genes, including oxytocin (OXT), and restoring circulating OXT peaks in BMAL1-PVN knockout (KO) mice rescues absent activity rhythms. While glutamatergic neurons undergo day/night changes in expression of genes involved in cell morphogenesis, astrocytes and oligodendrocytes show gene expression changes in cytoskeletal organization and oxidative phosphorylation. Collectively, our findings show diurnal gene regulation in neuronal and non-neuronal PVN cells and that BMAL1 contributes to diurnal OXT secretion, which is important for systemic diurnal rhythms.
Collapse
Affiliation(s)
- Rachel Van Drunen
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yulin Dai
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haichao Wei
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Baharan Fekry
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sina Noori
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Samay Shivshankar
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Rafael Bravo
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Seung-Hee Yoo
- MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Biochemistry and Cell Biology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nicholas Justice
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jia Qian Wu
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qingchun Tong
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kristin Eckel-Mahan
- UT Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Huang C, Liu D, Yang S, Huang Y, Wei X, Zhang P, Lin J, Xu B, Liu Y, Guo D, Li Y, Li J, Zhang H. Effect of time-restricted eating regimen on weight loss is mediated by gut microbiome. iScience 2024; 27:110202. [PMID: 38993674 PMCID: PMC11238135 DOI: 10.1016/j.isci.2024.110202] [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: 01/19/2024] [Revised: 04/16/2024] [Accepted: 06/01/2024] [Indexed: 07/13/2024] Open
Abstract
Time-restricted eating (TRE) is a promising obesity management strategy, but weight-loss efficacy varies among participants, and the underlying mechanism is unclear. The study aimed to investigate the role of gut microbiota in weight-loss response during long-term TRE intervention. We analyzed data from 51 obese adults in a 12-month TRE program, categorizing them into distinct weight loss groups (DG) and moderate weight loss groups (MG) based on their TRE responses. Shotgun metagenomic sequencing analysis revealed a significant increase in species closely associated with weight loss effectiveness and metabolic parameter changes in the DG group. Pathways related to fatty acid biosynthesis, glycogen biosynthesis, and nucleotide metabolism were reduced in the DG group and enhanced in the MG group. Next, we identified nine specific species at baseline that contributed better responses to TRE intervention and significant weight loss. Collectively, gut microbiota contributes to responsiveness heterogeneity in TRE and can predict weight-loss effectiveness.
Collapse
Affiliation(s)
- Chensihan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Deying Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shunyu Yang
- Department of Nutrition, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Peizhen Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Bingyan Xu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yating Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030000, Shanxi, China
- Core Laboratory, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University; Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan 030000, Shanxi, China
| | - Jin Li
- Department of Endocrinology and Metabolism, Shan Xi Medical University Second Hospital, Shan Xi Medical University, Taiyuan 030000, Shanxi, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
42
|
Pagidipati NJ, Taub PR, Ostfeld RJ, Kirkpatrick CF. Dietary patterns to promote cardiometabolic health. Nat Rev Cardiol 2024:10.1038/s41569-024-01061-7. [PMID: 39020052 DOI: 10.1038/s41569-024-01061-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 07/19/2024]
Abstract
Multiple professional societies recommend the Mediterranean and/or Dietary Approaches to Stop Hypertension dietary patterns in their cardiovascular disease prevention guidelines because these diets can improve cardiometabolic health and reduce the risk of cardiovascular events. Furthermore, low sodium intake can be particularly beneficial for patients with hypertension. Carbohydrate restriction, with an emphasis on including high-quality carbohydrates and limiting refined starches and foods and beverages with added sugars, can promote weight loss and cardiometabolic benefits in the short term, compared with higher carbohydrate intake. Evidence is lacking for sustained, long-term effects of low carbohydrate and very low carbohydrate intake on cardiometabolic risk and cardiovascular outcomes. Time-restricted eating, in the context of an overall healthy dietary pattern, can promote cardiometabolic health by aligning food intake with the circadian rhythm, although its effect on hard clinical outcomes remains to be proven. Although there is no one dietary pattern that is appropriate for all patients, engaging in shared decision-making with patients, utilizing behaviour-change principles and engaging members of the health-care team, such as registered dietitian nutritionists, can lead to substantial improvement in the lifestyle and overall health trajectory of a patient. Emphasizing the similarities, rather than differences, of recommended dietary patterns, which include an emphasis on vegetables, fruits, legumes, nuts, whole grains and minimally processed protein foods, such as fatty fish or plant-based proteins, can simplify the process for both patients and clinicians alike.
