201
|
The Circadian Regulation of Nutrient Metabolism in Diet-Induced Obesity and Metabolic Disease. Nutrients 2022; 14:nu14153136. [PMID: 35956312 PMCID: PMC9370226 DOI: 10.3390/nu14153136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity and other metabolic diseases are major public health issues that are particularly prevalent in industrialized societies where circadian rhythmicity is disturbed by shift work, jet lag, and/or social obligations. In mammals, daylight entrains the hypothalamic suprachiasmatic nucleus (SCN) to a ≈24 h cycle by initiating a transcription/translation feedback loop (TTFL) of molecular clock genes. The downstream impacts of the TTFL on clock-controlled genes allow the SCN to set the rhythm for the majority of physiological, metabolic, and behavioral processes. The TTFL, however, is ubiquitous and oscillates in tissues throughout the body. Tissues outside of the SCN are entrained to other signals, such as fed/fasting state, rather than light input. This system requires a considerable amount of biological flexibility as it functions to maintain homeostasis across varying conditions contained within a 24 h day. In the face of either circadian disruption (e.g., jet lag and shift work) or an obesity-induced decrease in metabolic flexibility, this finely tuned mechanism breaks down. Indeed, both human and rodent studies have found that obesity and metabolic disease develop when endogenous circadian pacing is at odds with the external cues. In the following review, we will delve into what is known on the circadian rhythmicity of nutrient metabolism and discuss obesity as a circadian disease.
Collapse
|
202
|
ISX-9 potentiates CaMKIIδ-mediated BMAL1 activation to enhance circadian amplitude. Commun Biol 2022; 5:750. [PMID: 35902736 PMCID: PMC9334596 DOI: 10.1038/s42003-022-03725-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 07/15/2022] [Indexed: 11/09/2022] Open
Abstract
Circadian dysregulation associates with numerous diseases including metabolic dysfunction, sleep disorder, depression and aging. Given that declined circadian amplitude is a trait commonly found with compromised health, interventions that design in precluding circadian amplitude from dampening will aid to mitigate complex, circadian-related diseases. Here we identify a neurogenic small molecule ISX-9 that is able to support persistent and higher amplitude of circadian oscillations. ISX-9 improves diurnal metabolic rhythms in middle-aged mice. Moreover, the ISX-9-treated mice show better sleep homeostasis with increased delta power during the day time and higher locomotive activity in the dark period. ISX-9 augments CaMKIIδ expression and increases BMAL1 activity via eliciting CaMKIIδ-mediated phosphorylation on BMAL1 residues S513/S515/S516, accordingly composes a positive feedback effect on enhancing circadian amplitude. CaMKIIδ-targeting, and the use of ISX-9 may serve as decent choices for treating circadian-related disorders.
Collapse
|
203
|
Tang Q, Assali DR, Güler AD, Steele AD. Dopamine systems and biological rhythms: Let's get a move on. Front Integr Neurosci 2022; 16:957193. [PMID: 35965599 PMCID: PMC9364481 DOI: 10.3389/fnint.2022.957193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023] Open
Abstract
How dopamine signaling regulates biological rhythms is an area of emerging interest. Here we review experiments focused on delineating dopamine signaling in the suprachiasmatic nucleus, nucleus accumbens, and dorsal striatum to mediate a range of biological rhythms including photoentrainment, activity cycles, rest phase eating of palatable food, diet-induced obesity, and food anticipatory activity. Enthusiasm for causal roles for dopamine in the regulation of circadian rhythms, particularly those associated with food and other rewarding events, is warranted. However, determining that there is rhythmic gene expression in dopamine neurons and target structures does not mean that they are bona fide circadian pacemakers. Given that dopamine has such a profound role in promoting voluntary movements, interpretation of circadian phenotypes associated with locomotor activity must be differentiated at the molecular and behavioral levels. Here we review our current understanding of dopamine signaling in relation to biological rhythms and suggest future experiments that are aimed at teasing apart the roles of dopamine subpopulations and dopamine receptor expressing neurons in causally mediating biological rhythms, particularly in relation to feeding, reward, and activity.
Collapse
Affiliation(s)
- Qijun Tang
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| | - Dina R. Assali
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA, United States
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Andrew D. Steele
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States
| |
Collapse
|
204
|
Yong YN, Henry CJ, Haldar S. Is There a Utility of Chrono-Specific Diets in Improving Cardiometabolic Health? Mol Nutr Food Res 2022; 66:e2200043. [PMID: 35856629 DOI: 10.1002/mnfr.202200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/31/2022] [Indexed: 11/10/2022]
Abstract
Modern lifestyle is generally associated with the consumption of three main meals per day, one of which is typically in the evening or at night. It is also well established that consumption of meals in the later part of the day, notably in the evenings, is associated with circadian desynchrony, which in turn increases the risk of non-communicable diseases, particularly cardiometabolic diseases. While it is not feasible to avoid food consumption during the evenings altogether, there is an opportunity to provide chrono-specific, diet-based solutions to mitigate some of these risks. To date, there has been substantial progress in the understanding of chrononutrition, with evidence derived mainly from in vitro and in vivo animal studies. Some of these approaches include the manipulation of the quality and quantity of certain nutrients to be consumed at specific times of the day, as well as incorporating certain dietary components (macronutrients, micronutrients, or non-nutrient bioactives, including polyphenols) with the ability to modulate circadian rhythmicity. However, robust human studies are generally lacking. In this review, the study has consolidated and critically appraised the current evidence base, with an aim to translate these findings to improve cardiometabolic health and provides recommendations to move this field forward.
Collapse
Affiliation(s)
- Yi Ning Yong
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117599, Singapore
| | - Sumanto Haldar
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore
| |
Collapse
|
205
|
Touati H, Ouali-Hassenaoui S, Dekar-Madoui A, Benhafri N, Boumansour L, Challet E, Pévet P, Vuillez P. Osmoregulatory neurons clockwork is altered during metabolic disorder induced by high energy diet in the Sand rat Psammomys obesus. BIOL RHYTHM RES 2022. [DOI: 10.1080/09291016.2022.2102710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Affiliation(s)
- Hanane Touati
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Biology and Physiology of Organisms, Bab Ezzouar, Algeria
| | - Saliha Ouali-Hassenaoui
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Biology and Physiology of Organisms, Bab Ezzouar, Algeria
| | - Aicha Dekar-Madoui
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Biology and Physiology of Organisms, Bab Ezzouar, Algeria
| | - Nadir Benhafri
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Biology and Physiology of Organisms, Bab Ezzouar, Algeria
| | - Lydia Boumansour
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Biology and Physiology of Organisms, Bab Ezzouar, Algeria
| | - Etienne Challet
- Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, CNRS, University of Strasbourg, Strasbourg, France
| | - Paul Pévet
- Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, CNRS, University of Strasbourg, Strasbourg, France
| | - Patrick Vuillez
- Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, CNRS, University of Strasbourg, Strasbourg, France
| |
Collapse
|
206
|
Vandenberghe A, Lefranc M, Furlan A. An Overview of the Circadian Clock in the Frame of Chronotherapy: From Bench to Bedside. Pharmaceutics 2022; 14:pharmaceutics14071424. [PMID: 35890319 PMCID: PMC9317821 DOI: 10.3390/pharmaceutics14071424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Most living organisms in both the plant and animal kingdoms have evolved processes to stay in tune with the alternation of day and night, and to optimize their physiology as a function of light supply. In mammals, a circadian clock relying on feedback loops between key transcription factors will thus control the temporally regulated pattern of expression of most genes. Modern ways of life have highly altered the synchronization of human activities with their circadian clocks. This review discusses the links between an altered circadian clock and the rise of pathologies. We then sum up the proofs of concept advocating for the integration of circadian clock considerations in chronotherapy for health care, medicine, and pharmacotherapy. Finally, we discuss the current challenges that circadian biology must face and the tools to address them.
Collapse
Affiliation(s)
- Alan Vandenberghe
- Univ. Lille, CNRS, UMR 8523-PhLAM-Physique des Lasers Atomes et Molécules, F-59000 Lille, France;
| | - Marc Lefranc
- Univ. Lille, CNRS, UMR 8523-PhLAM-Physique des Lasers Atomes et Molécules, F-59000 Lille, France;
- Correspondence: (M.L.); (A.F.)
| | - Alessandro Furlan
- Univ. Lille, CNRS, UMR 8523-PhLAM-Physique des Lasers Atomes et Molécules, F-59000 Lille, France;
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Correspondence: (M.L.); (A.F.)
| |
Collapse
|
207
|
Dantas Machado AC, Brown SD, Lingaraju A, Sivaganesh V, Martino C, Chaix A, Zhao P, Pinto AFM, Chang MW, Richter RA, Saghatelian A, Saltiel AR, Knight R, Panda S, Zarrinpar A. Diet and feeding pattern modulate diurnal dynamics of the ileal microbiome and transcriptome. Cell Rep 2022; 40:111008. [PMID: 35793637 PMCID: PMC9296000 DOI: 10.1016/j.celrep.2022.111008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 03/28/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022] Open
Abstract
Compositional oscillations of the gut microbiome are essential for normal peripheral circadian rhythms, both of which are disrupted in diet-induced obesity (DIO). Although time-restricted feeding (TRF) maintains circadian synchrony and protects against DIO, its impact on the dynamics of the cecal gut microbiome is modest. Thus, other regions of the gut, particularly the ileum, the nexus for incretin and bile acid signaling, may play an important role in entraining peripheral circadian rhythms. We demonstrate the effect of diet and feeding rhythms on the ileal microbiome composition and transcriptome in mice. The dynamic rhythms of ileal microbiome composition and transcriptome are dampened in DIO. TRF partially restores diurnal rhythms of the ileal microbiome and transcriptome, increases GLP-1 release, and alters the ileal bile acid pool and farnesoid X receptor (FXR) signaling, which could explain how TRF exerts its metabolic benefits. Finally, we provide a web resource for exploration of ileal microbiome and transcriptome circadian data.
