1
|
Ratwani M, Bisht S, Prakash S. Association between sleep disturbance and metabolic dysfunctions in adipose tissue: Insights into melatonin's role. Biochem Biophys Res Commun 2025; 770:151978. [PMID: 40378618 DOI: 10.1016/j.bbrc.2025.151978] [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: 01/23/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
The increased prevalence of sleep disturbances in modern society is frequently linked to various metabolic disorders, including insulin resistance, obesity, hypertension, fatty liver disease, and cardiometabolic complications. Melatonin, a pineal gland-secreted neurohormone, plays a pivotal role in maintaining the circadian rhythm. It is involved in regulating adipose tissue development, lipid accumulation, browning of white adipose tissue, and activation of brown adipose tissue. The adipose tissue is a dynamic endocrine organ that secretes hormones and cytokines. Recent research has highlighted the significant role of melatonin in the modulation of lipid metabolism, adipogenesis, and thermogenesis in adipose tissues. Circadian rhythms are important in synchronizing metabolic functions with environmental cues, such as light and dark, feeding-fasting states, etc. Irregular sleep patterns, shift work, and exposure to artificial light at night disrupt these rhythms, affecting circadian regulation and compromising metabolic health. Melatonin imbalance due to sleep disturbances results in metabolic dysfunction, increased fat storage, and adipose tissue inflammation. As circadian rhythm and melatonin are both related, a change in circadian rhythm affects the physiology of adipose tissues thereby precipitating metabolic complications through melatonin signaling. This study attempted to understand the mechanisms by which melatonin influences adipose tissue activity, highlighting the role of circadian rhythms in this process. This will enable the development of melatonin-based therapies to mitigate the adverse effects of chronobiological disturbances on the physiology of adipose tissue. Understanding these interactions will provide novel insights for combating obesity and related metabolic conditions.
Collapse
Affiliation(s)
- Mishthi Ratwani
- Amity Institute of Pharmacy, Amity University, Sector 125, Uttar Pradesh, Lucknow, 201313, India
| | - Shradha Bisht
- College of Pharmacy, Shivalik Campus, Dehradun, 248197, Uttarakhand, India
| | - Swati Prakash
- Amity Institute of Pharmacy, Amity University, Sector 125, Uttar Pradesh, Lucknow, 201313, India.
| |
Collapse
|
2
|
Cano-Barquilla P, Jiménez-Ortega V, Fernández-Mateos P, Virto L, Maldonado Bautista E, Perez-Miguelsanz J, Esquifino AI. Daily Lipolysis Gene Expression in Male Rat Mesenteric Adipose Tissue: Obesity and Melatonin Effects. Int J Mol Sci 2025; 26:577. [PMID: 39859293 PMCID: PMC11765279 DOI: 10.3390/ijms26020577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Melatonin is involved in various functions such as the timing of circadian rhythms, energy metabolism, and body mass gain in experimental animals. However, its effects on adipose tissue lipid metabolism are still unclear. This study analyzes the effects of melatonin on the relative gene expression of lipolytic proteins in rat mesenteric adipose tissue and free fatty acid (FFA) and glycerol plasma levels of male Wistar rats fed a high-fat (HFD) or maintenance diet. Four experimental groups were established: control, obese, and control or obese plus 2.3 mg/kg/day of melatonin in tap water. After 11 weeks, animals were sacrificed at different times throughout a 24 h cycle, and mesenteric adipose tissue and plasma samples were collected and analyzed. Cgi58, Perilipin, and Dgat1 gene expression, as well as FFA and glycerol concentrations, showed rhythm patterns in the control group. HFD disrupted those rhythm patterns and increased FFA and glycerol concentrations during the dark photoperiod. In both melatonin-treated groups, almost all analyzed genes showed circadian patterns. Notably, melatonin significantly prevented the increase in FFA levels during the dark photoperiod in obese rats (obese group: ~1100 mM vs. obese + melatonin group: ~600 μM, similar to control levels). However, the rhythmic pattern observed in control animals was not sustained. According to our results, melatonin could regulate circadian gene transcription of mesenteric adipose tissue lipolysis proteins. The effect of melatonin on preventing elevated FFA plasma levels associated with high-fat diet intake warrants further investigation.