Collapse
Affiliation(s)
- Neha J Pagidipati
- Division of Cardiology, Duke University School of Medicine, Duke Clinical Research Institute, Durham, NC, USA.
| | - Pam R Taub
- Division of Cardiovascular Medicine, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Robert J Ostfeld
- Division of Cardiology, Montefiore Health System, Bronx, NY, USA
| | - Carol F Kirkpatrick
- Midwest Biomedical Research, Addison, IL, USA
- Kasiska Division of Health Sciences, Idaho State University, Pocatello, ID, USA
| |
Collapse
|
43
|
Schrader LA, Ronnekleiv-Kelly SM, Hogenesch JB, Bradfield CA, Malecki KM. Circadian disruption, clock genes, and metabolic health. J Clin Invest 2024; 134:e170998. [PMID: 39007272 PMCID: PMC11245155 DOI: 10.1172/jci170998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
A growing body of research has identified circadian-rhythm disruption as a risk factor for metabolic health. However, the underlying biological basis remains complex, and complete molecular mechanisms are unknown. There is emerging evidence from animal and human research to suggest that the expression of core circadian genes, such as circadian locomotor output cycles kaput gene (CLOCK), brain and muscle ARNT-Like 1 gene (BMAL1), period (PER), and cyptochrome (CRY), and the consequent expression of hundreds of circadian output genes are integral to the regulation of cellular metabolism. These circadian mechanisms represent potential pathophysiological pathways linking circadian disruption to adverse metabolic health outcomes, including obesity, metabolic syndrome, and type 2 diabetes. Here, we aim to summarize select evidence from in vivo animal models and compare these results with epidemiologic research findings to advance understanding of existing foundational evidence and potential mechanistic links between circadian disruption and altered clock gene expression contributions to metabolic health-related pathologies. Findings have important implications for the treatment, prevention, and control of metabolic pathologies underlying leading causes of death and disability, including diabetes, cardiovascular disease, and cancer.
Collapse
Affiliation(s)
| | - Sean M Ronnekleiv-Kelly
- Molecular and Environmental Toxicology Center and
- Department of Surgery, Division of Surgical Oncology, School of Medicine and Public Health, University of Wisconsin, Madison Wisconsin, USA
| | - John B Hogenesch
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Kristen Mc Malecki
- Molecular and Environmental Toxicology Center and
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
- Division of Environmental and Occupational Health Sciences, University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
44
|
Geronikolou SA, Pavlopoulou A, Uça Apaydin M, Albanopoulos K, Cokkinos DV, Chrousos G. Non-Hereditary Obesity Type Networks and New Drug Targets: An In Silico Approach. Int J Mol Sci 2024; 25:7684. [PMID: 39062927 PMCID: PMC11277295 DOI: 10.3390/ijms25147684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity, a chronic, preventable disease, has significant comorbidities that are associated with a great human and financial cost for society. The aim of the present work is to reconstruct the interactomes of non-hereditary obesity to highlight recent advances of its pathogenesis, and discover potential therapeutic targets. Obesity and biological-clock-related genes and/or gene products were extracted from the biomedical literature databases PubMed, GeneCards and OMIM. Their interactions were investigated using STRING v11.0 (a database of known and predicted physical and indirect associations among genes/proteins), and a high confidence interaction score of >0.7 was set. We also applied virtual screening to discover natural compounds targeting obesity- and circadian-clock-associated proteins. Two updated and comprehensive interactomes, the (a) stress- and (b) inflammation-induced obesidomes involving 85 and 93 gene/gene products of known and/or predicted interactions with an average node degree of 9.41 and 10.8, respectively, were produced. Moreover, 15 of these were common between the two non-hereditary entities, namely, ADIPOQ, ADRB2/3, CCK, CRH, CXCL8, FOS, GCG, GNRH1, IGF1, INS, LEP, MC4R, NPY and POMC, while phelligridin E, a natural product, may function as a potent FOX1-DBD interaction blocker. Molecular networks may contribute to the understanding of the integrated regulation of energy balance/obesity pathogenesis and may associate chronopharmacology schemes with natural products.