Collapse
Affiliation(s)
- Ana Carolina Dantas Machado
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Steven D Brown
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Amulya Lingaraju
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Vignesh Sivaganesh
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Cameron Martino
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Peng Zhao
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Antonio F M Pinto
- Clayton Foundation Laboratories for Peptide Biology, the Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Max W Chang
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - R Alexander Richter
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, the Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Alan R Saltiel
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Institute of Diabetes and Metabolic Health, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA
| | - Rob Knight
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, CA, USA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; Institute of Diabetes and Metabolic Health, University of California, San Diego, 9500 Gilman Drive, MC 0983, La Jolla, CA, USA; VA Health Sciences, San Diego, La Jolla, CA, USA.
| |
Collapse
|
208
|
Soliman RH, Jin C, Taylor CM, Moura Coelho da Silva E, Pollock DM. Sex Differences in Diurnal Sodium Handling During Diet-Induced Obesity in Rats. Hypertension 2022; 79:1395-1408. [PMID: 35545941 PMCID: PMC9186154 DOI: 10.1161/hypertensionaha.121.18690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Emerging evidence over the past several years suggests that diurnal control of sodium excretion is sex dependent and involves the renal endothelin system. Given recent awareness of disruptions of circadian function in obesity, we determined whether diet-induced obesity impairs renal handling of an acute salt load at different times of day and whether this varies by sex and is associated with renal endothelin dysfunction. METHODS Male and female Sprague-Dawley rats were placed on a high-fat diet for 8 weeks before assessing renal sodium handling and blood pressure. RESULTS Male, but not female, rats on high fat had a significantly reduced natriuretic response to acute NaCl injection at the beginning of their active period that was associated with lower endothelin 1 (ET-1) excretion, lower ET-1 mRNA expression in the cortex and outer medulla as well as lower ETB receptor expression in the outer medulla of the high-fat rats. Obese males also had significantly higher blood pressure (telemetry) that was exacerbated by adding high salt to the diet during the last 2 weeks. While female rats developed hypertension with a high-fat diet, they were not salt sensitive and ET-1 excretion was unchanged. CONCLUSIONS These data identify diet-induced obesity as a sex-specific disruptive factor for maintaining proper sodium handling. Although high-fat diets induce hypertension in both sexes, these data reveal that males are at greater risk of salt-dependent hypertension and further suggest that females have more redundant systems that can be productive against salt-sensitive hypertension in at least some circumstances.
Collapse
Affiliation(s)
- Reham H. Soliman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham
| | - Chunhua Jin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham
| | - Crystal M. Taylor
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham
| | - Emile Moura Coelho da Silva
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham
| | - David M. Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham
| |
Collapse
|
209
|
Pickel L, Lee JH, Maughan H, Shi IQ, Verma N, Yeung C, Guttman D, Sung H. Circadian rhythms in metabolic organs and the microbiota during acute fasting in mice. Physiol Rep 2022; 10:e15393. [PMID: 35851583 PMCID: PMC9295129 DOI: 10.14814/phy2.15393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 04/16/2023] Open
Abstract
The circadian clock regulates metabolism in anticipation of regular changes in the environment. It is found throughout the body, including in key metabolic organs such as the liver, adipose tissues, and intestine, where the timing of the clock is set largely by nutrient signaling. However, the circadian clocks of these tissues during the fasted state have not been completely characterized. Moreover, the sufficiency of a functioning host clock to produce diurnal rhythms in the composition of the microbiome in fasted animals has not been explored. To this end, mice were fasted 24 h prior to collection of key metabolic tissues and fecal samples for the analysis of circadian clock gene expression and microbiome composition. Rhythm characteristics were determined using CircaCompare software. We identify tissue-specific changes to circadian clock rhythms upon fasting, particularly in the brown adipose tissue, and for the first time demonstrate the rhythmicity of the microbiome in fasted animals.
Collapse
Affiliation(s)
- Lauren Pickel
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
| | - Ju Hee Lee
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| | | | - Irisa Qianwen Shi
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
| | - Navkiran Verma
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| | - Christy Yeung
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| | - David Guttman
- Centre for the Analysis of Genome Evolution & FunctionUniversity of TorontoTorontoOntarioUSA
| | - Hoon‐Ki Sung
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| |
Collapse
|
210
|
Ma Z, Eaton M, Liu Y, Zhang J, Chen X, Tu X, Shi Y, Que Z, Wettschurack K, Zhang Z, Shi R, Chen Y, Kimbrough A, Lanman NA, Schust L, Huang Z, Yang Y. Deficiency of autism-related Scn2a gene in mice disrupts sleep patterns and circadian rhythms. Neurobiol Dis 2022; 168:105690. [PMID: 35301122 PMCID: PMC9018617 DOI: 10.1016/j.nbd.2022.105690] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/21/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) affects ~2% of the population in the US, and monogenic forms of ASD often result in the most severe manifestation of the disorder. Recently, SCN2A has emerged as a leading gene associated with ASD, of which abnormal sleep pattern is a common comorbidity. SCN2A encodes the voltage-gated sodium channel NaV1.2. Predominantly expressed in the brain, NaV1.2 mediates the action potential firing of neurons. Clinical studies found that a large portion of children with SCN2A deficiency have sleep disorders, which severely impact the quality of life of affected individuals and their caregivers. The underlying mechanism of sleep disturbances related to NaV1.2 deficiency, however, is not known. Using a gene-trap Scn2a-deficient mouse model (Scn2atrap), we found that Scn2a deficiency results in increased wakefulness and reduced non-rapid-eye-movement (NREM) sleep. Brain region-specific Scn2a deficiency in the suprachiasmatic nucleus (SCN) containing region, which is involved in circadian rhythms, partially recapitulates the sleep disturbance phenotypes. At the cellular level, we found that Scn2a deficiency disrupted the firing pattern of spontaneously firing neurons in the SCN region. At the molecular level, RNA-sequencing analysis revealed differentially expressed genes in the circadian entrainment pathway including core clock genes Per1 and Per2. Performing a transcriptome-based compound discovery, we identified dexanabinol (HU-211), a putative glutamate receptor modulator, that can partially reverse the sleep disturbance in mice. Overall, our study reveals possible molecular and cellular mechanisms underlying Scn2a deficiency-related sleep disturbances, which may inform the development of potential pharmacogenetic interventions for the affected individuals.
Collapse
Affiliation(s)
- Zhixiong Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Muriel Eaton
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Yushuang Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Jingliang Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Xiaoling Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yiqiang Shi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhefu Que
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Kyle Wettschurack
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA
| | - Zaiyang Zhang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Yueyi Chen
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Nadia A Lanman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Leah Schust
- FamilieSCN2A Foundation, P.O. Box 82, East Longmeadow, MA 01028, USA
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy & Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47906, USA.
| |
Collapse
|
211
|
Neves AR, Albuquerque T, Quintela T, Costa D. Circadian rhythm and disease: Relationship, new insights, and future perspectives. J Cell Physiol 2022; 237:3239-3256. [PMID: 35696609 DOI: 10.1002/jcp.30815] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 02/01/2023]
Abstract
The circadian system is responsible for internal functions and regulation of the organism according to environmental cues (zeitgebers). Circadian rhythm dysregulation or chronodisruption has been associated with several diseases, from mental to autoimmune diseases, and with life quality change. Following this, some therapies have been developed to correct circadian misalignments, such as light therapy and chronobiotics. In this manuscript, we describe the circadian-related diseases so far investigated, and studies reporting relevant data on this topic, evidencing this relationship, are included. Despite the actual limitations in published work, there is clear evidence of the correlation between circadian rhythm dysregulation and disease origin/development, and, in this way, clock-related therapies emerge as great progress in the clinical field. Future improvements in such interventions can lead to the development of successful chronotherapy strategies, deeply contributing to enhanced therapeutic outcomes.
Collapse
Affiliation(s)
- Ana R Neves
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Tânia Albuquerque
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal.,Unidade de Investigação para o Desenvolvimento do Interior (UDI-IPG), Instituto Politécnico da Guarda, Guarda, Portugal
| | - Diana Costa
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| |
Collapse
|
212
|
Evening chronotype is associated with severe NAFLD in obesity. Int J Obes (Lond) 2022; 46:1638-1643. [PMID: 35676442 DOI: 10.1038/s41366-022-01159-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND/OBJECTIVES Chronotype, i.e., the individual attitude to carry out the daily activities at a certain time of the day, has been reported to play a role in metabolic diseases. Thus, the aim of our study was to investigate the association of non-alcoholic fatty liver disease (NAFLD) with chronotype categories in individuals with obesity. SUBJECTS/METHODS In this cross-sectional study, participants underwent to anthropometric, biochemical, and the Visceral Adiposity Index (VAI) assessment. Chronotype categories were assessed by the Horne-Ostberg Morningness-Eveningness Questionnaire (MEQ). The following indirect indices of NAFLD were calculated: Liver Fat Equation (LFE), Hepatic Steatosis Index (HSI) and Index of Non-alcoholic steatohepatitis (ION). RESULTS The study population consisted of 87 participants (40.3 ± 12.3 years; BMI 45.6 ± 6.3 kg/m2; 64F/23M). According to the chronotype score, 31.0% were morning (MC), 36.8% were intermediate (IC), and 32.2% evening chronotype (EC). Individuals with IC were significantly younger than those with MC and EC (p = 0.002 and p = 0.047, respectively). EC presented significantly higher BMI (p < 0.001), waist circumference (p = 0.007), and hip circumference (p = 0.001) than MC. Moreover, EC had higher BMI (p < 0.001) and waist circumference (p = 0.011) than IC. All participants presented NAFLD (HSI ≥ 36 and LFE ≥ 5). Nevertheless, only individuals with EC were above the threshold of non-alcoholic steatohepatitis evaluated as ION index (ION ≥ 50). Individuals with EC presented significantly higher VAI (p = 0.036), LFE (p = 0.034), HSI (p < 0.001) and ION (p = 0.014) than MC, also after the adjustment for age, gender, and BMI. CONCLUSION EC is associated with more severe NAFLD independently of age, gender, and BMI than MC and IC in individuals with obesity.
Collapse
|
213
|
Samad M, Agostinelli F, Sato T, Shimaji K, Baldi P. CircadiOmics: circadian omic web portal. Nucleic Acids Res 2022; 50:W183-W190. [PMID: 35657089 PMCID: PMC9252794 DOI: 10.1093/nar/gkac419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/14/2022] [Accepted: 06/01/2022] [Indexed: 11/12/2022] Open
Abstract
Circadian rhythms are a foundational aspect of biology. These rhythms are found at the molecular level in every cell of every living organism and they play a fundamental role in homeostasis and a variety of physiological processes. As a result, biomedical research of circadian rhythms continues to expand at a rapid pace. To support this research, CircadiOmics (http://circadiomics.igb.uci.edu/) is the largest annotated repository and analytic web server for high-throughput omic (e.g. transcriptomic, metabolomic, proteomic) circadian time series experimental data. CircadiOmics contains over 290 experiments and over 100 million individual measurements, across >20 unique tissues/organs, and 11 different species. Users are able to visualize and mine these datasets by deriving and comparing periodicity statistics for oscillating molecular species including: period, amplitude, phase, P-value and q-value. These statistics are obtained from BIO_CYCLE and JTK_CYCLE and are intuitively aggregated and displayed for comparison. CircadiOmics is the most up-to-date and cutting-edge web portal for searching and analyzing circadian omic data and is used by researchers around the world.
Collapse
Affiliation(s)
- Muntaha Samad
- Department of Computer Science, University of California Irvine, Irvine, CA 92697, USA.,Institute for Genomics and Bioinformatics, University of California, Irvine CA 92697, USA
| | - Forest Agostinelli
- Department of Computer Science and Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Tomoki Sato
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Kohei Shimaji
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Pierre Baldi
- Department of Computer Science, University of California Irvine, Irvine, CA 92697, USA.,Institute for Genomics and Bioinformatics, University of California, Irvine CA 92697, USA
| |
Collapse
|
214
|
Ratiner K, Shapiro H, Goldenberg K, Elinav E. Time-limited diets and the gut microbiota in cardiometabolic disease. J Diabetes 2022; 14:377-393. [PMID: 35698246 PMCID: PMC9366560 DOI: 10.1111/1753-0407.13288] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, intermittent fasting (IF), including periodic fasting and time-restricted feeding (TRF), has been increasingly suggested to constitute a promising treatment for cardiometabolic diseases (CMD). A deliberate daily pause in food consumption influences the gut microbiome and the host circadian clock, resulting in improved cardiometabolic health. Understanding the molecular mechanisms by which circadian host-microbiome interactions affect host metabolism and immunity may add a potentially important dimension to effective implementation of IF diets. In this review, we discuss emerging evidence potentially linking compositional and functional alterations of the gut microbiome with IF impacts on mammalian metabolism and risk of development of hypertension, type 2 diabetes (T2D), obesity, and their long-term micro- and macrovascular complications. We highlight the challenges and unknowns in causally linking diurnal bacterial signals with dietary cues and downstream metabolic consequences and means of harnessing these signals toward future microbiome integration into precision medicine.
Collapse
Affiliation(s)
- Karina Ratiner
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Hagit Shapiro
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Kim Goldenberg
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Eran Elinav
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
- Microbiome & Cancer Division, DKFZHeidelbergGermany
| |
Collapse
|
215
|
Garcia-Rios A, Ordovas JM. Chronodisruption and cardiovascular disease. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2022; 34 Suppl 1:S32-S37. [PMID: 35125247 DOI: 10.1016/j.arteri.2021.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 06/14/2023]
Abstract
Cardiovascular disease (CVD) is an important challenge for clinicians, researchers and governments to reduce the impact on the global health burden and socioeconomic costs. Moreover, far from diminishing, cardiometabolic risk factors leading to CVD development are on the rise. In order to stop the CVD pandemic, it is not enough to merely attempt to control traditional risk factors. In this regard, chronobiology, the science that studies biological rhythms, has become an important field in research in the last years. Circadian disruption or chronodisruption, defined as a relevant disturbance of the internal temporal order of physiological circadian rhythms significantly increases the risk of CVD. In this article we review some of the evidence that has made chronobiology one of the most emerging scenarios to take into account in routine clinical practice in which a translation of all this evidence should be mandatory.
Collapse
Affiliation(s)
- Antonio Garcia-Rios
- Lipids and Atherosclerosis Unit, Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Jose M Ordovas
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain; Nutrition and Genomics Laboratory, J.M, US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| |
Collapse
|
216
|
Circadian rhythm of lipid metabolism. Biochem Soc Trans 2022; 50:1191-1204. [PMID: 35604112 DOI: 10.1042/bst20210508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Lipids comprise a diverse group of metabolites that are indispensable as energy storage molecules, cellular membrane components and mediators of inter- and intra-cellular signaling processes. Lipid homeostasis plays a crucial role in maintaining metabolic health in mammals including human beings. A growing body of evidence suggests that the circadian clock system ensures temporal orchestration of lipid homeostasis, and that perturbation of such diurnal regulation leads to the development of metabolic disorders comprising obesity and type 2 diabetes. In view of the emerging role of circadian regulation in maintaining lipid homeostasis, in this review, we summarize the current knowledge on lipid metabolic pathways controlled by the mammalian circadian system. Furthermore, we review the emerging connection between the development of human metabolic diseases and changes in lipid metabolites that belong to major classes of lipids. Finally, we highlight the mechanisms underlying circadian organization of lipid metabolic rhythms upon the physiological situation, and the consequences of circadian clock dysfunction for dysregulation of lipid metabolism.
Collapse
|
217
|
Litichevskiy L, Thaiss CA. The Oscillating Gut Microbiome and Its Effects on Host Circadian Biology. Annu Rev Nutr 2022; 42:145-164. [PMID: 35576592 DOI: 10.1146/annurev-nutr-062320-111321] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The microbial community colonizing the gastrointestinal tract, collectively termed the gut microbiota, is an important element of the host organism due to its impact on multiple aspects of health. The digestion of food, secretion of immunostimulatory molecules, performance of chemical reactions in the intestine, and production of metabolites by the microbiota contribute to host homeostasis and disease. Recent discoveries indicate that these major functions are not constantly performed over the course of a day, but rather undergo diurnal fluctuations due to compositional and biogeographical oscillations in the microbiota. Here, we summarize the characteristics and origins of diurnal microbiome rhythms as well as their functional consequences for the circadian biology of the host. We describe the major known pathways of circadian host-microbiome communication and discuss possible implications of altered diurnal microbiome rhythms for human disease. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lev Litichevskiy
- Microbiology Department, Institute for Immunology, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
| | - Christoph A Thaiss
- Microbiology Department, Institute for Immunology, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
| |
Collapse
|
218
|
Ribas-Latre A, Eckel-Mahan K. Nutrients and the Circadian Clock: A Partnership Controlling Adipose Tissue Function and Health. Nutrients 2022; 14:2084. [PMID: 35631227 PMCID: PMC9147080 DOI: 10.3390/nu14102084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 01/14/2023] Open
Abstract
White adipose tissue (WAT) is a metabolic organ with flexibility to retract and expand based on energy storage and utilization needs, processes that are driven via the coordination of different cells within adipose tissue. WAT is comprised of mature adipocytes (MA) and cells of the stromal vascular cell fraction (SVF), which include adipose progenitor cells (APCs), adipose endothelial cells (AEC) and infiltrating immune cells. APCs have the ability to proliferate and undergo adipogenesis to form MA, the main constituents of WAT being predominantly composed of white, triglyceride-storing adipocytes with unilocular lipid droplets. While adiposity and adipose tissue health are controlled by diet and aging, the endogenous circadian (24-h) biological clock of the body is highly active in adipose tissue, from adipocyte progenitor cells to mature adipocytes, and may play a unique role in adipose tissue health and function. To some extent, 24-h rhythms in adipose tissue rely on rhythmic energy intake, but individual circadian clock proteins are also thought to be important for healthy fat. Here we discuss how and why the clock might be so important in this metabolic depot, and how temporal and qualitative aspects of energy intake play important roles in maintaining healthy fat throughout aging.
Collapse
Affiliation(s)
- Aleix Ribas-Latre
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA;
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, University Hospital Leipzig, D-04103 Leipzig, Germany
| | - Kristin Eckel-Mahan
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA;
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
219
|
Sato T, Sassone-Corsi P. Nutrition, metabolism, and epigenetics: pathways of circadian reprogramming. EMBO Rep 2022; 23:e52412. [PMID: 35412705 PMCID: PMC9066069 DOI: 10.15252/embr.202152412] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/28/2021] [Accepted: 03/30/2022] [Indexed: 01/07/2023] Open
Abstract
Food intake profoundly affects systemic physiology. A large body of evidence has indicated a link between food intake and circadian rhythms, and ~24-h cycles are deemed essential for adapting internal homeostasis to the external environment. Circadian rhythms are controlled by the biological clock, a molecular system remarkably conserved throughout evolution. The circadian clock controls the cyclic expression of numerous genes, a regulatory program common to all mammalian cells, which may lead to various metabolic and physiological disturbances if hindered. Although the circadian clock regulates multiple metabolic pathways, metabolic states also provide feedback on the molecular clock. Therefore, a remarkable feature is reprogramming by nutritional challenges, such as a high-fat diet, fasting, ketogenic diet, and caloric restriction. In addition, various factors such as energy balance, histone modifications, and nuclear receptor activity are involved in the remodeling of the clock. Herein, we review the interaction of dietary components with the circadian system and illustrate the relationships linking the molecular clock to metabolism and critical roles in the remodeling process.