Collapse
Affiliation(s)
- Styliani A. Geronikolou
- Clinical, Translational Research and Experimental Surgery Centre, Biomedical Research Foundation of the Academy of Athens, 4, Soranou Ephessiou Str., 11527 Athens, Greece; (D.V.C.); (G.C.)
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens Medical School, Levadias 8, 11527 Athens, Greece
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center (IBG), 35340 Izmir, Türkiye; (A.P.)
- Izmir International Biomedicine and Genome Institute, Genomics and Molecular Biotechnology Department, Dokuz Eylül University, 35340 Izmir, Türkiye
| | - Merve Uça Apaydin
- Izmir Biomedicine and Genome Center (IBG), 35340 Izmir, Türkiye; (A.P.)
- Izmir International Biomedicine and Genome Institute, Genomics and Molecular Biotechnology Department, Dokuz Eylül University, 35340 Izmir, Türkiye
| | | | - Dennis V. Cokkinos
- Clinical, Translational Research and Experimental Surgery Centre, Biomedical Research Foundation of the Academy of Athens, 4, Soranou Ephessiou Str., 11527 Athens, Greece; (D.V.C.); (G.C.)
| | - George Chrousos
- Clinical, Translational Research and Experimental Surgery Centre, Biomedical Research Foundation of the Academy of Athens, 4, Soranou Ephessiou Str., 11527 Athens, Greece; (D.V.C.); (G.C.)
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens Medical School, Levadias 8, 11527 Athens, Greece
| |
Collapse
|
45
|
Han Y, Shon J, Kwon SY, Park YJ. Effects of Dietary Protein Intake Levels on Peripheral Circadian Rhythm in Mice. Int J Mol Sci 2024; 25:7373. [PMID: 39000480 PMCID: PMC11242084 DOI: 10.3390/ijms25137373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The regulation of the circadian clock plays an important role in influencing physiological conditions. While it is reported that the timing and quantity of energy intake impact circadian regulation, the underlying mechanisms remain unclear. This study investigated the impact of dietary protein intake on peripheral clocks. Firstly, transcriptomic analysis was conducted to investigate molecular targets of low-protein intake. Secondly, mPer2::Luc knock-in mice, fed with either a low-protein, normal, or high-protein diet for 6 weeks, were analyzed for the oscillation of PER2 expression in peripheral tissues and for the expression profiles of circadian and metabolic genes. Lastly, the candidate pathway identified by the in vivo analysis was validated using AML12 cells. As a result, using transcriptomic analysis, we found that the low-protein diet hardly altered the circadian rhythm in the central clock. In animal experiments, expression levels and period lengths of PER2 were different in peripheral tissues depending on dietary protein intake; moreover, mRNA levels of clock-controlled genes and endoplasmic reticulum (ER) stress genes were affected by dietary protein intake. Induction of ER stress in AML12 cells caused an increased amplitude of Clock and Bmal1 and an advanced peak phase of Per2. This result shows that the intake of different dietary protein ratios causes an alteration of the circadian rhythm, especially in the peripheral clock of mice. Dietary protein intake modifies the oscillation of ER stress genes, which may play key roles in the regulation of the circadian clock.
Collapse
Affiliation(s)
- Yerim Han
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinyoung Shon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - So Young Kwon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yoon Jung Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
46
|
Masuda K, Sakurai T, Hirano A. A coupled model between circadian, cell-cycle, and redox rhythms reveals their regulation of oxidative stress. Sci Rep 2024; 14:15479. [PMID: 38969743 PMCID: PMC11226698 DOI: 10.1038/s41598-024-66347-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
Most organisms possess three biological oscillators, circadian clock, cell cycle, and redox rhythm, which are autonomous but interact each other. However, whether their interactions and autonomy are beneficial for organisms remains unclear. Here, we modeled a coupled oscillator system where each oscillator affected the phase of the other oscillators. We found that multiple types of coupling prevent a high H2O2 level in cells at M phase. Consequently, we hypothesized a high H2O2 sensitivity at the M phase and found that moderate coupling reduced cell damage due to oxidative stress by generating appropriate phase relationships between three rhythms, whereas strong coupling resulted in an elevated cell damage by increasing the average H2O2 level and disrupted the cell cycle. Furthermore, the multicellularity model revealed that phase variations among cells confer flexibility in synchronization with environments at the expense of adaptability to the optimal environment. Thus, both autonomy and synchrony among the oscillators are important for coordinating their phase relationships to minimize oxidative stress, and couplings balance them depending on environments.