Collapse
Affiliation(s)
- Tomoki Sato
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, INSERM U1233, University of California, Irvine, CA, USA
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, INSERM U1233, University of California, Irvine, CA, USA
| |
Collapse
|
220
|
Martchenko A, Biancolin AD, Martchenko SE, Brubaker PL. Nobiletin ameliorates high fat-induced disruptions in rhythmic glucagon-like peptide-1 secretion. Sci Rep 2022; 12:7271. [PMID: 35508494 PMCID: PMC9068808 DOI: 10.1038/s41598-022-11223-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/20/2022] [Indexed: 02/06/2023] Open
Abstract
The incretin hormone glucagon-like peptide-1 (GLP-1) is secreted by the intestinal L cell in response to nutrient intake. However, GLP-1 secretion also follows a circadian rhythm which is disrupted by the saturated fatty acid palmitate in vitro and high-fat diet (HFD) feeding in vivo. The flavonoid nobiletin is a clock enhancer which improves metabolic homeostasis. Therefore, the aim of this study was to elucidate whether and how nobiletin mitigates the negative effects of palmitate and HFD-feeding on rhythmic GLP-1 release. Pre-treatment of murine GLUTag L cells with palmitate dampened the GLP-1 secretory response at the normal peak of secretion, while nobiletin co-treatment restored GLP-1 secretion and upregulated the ‘metabolic pathway’ transcriptome. Mice fed a HFD also lost their GLP-1 secretory rhythm in association with markedly increased GLP-1 levels and upregulation of L cell transcriptional pathways related to ‘sensing’ and ‘transducing’ cellular stimuli at the normal peak of GLP-1 release. Nobiletin co-administration reduced GLP-1 levels to more physiological levels and upregulated L cell ‘oxidative metabolism’ transcriptional pathways. Furthermore, nobiletin improved colonic microbial 16S rRNA gene diversity and reduced the levels of Proteobacteria in HFD-fed mice. Collectively, this study establishes that nobiletin improves the normal rhythm in GLP-1 secretion following fat-induced disruption.
Collapse
Affiliation(s)
- Alexandre Martchenko
- Department of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Andrew D Biancolin
- Department of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Sarah E Martchenko
- Department of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
221
|
Downton P, Sanna F, Maidstone R, Poolman TM, Hayter EA, Dickson SH, Ciccone NA, Early JO, Adamson A, Spiller DG, Simpkins DA, Baxter M, Fischer R, Rattray M, Loudon ASI, Gibbs JE, Bechtold DA, Ray DW. Chronic inflammatory arthritis drives systemic changes in circadian energy metabolism. Proc Natl Acad Sci U S A 2022; 119:e2112781119. [PMID: 35482925 PMCID: PMC9170023 DOI: 10.1073/pnas.2112781119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 03/01/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic inflammation underpins many human diseases. Morbidity and mortality associated with chronic inflammation are often mediated through metabolic dysfunction. Inflammatory and metabolic processes vary through circadian time, suggesting an important temporal crosstalk between these systems. Using an established mouse model of rheumatoid arthritis, we show that chronic inflammatory arthritis results in rhythmic joint inflammation and drives major changes in muscle and liver energy metabolism and rhythmic gene expression. Transcriptional and phosphoproteomic analyses revealed alterations in lipid metabolism and mitochondrial function associated with increased EGFR-JAK-STAT3 signaling. Metabolomic analyses confirmed rhythmic metabolic rewiring with impaired β-oxidation and lipid handling and revealed a pronounced shunt toward sphingolipid and ceramide accumulation. The arthritis-related production of ceramides was most pronounced during the day, which is the time of peak inflammation and increased reliance on fatty acid oxidation. Thus, our data demonstrate that localized joint inflammation drives a time-of-day–dependent build-up of bioactive lipid species driven by rhythmic inflammation and altered EGFR-STAT signaling.
Collapse
Affiliation(s)
- Polly Downton
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Fabio Sanna
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Robert Maidstone
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Toryn M. Poolman
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Edward A. Hayter
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Suzanna H. Dickson
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Nick A. Ciccone
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - James O. Early
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Antony Adamson
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David G. Spiller
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Devin A. Simpkins
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Matthew Baxter
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, OX3 7FZ, United Kingdom
| | - Magnus Rattray
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Andrew S. I. Loudon
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Julie E. Gibbs
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David A. Bechtold
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David W. Ray
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
222
|
Chen S, Ali I, Li X, Long D, Zhang Y, Long R, Huang X. Shifts in Fecal Metabolite Profiles Associated With Ramadan Fasting Among Chinese and Pakistani Individuals. Front Nutr 2022; 9:845086. [PMID: 35600819 PMCID: PMC9113920 DOI: 10.3389/fnut.2022.845086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
The human gut microbiota has been proposed to serve as a multifunctional organ in host metabolism, contributing effects to nutrient acquisition, immune response, and digestive health. Fasting during Ramadan may alter the composition of gut microbiota through changes in dietary behavior, which ultimately affects the contents of various metabolites in the gut. Here, we used liquid chromatography–mass spectrometry-based metabolomics to investigate the composition of fecal metabolites in Chinese and Pakistani individuals before and after Ramadan fasting. Principal component analysis showed distinct separation of metabolite profiles among ethnic groups as well as between pre- and post-fasting samples. After Ramadan fasting, the Chinese and Pakistani groups showed significant differences in their respective contents of various fecal metabolites. In particular, L-histidine, lycofawcine, and cordycepin concentrations were higher after Ramadan fasting in the Chinese group, while brucine was enriched in the Pakistani group. The KEGG analysis suggested that metabolites related to purine metabolism, 2-oxocarboxylic acid metabolism, and lysine degradation were significantly enriched in the total subject population pre-fasting vs. post-fasting comparisons. Several bacterial taxa were significantly correlated with specific metabolites unique to each ethnic group, suggesting that changes in fecal metabolite profiles related to Ramadan fasting may be influenced by associated shifts in gut microbiota. The fasting-related differences in fecal metabolite profile, together with these group-specific correlations between taxa and metabolites, support our previous findings that ethnic differences in dietary composition also drive variation in gut microbial composition and diversity. This landscape view of interconnected dietary behaviors, microbiota, and metabolites contributes to the future development of personalized, diet-based therapeutic strategies for gut-related disorders.
Collapse
Affiliation(s)
- Siyu Chen
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Ikram Ali
- School of Public Health, Lanzhou University, Lanzhou, China
- College of Ecology, Lanzhou University, Lanzhou, China
| | - Xin Li
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Danfeng Long
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Ying Zhang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Ruijun Long
- College of Ecology, Lanzhou University, Lanzhou, China
- Ruijun Long
| | - Xiaodan Huang
- School of Public Health, Lanzhou University, Lanzhou, China
- *Correspondence: Xiaodan Huang
| |
Collapse
|
223
|
Zou S, Jiao X, Liu J, Qi D, Pei X, Lu D, Huang S, Li Z. High-Fat Nutritional Challenge Reshapes Circadian Signatures in Murine Extraorbital Lacrimal Glands. Invest Ophthalmol Vis Sci 2022; 63:23. [PMID: 35588356 PMCID: PMC9123521 DOI: 10.1167/iovs.63.5.23] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose A high-fat diet (HFD) increases the risk of developing many systemic diseases; however, the effects of high fat intake on lacrimal gland functions and the molecular mechanisms underlying these effects are unknown. We explored the effects of an HFD on the circadian rhythms of the extraorbital lacrimal glands (ELGs). Methods Male C57BL/6J mice maintained on a 12/12-hour light/dark cycle were fed an ad libitum HFD or normal chow (NC) for 2 weeks. The ELGs were collected from euthanized animals every 3 hours throughout the circadian cycle (24 hours). Using high-throughput RNA-sequencing (RNA-Seq), we studied the circadian transcriptomic profile of the ELGs. Circadian oscillations in cell size, secretion response, lipid deposition, and immune cell trafficking of the ELGs were also analyzed. Results An HFD modulated the circadian transcriptomic profile of the ELGs, including the composition, phase, and amplitude of cyclical transcript oscillations, and affected the associated signaling pathways at spatiotemporal levels. HFD feeding significantly altered the normal rhythmic oscillations of ELG cell size, immune cell trafficking, secretion response, and lipid deposition. After dietary reversal in HFD-fed animals, the activity, core temperature, and lipid accumulation in lacrimal glands recovered partially to the level of NC-fed animals. However, the average cell size of the ELGs, the recruitment of immune cells, and the rhythm of lacrimal secretion did not return to the levels of the NC-fed group. Conclusions HFD perturbation interferes with the cyclical transcriptomic profile, cell size, immune cell trafficking, and secretion function of the ELGs with a strikingly high sensitivity.
Collapse
Affiliation(s)
- Sen Zou
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Jiangman Liu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou City, China
| |
Collapse
|
224
|
Apple polyphenol extract targets circadian rhythms to improve liver biological clock and lipid homeostasis in C57BL/6 male mice with mistimed high-fat diet feeding. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
225
|
Sun J, She Y, Fang P, Gu X, Zhang Z. Time-restricted feeding prevents metabolic diseases through the regulation of galanin/GALR1 expression in the hypothalamus of mice. Eat Weight Disord 2022; 27:1415-1425. [PMID: 34370270 DOI: 10.1007/s40519-021-01280-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Time-restricted feeding (TRF) reverses obesity and insulin resistance, yet the central mechanisms underlying its beneficial effects are not fully understood. Recent studies suggest a critical role of hypothalamic galanin and its receptors in the regulation of energy balance. It is yet unclear whether TRF could regulate the expression of galanin and its receptors in the hypothalamus of mice fed a high-fat diet. METHODS To test this effect, we subjected mice to either ad lib or TRF of a high-fat diet for 8 h per day. After 4 weeks, galanin and many neuropeptides associated with the function of metabolism were examined. RESULTS The present findings showed that mice under TRF consume equivalent calories from a high-fat diet as those with ad lib access, yet are protected against obesity and have improved glucose metabolism. Plasma galanin, orexin A, irisin and adropin levels were significantly reversed by TRF regimen. Besides, TRF regimen reversed the progression of metabolic disorders in mice by increasing GLUT4 and PGC-1α expression in skeletal muscles. Moreover, the levels of galanin and GALR1 expression were severely diminished in the hypothalamus of the TRF mice, whereas GALR2 was highly expressed. CONCLUSIONS TRF diminished galanin and GALR1 expression, and increased GALR2 expression in the hypothalamus of mice fed a high-fat diet. The current studies provide additional evidence that TRF is effective in improving HFD-induced hyperglycemia and insulin resistance in mice, and this effect could be associated with TRF-induced changes of the galanin systems in the hypothalamus. LEVEL OF EVIDENCE No level of evidence, animal studies.
Collapse
Affiliation(s)
- Jingjing Sun
- Affiliated Hospital of Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Yuqing She
- Department of Endocrinology, Pukou Branch of Jiangsu People's Hospital, Nanjing, China
| | - Penghua Fang
- Department of Physiology, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China
| | - Xuewen Gu
- Department of Pathology, Clinical Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
226
|
Bermudez Contreras E, Sutherland RJ, Mohajerani MH, Whishaw IQ. Challenges of a small world analysis for the continuous monitoring of behavior in mice. Neurosci Biobehav Rev 2022; 136:104621. [PMID: 35307475 DOI: 10.1016/j.neubiorev.2022.104621] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/14/2022] [Accepted: 03/11/2022] [Indexed: 12/18/2022]
Abstract
Documenting a mouse's "real world" behavior in the "small world" of a laboratory cage with continuous video recordings offers insights into phenotypical expression of mouse genotypes, development and aging, and neurological disease. Nevertheless, there are challenges in the design of a small world, the behavior selected for analysis, and the form of the analysis used. Here we offer insights into small world analyses by describing how acute behavioral procedures can guide continuous behavioral methodology. We show how algorithms can identify behavioral acts including walking and rearing, circadian patterns of action including sleep duration and waking activity, and the organization of patterns of movement into home base activity and excursions, and how they are altered with aging. We additionally describe how specific tests can be incorporated within a mouse's living arrangement. We emphasize how machine learning can condense and organize continuous activity that extends over extended periods of time.
Collapse
Affiliation(s)
| | - Robert J Sutherland
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada
| | - Majid H Mohajerani
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada.
| | - Ian Q Whishaw
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada
| |
Collapse
|
227
|
Zitting KM, Vetrivelan R, Yuan RK, Vujovic N, Wang W, Bandaru SS, Quan SF, Klerman EB, Scheer FAJL, Buxton OM, Williams JS, Duffy JF, Saper CB, Czeisler CA. Chronic circadian disruption on a high-fat diet impairs glucose tolerance. Metabolism 2022; 130:155158. [PMID: 35150732 PMCID: PMC11797635 DOI: 10.1016/j.metabol.2022.155158] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Nearly 14% of Americans experience chronic circadian disruption due to shift work, increasing their risk of obesity, diabetes, and other cardiometabolic disorders. These disorders are also exacerbated by modern eating habits such as frequent snacking and consumption of high-fat foods. METHODS We investigated the effects of recurrent circadian disruption (RCD) on glucose metabolism in C57BL/6 mice and in human participants exposed to non-24-h light-dark (LD) schedules vs. those on standard 24-h LD schedules. These LD schedules were designed to induce circadian misalignment between behaviors including rest/activity and fasting/eating with the output of the near-24-h central circadian pacemaker, while minimizing sleep loss, and were maintained for 12 weeks in mice and 3 weeks in humans. We examined interactions of these circadian-disrupted schedules compared to control 24-h schedules with a lower-fat diet (LFD, 13% in mouse and 25-27% in humans) and high-fat diet (HFD, 45% in mouse and 45-50% in humans). We also used young vs. older mice to determine whether they would respond differently to RCD. RESULTS When combined with a HFD, we found that RCD caused significant weight gain in mice and increased body fat in humans, and significantly impaired glucose tolerance and insulin sensitivity in both mice and humans, but this did not occur when RCD was combined with a LFD. This effect was similar in both young and older mice. CONCLUSION These results in both humans and a model organism indicate that circadian disruption has an adverse effect on metabolism among individuals eating a high-fat Western-style diet, even in the absence of significant sleep loss, and suggest that reducing dietary fat may protect against the metabolic consequences of a lifestyle (such as shift work) that involves chronic circadian disruption.
Collapse
Affiliation(s)
- Kirsi-Marja Zitting
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ramalingam Vetrivelan
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology, Program in Neuroscience and Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Robin K Yuan
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nina Vujovic
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Wang
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Sathyajit S Bandaru
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology, Program in Neuroscience and Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Stuart F Quan
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Orfeu M Buxton
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Biobehavioral Health, University Park PA 16802, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jeanne F Duffy
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford B Saper
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology, Program in Neuroscience and Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Charles A Czeisler
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
228
|
Abstract
Circadian rhythms are approximately 24-hour cycles of variation in physiological processes, gene expression, and behavior. They result from the interplay of internal biological clocks with daily environmental rhythms, including light/dark and feeding/fasting. Note that 24-hour rhythms of liver metabolic processes have been known for almost 100 years. Modern studies reveal that, like metabolism, hepatic gene expression is highly rhythmic. Genetic or environmental changes can disrupt the circadian rhythms of the liver, leading to metabolic disorders and hepatocellular carcinoma. In this review, we summarize the current understanding of mechanisms regulating rhythmic gene expression in the liver, highlighting the roles of transcription factors that comprise the core clock molecular as well as noncanonical regulators. We emphasize the plasticity of circadian rhythms in the liver as it responds to multiple inputs from the external and internal environments as well as the potential of circadian medicine to impact liver-related diseases.
Collapse
Affiliation(s)
- Dongyin Guan
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX,Correspondence: Dongyin Guan, PhD (); Mitchell A. Lazar, MD, PhD ()
| | - Mitchell A. Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA,Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA,Correspondence: Dongyin Guan, PhD (); Mitchell A. Lazar, MD, PhD ()
| |
Collapse
|
229
|
Soliz-Rueda JR, López-Fernández-Sobrino R, Bravo FI, Aragonès G, Suarez M, Muguerza B. Grape Seed Proanthocyanidins Mitigate the Disturbances Caused by an Abrupt Photoperiod Change in Healthy and Obese Rats. Nutrients 2022; 14:nu14091834. [PMID: 35565801 PMCID: PMC9100649 DOI: 10.3390/nu14091834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Variations in the light/dark cycle and obesogenic diets trigger physiological and behavioral disorders. Proanthocyanidins, in addition to their healthy properties, have recently demonstrated a modulating effect on biological rhythms. Therefore, the aim of this study was to evaluate the administration of a grape seed proanthocyanidin-rich extract (GSPE) to mitigate the disruption caused by a sudden photoperiod change in healthy and cafeteria (CAF)-diet obese rats. For this, 48 photoperiod-sensitive Fischer 344 rats were fed standard or CAF diets for 6 weeks under a standard (12 h light/day, L12) conditions. Then, rats were switched to a long (18 h light/day, L18) or short (6 h light/day, L6) photoperiod and administered vehicle or GSPE (25 mg/kg) for 1 week. Body weight (BW) and food intake (FI) were recorded weekly. Animal activity and serum hormone concentrations were studied before and after the photoperiod change. Hormone levels were measured both at 3 h (ZT3) and 15 h (ZT15) after the onset of light. Results showed the impact of the CAF diet and photoperiod on the BW, FI, activity, and hormonal status of the animals. GSPE administration resulted in an attenuation of the changes produced by the photoperiod disruption. Specifically, GSPE in L6 CAF-fed rats reduced serum corticosterone concentration, restoring its circadian rhythm, increased the T3-to-T4 ratio, and increased light phase activity, while under L18, it decreased BW and testosterone concentration and increased the animal activity. These results suggest that GSPE may contribute to the adaptation to the new photoperiods. However, further studies are needed to elucidate the metabolic pathways and processes involved in these events.
Collapse
|
230
|
Lee S, Hong CI. Organoids as Model Systems to Investigate Circadian Clock-Related Diseases and Treatments. Front Genet 2022; 13:874288. [PMID: 35559029 PMCID: PMC9086274 DOI: 10.3389/fgene.2022.874288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Circadian rhythms exist in most cell types in mammals regulating temporal organization of numerous cellular and physiological processes ranging from cell cycle to metabolism. The master clock, suprachiasmatic nucleus (SCN) in the hypothalamus, processes light input and coordinates peripheral clocks optimizing organisms' survival and functions aligning with external conditions. Intriguingly, it was demonstrated that circadian rhythms in the mouse liver can be decoupled from the master clock under time-restricted feeding regimen when food was provided during their inactive phase. Furthermore, mouse liver showed clock-controlled gene expression even in the absence of the master clock demonstrating independent functions of peripheral clocks apart from the SCN. These findings suggest a dynamic relationship between the master and peripheral clocks and highlight potential functions of peripheral clocks independent of the master clock. Importantly, disruption of circadian rhythms correlates with numerous human ailments including cancer and metabolic diseases, suggesting that diseases may be exacerbated by disruption of circadian rhythms in the SCN and/or peripheral clocks. However, molecular mechanisms providing causative links between circadian rhythms and human diseases remain largely unknown. Recent technical advances highlighted PCS- and tissue-derived 3-dimensional organoids as in vitro organs that possess numerous applications ranging from disease modeling to drug screening. In this mini-review, we highlight recent findings on the importance and contributions of peripheral clocks and potential uses of 3D organoids investigating complex circadian clock-related diseases.
Collapse
Affiliation(s)
| | - Christian I. Hong
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
231
|
Burckard O, Teboul M, Delaunay F, Chaves M. Cycle dynamics and synchronization in a coupled network of peripheral circadian clocks. Interface Focus 2022; 12:20210087. [PMID: 35464139 PMCID: PMC9010852 DOI: 10.1098/rsfs.2021.0087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/08/2022] [Indexed: 12/18/2022] Open
Abstract
The intercellular interactions between peripheral circadian clocks, located in tissues and organs other than the suprachiasmatic nuclei of the hypothalamus, are still very poorly understood. We propose a theoretical and computational study of the coupling between two or more clocks, using a calibrated, reduced model of the circadian clock to describe some synchronization properties between peripheral cellular clocks. Based on a piecewise linearization of the dynamics of the mutual CLOCK:BMAL1/PER:CRY inactivation term, we suggest a segmentation of the circadian cycle into six stages, to help analyse different types of synchronization between two clocks, including single stage duration, total period and maximal amplitudes. Finally, our model reproduces some recent experimental results on the effects of different regimes of time-restricted feeding in liver circadian clocks of mice.