Collapse
Affiliation(s)
- Kosaku Masuda
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Takeshi Sakurai
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
- Life Science Center for Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Arisa Hirano
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
47
|
Wang R, Liao Y, Deng Y, Shuang R. Unraveling the Health Benefits and Mechanisms of Time-Restricted Feeding: Beyond Caloric Restriction. Nutr Rev 2024:nuae074. [PMID: 38954563 DOI: 10.1093/nutrit/nuae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024] Open
Abstract
Time-restricted feeding (TRF) is a lifestyle intervention that aims to maintain a consistent daily cycle of feeding and fasting to support robust circadian rhythms. Recently, it has gained scientific, medical, and public attention due to its potential to enhance body composition, extend lifespan, and improve overall health, as well as induce autophagy and alleviate symptoms of diseases like cardiovascular diseases, type 2 diabetes, neurodegenerative diseases, cancer, and ischemic injury. However, there is still considerable debate on the primary factors that contribute to the health benefits of TRF. Despite not imposing strict limitations on calorie intake, TRF consistently led to reductions in calorie intake. Therefore, while some studies suggest that the health benefits of TRF are primarily due to caloric restriction (CR), others argue that the key advantages of TRF arise not only from CR but also from factors like the duration of fasting, the timing of the feeding period, and alignment with circadian rhythms. To elucidate the roles and mechanisms of TRF beyond CR, this review incorporates TRF studies that did not use CR, as well as TRF studies with equivalent energy intake to CR, which addresses the previous lack of comprehensive research on TRF without CR and provides a framework for future research directions.
Collapse
Affiliation(s)
- Ruhan Wang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Yan Deng
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Rong Shuang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| |
Collapse
|
48
|
Guo Y, Abou Daya F, Le HD, Panda S, Melkani GC. Diurnal expression of Dgat2 induced by time-restricted feeding maintains cardiac health in the Drosophila model of circadian disruption. Aging Cell 2024; 23:e14169. [PMID: 38616316 PMCID: PMC11258440 DOI: 10.1111/acel.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/16/2024] Open
Abstract
Circadian disruption is associated with an increased risk of cardiometabolic disorders and cardiac diseases. Time-restricted feeding/eating (TRF/TRE), restricting food intake within a consistent window of the day, has shown improvements in heart function from flies and mice to humans. However, whether and how TRF still conveys cardiac benefits in the context of circadian disruption remains unclear. Here, we demonstrate that TRF sustains cardiac performance, myofibrillar organization, and regulates cardiac lipid accumulation in Drosophila when the circadian rhythm is disrupted by constant light. TRF induces oscillations in the expression of genes associated with triglyceride metabolism. In particular, TRF induces diurnal expression of diacylglycerol O-acyltransferase 2 (Dgat2), peaking during the feeding period. Heart-specific manipulation of Dgat2 modulates cardiac function and lipid droplet accumulation. Strikingly, heart-specific overexpression of human Dgat2 at ZT 0-10 significantly improves cardiac performance in flies exposed to constant light. We have demonstrated that TRF effectively attenuates cardiac decline induced by circadian disruption. Moreover, our data suggests that diurnal expression of Dgat2 induced by TRF is beneficial for heart health under circadian disruption. Overall, our findings have underscored the relevance of TRF in preserving heart health under circadian disruptions and provided potential targets, such as Dgat2, and strategies for therapeutic interventions in mitigating cardiac aging, metabolic disorders, and cardiac diseases in humans.