Collapse
Affiliation(s)
- Odile Burckard
- Université Côte d’Azur, Inria, INRAE, CNRS, Sorbonne Université, Biocore team, Sophia Antipolis, France
| | | | | | - Madalena Chaves
- Université Côte d’Azur, Inria, INRAE, CNRS, Sorbonne Université, Biocore team, Sophia Antipolis, France
| |
Collapse
|
232
|
Fougeray T, Polizzi A, Régnier M, Fougerat A, Ellero-Simatos S, Lippi Y, Smati S, Lasserre F, Tramunt B, Huillet M, Dopavogui L, Salvi J, Nédélec E, Gigot V, Smith L, Naylies C, Sommer C, Haas JT, Wahli W, Duez H, Gourdy P, Gamet-Payrastre L, Benani A, Burnol AF, Loiseau N, Postic C, Montagner A, Guillou H. The hepatocyte insulin receptor is required to program the liver clock and rhythmic gene expression. Cell Rep 2022; 39:110674. [PMID: 35417722 DOI: 10.1016/j.celrep.2022.110674] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/03/2022] [Accepted: 03/23/2022] [Indexed: 12/30/2022] Open
Abstract
Liver physiology is circadian and sensitive to feeding and insulin. Food intake regulates insulin secretion and is a dominant signal for the liver clock. However, how much insulin contributes to the effect of feeding on the liver clock and rhythmic gene expression remains to be investigated. Insulin action partly depends on changes in insulin receptor (IR)-dependent gene expression. Here, we use hepatocyte-restricted gene deletion of IR to evaluate its role in the regulation and oscillation of gene expression as well as in the programming of the circadian clock in the adult mouse liver. We find that, in the absence of IR, the rhythmicity of core-clock gene expression is altered in response to day-restricted feeding. This change in core-clock gene expression is associated with defective reprogramming of liver gene expression. Our data show that an intact hepatocyte insulin receptor is required to program the liver clock and associated rhythmic gene expression.
Collapse
Affiliation(s)
- Tiffany Fougeray
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), UMR1297, INSERM/UPS, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse, France
| | - Arnaud Polizzi
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Marion Régnier
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Yannick Lippi
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Sarra Smati
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), UMR1297, INSERM/UPS, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse, France; Université de Nantes, INSERM, CNRS, CHU Nantes, Institut du Thorax, 44000 Nantes, France
| | - Frédéric Lasserre
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Blandine Tramunt
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), UMR1297, INSERM/UPS, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse, France; Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Université de Toulouse, Toulouse, France
| | - Marine Huillet
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Léonie Dopavogui
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Juliette Salvi
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Université Bourgogne Franche-Comté, Institut Agro Dijon, 21000 Dijon, France
| | - Emmanuelle Nédélec
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Université Bourgogne Franche-Comté, Institut Agro Dijon, 21000 Dijon, France
| | - Vincent Gigot
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Université Bourgogne Franche-Comté, Institut Agro Dijon, 21000 Dijon, France
| | - Lorraine Smith
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Claire Naylies
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Caroline Sommer
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Joel T Haas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France; Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore; Center for Integrative Genomics, University of Lausanne, Le Génopode, 1015 Lausanne, Switzerland
| | - Hélène Duez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), UMR1297, INSERM/UPS, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse, France; Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Université de Toulouse, Toulouse, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Université Bourgogne Franche-Comté, Institut Agro Dijon, 21000 Dijon, France
| | | | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France
| | - Catherine Postic
- Université de Paris, Institut Cochin, CNRS, INSERM, 75014 Paris, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), UMR1297, INSERM/UPS, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse, France.
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, 31027 Toulouse, France.
| |
Collapse
|
233
|
Brown MR, Matveyenko AV. It's What and When You Eat: An Overview of Transcriptional and Epigenetic Responses to Dietary Perturbations in Pancreatic Islets. Front Endocrinol (Lausanne) 2022; 13:842603. [PMID: 35355560 PMCID: PMC8960041 DOI: 10.3389/fendo.2022.842603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/07/2022] [Indexed: 01/07/2023] Open
Abstract
Our ever-changing modern environment is a significant contributor to the increased prevalence of many chronic diseases, and particularly, type 2 diabetes mellitus (T2DM). Although the modern era has ushered in numerous changes to our daily living conditions, changes in "what" and "when" we eat appear to disproportionately fuel the rise of T2DM. The pancreatic islet is a key biological controller of an organism's glucose homeostasis and thus plays an outsized role to coordinate the response to environmental factors to preserve euglycemia through a delicate balance of endocrine outputs. Both successful and failed adaptation to dynamic environmental stimuli has been postulated to occur due to changes in the transcriptional and epigenetic regulation of pathways associated with islet secretory function and survival. Therefore, in this review we examined and evaluated the current evidence elucidating the key epigenetic mechanisms and transcriptional programs underlying the islet's coordinated response to the interaction between the timing and the composition of dietary nutrients common to modern lifestyles. With the explosion of next generation sequencing, along with the development of novel informatic and -omic approaches, future work will continue to unravel the environmental-epigenetic relationship in islet biology with the goal of identifying transcriptional and epigenetic targets associated with islet perturbations in T2DM.
Collapse
Affiliation(s)
- Matthew R. Brown
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Aleksey V. Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
234
|
Manoogian ENC, Chow LS, Taub PR, Laferrère B, Panda S. Time-restricted Eating for the Prevention and Management of Metabolic Diseases. Endocr Rev 2022; 43:405-436. [PMID: 34550357 PMCID: PMC8905332 DOI: 10.1210/endrev/bnab027] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Indexed: 02/08/2023]
Abstract
Time-restricted feeding (TRF, animal-based studies) and time-restricted eating (TRE, humans) are an emerging behavioral intervention approach based on the understanding of the role of circadian rhythms in physiology and metabolism. In this approach, all calorie intake is restricted within a consistent interval of less than 12 hours without overtly attempting to reduce calories. This article will summarize the origin of TRF/TRE starting with concept of circadian rhythms and the role of chronic circadian rhythm disruption in increasing the risk for chronic metabolic diseases. Circadian rhythms are usually perceived as the sleep-wake cycle and dependent rhythms arising from the central nervous system. However, the recent discovery of circadian rhythms in peripheral organs and the plasticity of these rhythms in response to changes in nutrition availability raised the possibility that adopting a consistent daily short window of feeding can sustain robust circadian rhythm. Preclinical animal studies have demonstrated proof of concept and identified potential mechanisms driving TRF-related benefits. Pilot human intervention studies have reported promising results in reducing the risk for obesity, diabetes, and cardiovascular diseases. Epidemiological studies have indicated that maintaining a consistent long overnight fast, which is similar to TRE, can significantly reduce risks for chronic diseases. Despite these early successes, more clinical and mechanistic studies are needed to implement TRE alone or as adjuvant lifestyle intervention for the prevention and management of chronic metabolic diseases.
Collapse
Affiliation(s)
| | - Lisa S Chow
- University of Minnesota, Division of Diabetes, Endocrinology and Metabolism, Minneapolis, Minnesota 55455, USA
| | - Pam R Taub
- University of California, San Diego, Division of Cardiovascular Diseases, Department of Medicine, 9434 Medical Center Drive, La Jolla, California 92037, USA
| | - Blandine Laferrère
- New York Nutrition Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | | |
Collapse
|
235
|
Xiao Y, Gong X, Deng R, Liu W, Yang Y, Wang X, Wang J, Bao J, Shu X. Iron Chelation Remits Memory Deficits Caused by the High-Fat Diet in a Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2022; 86:1959-1971. [DOI: 10.3233/jad-215705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Obesity is a worldwide health problem that has been implicated in many diseases, including Alzheimer’s disease (AD). AD is one of the most common neurodegenerative disorders and is characterized by two pathologies, including extracellular senior plaques composed of amyloid-β (Aβ) and intracellular neurofibrillary tangles (NFTs) consisting of abnormally hyperphosphorylated tau. According to current research, a high-fat diet (HFD) could exacerbate Aβ accumulation, oxidative damage, and cognitive defects in AD mice. However, the accurate role of HFD in the pathogenesis of AD is far more unclear. Objective: To explore the accurate role of HFD in the pathogenesis of AD. Methods: Open Field, Barns Maze, Elevated zero-maze, Contextual fear condition, Tail suspension test, western blotting, immunofluorescence, Fluoro-Jade C Labeling, Perls’ Prussian blue staining, and ELISA were used. Results: HFD caused nonheme iron overload in the brains of APPswe/PS1dE9 (APP/PS1) mice. Furthermore, the administration of M30 (0.5 mg/kg) for iron chelation once every 2 days per os (p.o.) for 1 month remitted memory deficits caused by HFD in APP/PS1 mice. Notably, a variety of hematological parameters in whole blood had no difference after iron chelation. In addition, iron chelation effectively reduced synaptic impairment in hippocampus and neuronal degeneration in cortex in the HFD-fed APP/PS1 mice. Meanwhile, iron chelation decreased Aβ 1–40 and Aβ 1–42 level as well as neuroinflammation in HFD-fed APP/PS1 mice. Conclusion: These data enhance our understanding of how HFD aggravates AD pathology and cognitive impairments and might shed light on future preclinical studies.
Collapse
Affiliation(s)
- Yifan Xiao
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Xiaokang Gong
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Ronghua Deng
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Wei Liu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Youhua Yang
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Xiaochuan Wang
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianzhi Wang
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Bao
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Xiji Shu
- Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| |
Collapse
|
236
|
Polysaccharide Extracts Derived from Defloration Waste of Fruit Pitaya Regulates Gut Microbiota in a Mice Model. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8030108] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Flower thinning is often used during the planting of fruit trees to improve fruit quality and promote large fruit. Flower buds become an agricultural by-product of the planting process. Pitaya (Hylocereus undatus) is a popular fruit in many tropical regions, which is widely cultivated in Southeast Asian countries. Probiotics such as Lactobacillus plantarum have been shown to exhibit an anti-obesity effect by regulating gut microbiota. This study investigated the effect of polysaccharides from pitaya flower buds (PFW) extracted with water on the regulation of gut microbiota and body weight control in mice fed with a high-fat diet. The effects of PFW on the growth of L. plantarum were analyzed and the propagation of L. plantarum was promoted in an aqueous solution containing PFW. In an in vivo study, mice were fed with a high-fat diet supplemented with PFW for 12 weeks; PFW treatment effectively controlled body weight and reduced short bowel syndrome of mice induced by the high-fat diet. Gut microbiota sequencing revealed that Lachnospiraceae and Lactobacillaceae were the main bacteria targeted by PFW. Moreover, transcript analysis demonstrated that PFW alleviated obesity through amino acid metabolism, carbohydrate metabolism, and glycan metabolism. Overall, PFW is a valuable food supplement that can regulate gut microbiota and may have potential to ameliorate the physiological damage caused by a high-fat diet.
Collapse
|
237
|
Malik S, Stokes Iii J, Manne U, Singh R, Mishra MK. Understanding the significance of biological clock and its impact on cancer incidence. Cancer Lett 2022; 527:80-94. [PMID: 34906624 PMCID: PMC8816870 DOI: 10.1016/j.canlet.2021.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
The circadian clock is an essential timekeeper that controls, for humans, the daily rhythm of biochemical, physiological, and behavioral functions. Irregular performance or disruption in circadian rhythms results in various diseases, including cancer. As a factor in cancer development, perturbations in circadian rhythms can affect circadian homeostasis in energy balance, lead to alterations in the cell cycle, and cause dysregulation of chromatin remodeling. However, knowledge gaps remain in our understanding of the relationship between the circadian clock and cancer. Therefore, a mechanistic understanding by which circadian disruption enhances cancer risk is needed. This review article outlines the importance of the circadian clock in tumorigenesis and summarizes underlying mechanisms in the clock and its carcinogenic mechanisms, highlighting advances in chronotherapy for cancer treatment.
Collapse
Affiliation(s)
- Shalie Malik
- Cancer Biology Research and Training, Department of Biological Sciences, Alabama State University, Montgomery, AL, USA; Department of Zoology and Dr. Giri Lal Gupta Institute of Public Health and Public Affairs, University of Lucknow, Lucknow, UP, India
| | - James Stokes Iii
- Department of Biological and Environmental Sciences, Auburn University, Montgomery, AL, USA
| | - Upender Manne
- Departments of Pathology, Surgery and Epidemiology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Manoj K Mishra
- Cancer Biology Research and Training, Department of Biological Sciences, Alabama State University, Montgomery, AL, USA.
| |
Collapse
|
238
|
Early Time-Restricted Feeding Amends Circadian Clock Function and Improves Metabolic Health in Male and Female Nile Grass Rats. MEDICINES (BASEL, SWITZERLAND) 2022; 9:medicines9020015. [PMID: 35200758 PMCID: PMC8877212 DOI: 10.3390/medicines9020015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
Abstract
Lengthening the daily eating period contributes to the onset of obesity and metabolic syndrome. Dietary approaches, including energy restriction and time-restricted feeding, are promising methods to combat metabolic disorders. This study explored the effect of early and late time-restricted feeding (TRF) on weight and adiposity, food consumption, glycemic control, clock gene expression, and liver metabolite composition in diurnal Nile grass rats (NGRs). Adult male and female Nile grass rats were randomly assigned to one of three groups: (1) access to a 60% high-fat (HF) diet ad-libitum (HF-AD), (2) time-restricted access to the HF diet for the first 6 h of the 12 h light/active phase (HF-AM) or (3) the second 6 h of the 12 h light/active phase (HF-PM). Animals remained on their respective protocols for six weeks. TRF reduced total energy consumption and weight gain, and early TRF (HF-AM) reduced fasting blood glucose, restored Per1 expression, and reduced liver lipid levels. Although sex-dependent differences were observed for fat storage and lipid composition, TRF improved metabolic parameters in both male and female NGRs. In conclusion, this study demonstrated that early TRF protocol benefits weight management, improves lipid and glycemic control, and restores clock gene expression in NGRs.
Collapse
|
239
|
Oneda S, Cao S, Haraguchi A, Sasaki H, Shibata S. Wheel-Running Facilitates Phase Advances in Locomotor and Peripheral Circadian Rhythm in Social Jet Lag Model Mice. Front Physiol 2022; 13:821199. [PMID: 35250622 PMCID: PMC8890682 DOI: 10.3389/fphys.2022.821199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
The circadian clock maintains our health by controlling physiological functions. Social jet lag is one factor that can disrupt the body clock. This is caused by the difference in sleeping hours between weekdays when we live according to social time and holidays when we live according to our body clock. The body clock can be altered by exercise, nutrition, and stress, and several studies have reported that these factors can be used to improve a disturbed body clock. Here we focused on exercise and examined whether continuous wheel-running could improve the disordered body clock in a mouse model that mimics social jet lag. The results showed that the wheel-running exercise group showed faster synchronization of the onset of activities on weekdays which had been delayed by social jet lag and the results were even more pronounced in the high-fat diet feeding condition. Also, when the expression rhythms of the clock genes were examined, they experienced a sudden time shift in the advance light condition or social jet lag condition, it was found that the wheel-running group had a higher ability to adapt to the advance direction. Thus, it is possible that the effective inclusion of exercise in human, especially those who eat high-fat foods, life can improve the disordered body clock in terms of social jet lag.
Collapse
|
240
|
Murakami M, Tognini P. Molecular Mechanisms Underlying the Bioactive Properties of a Ketogenic Diet. Nutrients 2022; 14:nu14040782. [PMID: 35215432 PMCID: PMC8879219 DOI: 10.3390/nu14040782] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
The consumption of a high-fat, low-carbohydrate diet (ketogenic diet) has diverse effects on health and is expected to have therapeutic value in neurological disorders, metabolic syndrome, and cancer. Recent studies have shown that a ketogenic diet not only pronouncedly shifts the cellular metabolism to pseudo-starvation, but also exerts a variety of physiological functions on various organs through metabolites that act as energy substrates, signaling molecules, and epigenetic modifiers. In this review, we highlight the latest findings on the molecular mechanisms of a ketogenic diet and speculate on the significance of these functions in the context of the epigenome and microbiome. Unraveling the molecular basis of the bioactive effects of a ketogenic diet should provide solid evidence for its clinical application in a variety of diseases including cancer.
Collapse
Affiliation(s)
- Mari Murakami
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
- Correspondence:
| | - Paola Tognini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
- Laboratory of Biology, Scuola Normale, Superiore, 56126 Pisa, Italy
| |
Collapse
|
241
|
Itoh H, Tanaka M. “Greedy Organs Hypothesis” for sugar and salt in the pathophysiology of non-communicable diseases in relation to sodium-glucose co-transporters in the intestines and the kidney. Metabol Open 2022; 13:100169. [PMID: 35198947 PMCID: PMC8844901 DOI: 10.1016/j.metop.2022.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 11/05/2022] Open
Abstract
Deposition of visceral fat and insulin resistance play central role in the development of non-communicable diseases (NCDs) including obesity, hypertension and type 2 diabetes. However, we shed more light upon the intestines and the kidney as a strong driver of NCDs. Based upon unexpected outcomes of clinical trials using sodium-glucose cotransporter (SGLT) 2 inhibitors to demonstrate their actions for not only body weight reduction and blood glucose fall but also remarkable cardiorenal protection, we speculate that hyperfunction of the intestines and the kidney is one of critical contributing factors for initiation of NCDs. By detecting high amount of glucose and sodium chloride around them by sweet/salt taste sensors, the intestines and the kidney are designed to (re)absorb these nutrients by up-regulating SGLT1 or SGLT2. We designate these hyperfunctioning organs for nutrient uptake as “greedy organs”. The greedy organs can induce NCDs (“greedy organ hypothesis”). SGLTs are regulated by glucose and sodium chloride, and SGLTs or other genes can be “greedy genes.” Regulating factors for greedy organs are renin-angiotensin system, renal sympathetic nervous activity, gut inflammation/microbiota or oxidative stress. Mitigation of organ greediness by SGLT2 inhibitors, ketone bodies, bariatric surgery, and regular lifestyle to keep rhythmicity of biological clock are promising. We propose the concept of “Greedy Organs” hypothesis as a possible cause of NCDs. Clinical implication of greedy kidney is supported by the effect of SGLT2 inhibitors. The significance of greedy intestines is suggested by the effect of bariatric surgery. The intestines and kidney become hyperactive through upregulation of SGLT1 or 2. To mitigate “greedy organs” should be a promising strategy against NCDs.
Collapse
|
242
|
Damgaard MV, Nielsen TS, Basse AL, Chubanava S, Trost K, Moritz T, Dellinger RW, Larsen S, Treebak JT. Intravenous nicotinamide riboside elevates mouse skeletal muscle NAD+ without impacting respiratory capacity or insulin sensitivity. iScience 2022; 25:103863. [PMID: 35198907 PMCID: PMC8844641 DOI: 10.1016/j.isci.2022.103863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 11/30/2022] Open
Abstract
In clinical trials, oral supplementation with nicotinamide riboside (NR) fails to increase muscle mitochondrial respiratory capacity and insulin sensitivity but also does not increase muscle NAD+ levels. This study tests the feasibility of chronically elevating skeletal muscle NAD+ in mice and investigates the putative effects on mitochondrial respiratory capacity, insulin sensitivity, and gene expression. Accordingly, to improve bioavailability to skeletal muscle, we developed an experimental model for administering NR repeatedly through a jugular vein catheter. Mice on a Western diet were treated with various combinations of NR, pterostilbene (PT), and voluntary wheel running, but the metabolic effects of NR and PT treatment were modest. We conclude that the chronic elevation of skeletal muscle NAD+ by the intravenous injection of NR is possible but does not affect muscle respiratory capacity or insulin sensitivity in either sedentary or physically active mice. Our data have implications for NAD+ precursor supplementation regimens. A model was developed for daily intravenous NR injections Intravenous NR stably elevates NAD+ of skeletal muscle and adipose, but not liver Voluntary running and intravenous NR synergize to boost mouse skeletal muscle NAD+ NR did not impact skeletal muscle insulin sensitivity or respiratory capacity
Collapse
Affiliation(s)
- Mads V. Damgaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Thomas S. Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Astrid L. Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sabina Chubanava
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kajetan Trost
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | | | - Steen Larsen
- Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Jonas T. Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Corresponding author
| |
Collapse
|
243
|
Shon J, Han Y, Park YJ. Effects of Dietary Fat to Carbohydrate Ratio on Obesity Risk Depending on Genotypes of Circadian Genes. Nutrients 2022; 14:nu14030478. [PMID: 35276838 PMCID: PMC8838281 DOI: 10.3390/nu14030478] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
Although the impacts of macronutrients and the circadian clock on obesity have been reported, the interactions between macronutrient distribution and circadian genes are unclear. The aim of this study was to explore macronutrient intake patterns in the Korean population and associations between the patterns and circadian gene variants and obesity. After applying the criteria, 5343 subjects (51.6% male, mean age 49.4 ± 7.3 years) from the Korean Genome and Epidemiology Study data and nine variants in seven circadian genes were analyzed. We defined macronutrient intake patterns by tertiles of the fat to carbohydrate ratio (FC). The very low FC (VLFC) was associated with a higher risk of obesity than the optimal FC (OFC). After stratification by the genotypes of nine variants, the obesity risk according to the patterns differed by the variants. In the female VLFC, the major homozygous allele of CLOCK rs11932595 and CRY1 rs3741892 had a higher abdominal obesity risk than those in the OFC. The GG genotype of PER2 rs2304672 in the VLFC showed greater risks for obesity and abdominal obesity. In conclusion, these findings suggest that macronutrient intake patterns were associated with obesity susceptibility, and the associations were different depending on the circadian clock genotypes of the CLOCK, PER2, and CRY1 loci.