Collapse
Affiliation(s)
- Yiming Guo
- Department of Pathology, Division of Molecular and Cellular PathologyHeersink School of Medicine, University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Farah Abou Daya
- Department of Pathology, Division of Molecular and Cellular PathologyHeersink School of Medicine, University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Hiep Dinh Le
- Regulatory Biology LaboratorySalk Institute for Biological StudiesLa JollaCaliforniaUSA
| | - Satchidananda Panda
- Regulatory Biology LaboratorySalk Institute for Biological StudiesLa JollaCaliforniaUSA
| | - Girish C. Melkani
- Department of Pathology, Division of Molecular and Cellular PathologyHeersink School of Medicine, University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
49
|
Goldman DM, Warbeck CB, Waterfall TJ, Sud A, Quarshie M, Craddock JC. Plant-based and Early Time-restricted Eating for Prevention and Treatment of Type 2 Diabetes in Adults: A Narrative Review. Can J Diabetes 2024; 48:341-347. [PMID: 38513822 DOI: 10.1016/j.jcjd.2024.03.002] [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: 12/11/2023] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
Type 2 diabetes (T2D) is a significant public health challenge for which effective lifestyle interventions are needed. A growing body of evidence supports the use of both plant-based eating patterns and early time-restricted eating (eTRE) for the prevention and treatment of T2D, but research has not yet explored the potential of these dietary strategies in combination. In this narrative review, we assessed the evidence by which plant-based diets, in conjunction with eTRE, could support T2D care. The electronic databases MEDLINE and the Web of Science were searched for relevant articles published throughout the last decade. Observational research has shown that healthy plant-based eating patterns and eTRE are associated with reductions in T2D risk. Interventional trials demonstrated that plant-based diets promote improvements in glycated hemoglobin, insulin resistance, glycemic management, and cardiometabolic risk factors. These changes may be mediated, in part, by reductions in oxidative stress, dietary acid load, and hepatocellular and intramyocellular lipids. The eTRE strategies were also shown to improve insulin resistance and glycemic management, and mechanisms of action included enhanced regulation of circadian rhythm and increased metabolic flexibility. Integrating these dietary strategies may produce additive benefits, mediated by reduced visceral adiposity and beneficial shifts in gut microbiota composition. However, potential barriers to concurrent implementation of these interventions may exist, including social challenges, scheduling constraints, and tolerance. Prospective trials are needed to examine their acceptability and clinical effects.
Collapse
Affiliation(s)
| | - Cassandra B Warbeck
- Department of Family Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Adam Sud
- Plant-Based for Positive Change, Austin, Texas, United States
| | | | - Joel C Craddock
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| |
Collapse
|
50
|
Holland JG, Prior KF, O'Donnell AJ, Reece SE. Testing the evolutionary drivers of malaria parasite rhythms and their consequences for host-parasite interactions. Evol Appl 2024; 17:e13752. [PMID: 39006006 PMCID: PMC11246599 DOI: 10.1111/eva.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Undertaking certain activities at the time of day that maximises fitness is assumed to explain the evolution of circadian clocks. Organisms often use daily environmental cues such as light and food availability to set the timing of their clocks. These cues may be the environmental rhythms that ultimately determine fitness, act as proxies for the timing of less tractable ultimate drivers, or are used simply to maintain internal synchrony. While many pathogens/parasites undertake rhythmic activities, both the proximate and ultimate drivers of their rhythms are poorly understood. Explaining the roles of rhythms in infections offers avenues for novel interventions to interfere with parasite fitness and reduce the severity and spread of disease. Here, we perturb several rhythms in the hosts of malaria parasites to investigate why parasites align their rhythmic replication to the host's feeding-fasting rhythm. We manipulated host rhythms governed by light, food or both, and assessed the fitness implications for parasites, and the consequences for hosts, to test which host rhythms represent ultimate drivers of the parasite's rhythm. We found that alignment with the host's light-driven rhythms did not affect parasite fitness metrics. In contrast, aligning with the timing of feeding-fasting rhythms may be beneficial for the parasite, but only when the host possess a functional canonical circadian clock. Because parasites in clock-disrupted hosts align with the host's feeding-fasting rhythms and yet derive no apparent benefit, our results suggest cue(s) from host food act as a proxy rather than being a key selective driver of the parasite's rhythm. Alternatively, parasite rhythmicity may only be beneficial because it promotes synchrony between parasite cells and/or allows parasites to align to the biting rhythms of vectors. Our results also suggest that interventions can disrupt parasite rhythms by targeting the proxies or the selective factors driving them without impacting host health.
Collapse
Affiliation(s)
- Jacob G. Holland
- Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
| | | | | | - Sarah E. Reece
- Institute of Ecology and EvolutionUniversity of EdinburghEdinburghUK
- Institute of Immunology and Infection ResearchUniversity of EdinburghEdinburghUK
| |
Collapse
|