Collapse
Affiliation(s)
- Jinyoung Shon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea; (J.S.); (Y.H.)
| | - Yerim Han
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea; (J.S.); (Y.H.)
- Graduate Program in System Health Science & Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Yoon Jung Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea; (J.S.); (Y.H.)
- Graduate Program in System Health Science & Engineering, Ewha Womans University, Seoul 03760, Korea
- Correspondence: ; Tel.: +82-2-3277-6533
| |
Collapse
|
244
|
A Growing Link between Circadian Rhythms, Type 2 Diabetes Mellitus and Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23010504. [PMID: 35008933 PMCID: PMC8745289 DOI: 10.3390/ijms23010504] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) patients are at a higher risk of developing Alzheimer’s disease (AD). Mounting evidence suggests the emerging important role of circadian rhythms in many diseases. Circadian rhythm disruption is considered to contribute to both T2DM and AD. Here, we review the relationship among circadian rhythm disruption, T2DM and AD, and suggest that the occurrence and progression of T2DM and AD may in part be associated with circadian disruption. Then, we summarize the promising therapeutic strategies targeting circadian dysfunction for T2DM and AD, including pharmacological treatment such as melatonin, orexin, and circadian molecules, as well as non-pharmacological treatments like light therapy, feeding behavior, and exercise.
Collapse
|
245
|
Tuning up an aged clock: Circadian clock regulation in metabolism and aging. TRANSLATIONAL MEDICINE OF AGING 2022. [DOI: 10.1016/j.tma.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
246
|
Kawai M. Disruption of the circadian rhythms and its relationship with pediatric obesity. Pediatr Int 2022; 64:e14992. [PMID: 34525248 DOI: 10.1111/ped.14992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/23/2021] [Accepted: 09/13/2021] [Indexed: 11/26/2022]
Abstract
The circadian clock system is an evolutionarily conserved system by which organisms adapt their metabolic activities to environmental inputs, including nutrient availability. The disruption of this system has been pathogenically linked to the disintegration of metabolic homeostasis, leading to the development of metabolic complications, including obesity. Lifestyle factors that disrupt this system have been found to be associated with the development of metabolic disorder, which is most evidenced by the finding that shift workers are at an increased risk of developing various disorders, such as obesity and obesity-related complications. Lifestyle factors that contribute to a misalignment between the internal clock system and environmental rhythms have also been identified in children. A short sleep duration and skipping breakfast are prevalent in children and there is mounting evidence that these factors are associated with an increased risk of pediatric obesity; however, the underlying mechanisms have not yet been elucidated in detail. Our current understanding of the impact of lifestyle factors that cause a misalignment between the internal clock system and environmental rhythms on the development of pediatric obesity is summarized herein, with a discussion of potential mechanistic factors.
Collapse
Affiliation(s)
- Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan.,Department of Gastroenterology, Nutrition, and Endocrinology, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| |
Collapse
|
247
|
Chaix A. Time-Restricted Feeding and Caloric Restriction: Two Feeding Regimens at the Crossroad of Metabolic and Circadian Regulation. Methods Mol Biol 2022; 2482:329-340. [PMID: 35610437 PMCID: PMC9254535 DOI: 10.1007/978-1-0716-2249-0_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In addition to diet quality and quantity, the "timing" of food intake recently emerged as a third key parameter in nutritional and metabolic health. The link between nutrition timing and metabolic homeostasis is in part due to the regulation of daily feeding:fasting cycles and metabolic pathways by the circadian clock. Preclinical feeding regimen studies in rodents are invaluable to further define the modalities of this relationship and get a better understanding of its mechanistic underpinnings. Time-restricted feeding (TRF) and caloric restriction (CR) are examples of feeding regimen at the crossroads of metabolic and circadian regulation. Here we propose methods to implement TRF and CR highlighting the parameters that are relevant to the study of circadian and metabolic health. We also provide methods to determine their impact on the output of the circadian clock by analyzing diurnal expression profiles using 24 h time-series collection as well as their impact on metabolic homeostasis using a glucose tolerance test (GTT).
Collapse
Affiliation(s)
- Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
248
|
Abstract
Cross-talk between peripheral tissues is essential to ensure the coordination of nutrient intake with disposition during the feeding period, thereby preventing metabolic disease. This mini-review considers the interactions between the key peripheral tissues that constitute the metabolic clock, each of which is considered in a separate mini-review in this collation of articles published in Endocrinology in 2020 and 2021, by Martchenko et al (Circadian rhythms and the gastrointestinal tract: relationship to metabolism and gut hormones); Alvarez et al (The microbiome as a circadian coordinator of metabolism); Seshadri and Doucette (Circadian regulation of the pancreatic beta cell); McCommis et al (The importance of keeping time in the liver); Oosterman et al (The circadian clock, shift work, and tissue-specific insulin resistance); and Heyde et al (Contributions of white and brown adipose tissues to the circadian regulation of energy metabolism). The use of positive- and negative-feedback signals, both hormonal and metabolic, between these tissues ensures that peripheral metabolic pathways are synchronized with the timing of food intake, thus optimizing nutrient disposition and preventing metabolic disease. Collectively, these articles highlight the critical role played by the circadian clock in maintaining metabolic homeostasis.
Collapse
Affiliation(s)
- Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8Canada
- Correspondence: P. L. Brubaker, PhD, Departments of Physiology and Medicine, University of Toronto, Medical Sciences Bldg, Rm 3366, 1 King’s College Cir, Toronto, ON M5S 1A8, Canada.
| | | |
Collapse
|
249
|
Abstract
Isolation of primary hepatocytes and culturing these cells ex vivo provides a powerful platform to model liver physiology in vivo. Primary hepatocytes can be cultured for several days, the circadian clock can be synchronized, and these primary cells can be utilized for functional gene regulation analysis and metabolic studies. In this chapter, we describe detailed methodology for isolation of viable primary hepatocytes, techniques for culturing these cells, methods for synchronization of the circadian clock, transfection and luciferase reporter analysis, as well as glucose production assays as a functional readout of metabolic state.
Collapse
Affiliation(s)
- Sung Kook Chun
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine (UCI), Irvine, CA, USA
| | - Selma Masri
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine (UCI), Irvine, CA, USA.
| |
Collapse
|
250
|
Fan X, Chen D, Wang Y, Tan Y, Zhao H, Zeng J, Li Y, Guo X, Qiu H, Gu Y. Light intensity alters the effects of light-induced circadian disruption on glucose and lipid metabolism in mice. Am J Physiol Endocrinol Metab 2022; 322:E1-E9. [PMID: 34719945 DOI: 10.1152/ajpendo.00025.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Circadian disruption induced by rotating light cycles has been linked to metabolic disorders. However, how the interaction of light intensity and light cycle affects metabolism under different diets remains to be explored. Eighty mice were first randomly stratified into the low-fat diet (LFD, n = 40) or high-fat diet (HFD, n = 40) groups. Each group was further randomly subdivided into four groups (n = 8-12 per group) in terms of different light intensities [lower (LI, 78 lx) or higher intensity (HI, 169 lx)] and light cycles [12-h light:12-h dark cycle or circadian-disrupting (CD) light cycle consisting of repeated 6-h light phase advancement]. Body weight was measured weekly. At the end of the 16-wk experiment, mice were euthanized for serum and pathological analysis. Glucose and insulin tolerance tests were performed during the last 2 wk. The CD cycle increased body weight gain, adipocyte area, glucose intolerance, and insulin resistance of LFD as well as HFD mice under HI but not LI condition. Moreover, the serum and hepatic triglyceride levels increased with LFD-HI treatment, regardless of light cycle. In addition, the CD cycle improved lipid and glucose metabolism under HFD-LI condition. In summary, the detrimental effects of the CD cycle on metabolism were alleviated under LI condition, especially in HFD mice. These results indicate that modulating light intensity is a potential strategy to prevent the negative metabolic consequences associated with jet lag or shift work.NEW & NOTEWORTHY Glucose and lipid homeostasis is altered by the CD cycles in a light-intensity-dependent manner. Lower-intensity light reverses the negative metabolic effects of the CD cycles, especially under HFD feeding. The interaction of light intensity and light cycle on metabolism is independent of energy intake and eating pattern. Glucose metabolic disorders caused by rotating light cycles occur along with compensatory β-cell mass expansion.
Collapse
Affiliation(s)
- Xiaojing Fan
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
- Department of Endocrinology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Defu Chen
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Ying Wang
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yizhou Tan
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongyou Zhao
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Jing Zeng
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yunqi Li
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xianghuan Guo
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haixia Qiu
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ying Gu
- Department of Laser Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
- Precision Laser Medical Diagnosis and Treatment Innovation Unit, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| |
Collapse
|