Collapse
Affiliation(s)
- Pilar Cano-Barquilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| | - Vanesa Jiménez-Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| | - Pilar Fernández-Mateos
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Leire Virto
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Anatomía y Embriología, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Estela Maldonado Bautista
- Departamento de Anatomía y Embriología, Faculta de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Juliana Perez-Miguelsanz
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Anatomía y Embriología, Faculta de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Ana I. Esquifino
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| |
Collapse
|
3
|
Rabadán-Chávez G, Díaz de la Garza RI, Jacobo-Velázquez DA. White adipose tissue: Distribution, molecular insights of impaired expandability, and its implication in fatty liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166853. [PMID: 37611674 DOI: 10.1016/j.bbadis.2023.166853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
We are far behind the 2025 World Health Organization (WHO) goal of a zero increase in obesity. Close to 360 million people in Latin America and the Caribbean are overweight, with the highest rates observed in the Bahamas, Mexico, and Chile. To achieve relevant progress against the obesity epidemic, scientific research is essential to establish uniform practices in the study of obesity pathophysiology (using pre-clinical and clinical models) that ensure accuracy, reproducibility, and transcendent outcomes. The present review focuses on relevant aspects of white adipose tissue (WAT) expansion, underlying mechanisms of inefficient expandability, and its repercussion in ectopic lipid accumulation in the liver during nutritional abundance. In addition, we highlight the potential role of disrupted circadian rhythm in WAT metabolism. Since genetic factors also play a key role in determining an individual's predisposition to weight gain, we describe the most relevant genes associated with obesity in the Mexican population, underlining that most of them are related to appetite control.
Collapse
Affiliation(s)
- Griselda Rabadán-Chávez
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico
| | - Rocío I Díaz de la Garza
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico.
| | - Daniel A Jacobo-Velázquez
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Guadalajara, Av. General Ramon Corona 2514, C.P. 45201 Zapopan, Jalisco, Mexico.
| |
Collapse
|
4
|
Zambrano C, Kulyté A, Luján J, Rivero-Gutierrez B, Sánchez de Medina F, Martínez-Augustin O, Ryden M, Scheer FAJL, Garaulet M. Habitual nappers and non-nappers differ in circadian rhythms of LIPE expression in abdominal adipose tissue explants. Front Endocrinol (Lausanne) 2023; 14:1166961. [PMID: 37361522 PMCID: PMC10289256 DOI: 10.3389/fendo.2023.1166961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Background and purpose Napping is a widespread practice worldwide and has in recent years been linked to increased abdominal adiposity. Lipase E or LIPE encodes the protein hormone-sensitive lipase (HSL), an enzyme that plays an important role in lipid mobilization and exhibits a circadian expression rhythm in human adipose tissue. We hypothesized that habitual napping may impact the circadian expression pattern of LIPE, which in turn may attenuate lipid mobilization and induce abdominal fat accumulation. Methods Abdominal adipose tissue explants from participants with obesity (n = 17) were cultured for a 24-h duration and analyzed every 4 h. Habitual nappers (n = 8) were selected to match non-nappers (n = 9) in age, sex, BMI, adiposity, and metabolic syndrome traits. Circadian LIPE expression rhythmicity was analyzed using the cosinor method. Results Adipose tissue explants exhibited robust circadian rhythms in LIPE expression in non-nappers. In contrast, nappers had a flattened rhythm. LIPE amplitude was decreased in nappers as compared with non-nappers (71% lower). The decrease in amplitude among nappers was related to the frequency of napping (times per week) where a lower rhythm amplitude was associated with a higher napping frequency (r = -0.80; P = 0.018). Confirmatory analyses in the activity of LIPE's protein (i.e., HSL) also showed a significant rhythm in non-nappers, whereas significance in the activity of HSL was lost among nappers. Conclusion Our results suggest that nappers display dysregulated circadian LIPE expression as well as dysregulated circadian HSL activity, which may alter lipid mobilization and contribute to increased abdominal obesity in habitual nappers.
Collapse
Affiliation(s)
- Carolina Zambrano
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca-Universidad de Murcia (UMU), University Clinical Hospital, Murcia, Spain
| | - Agné Kulyté
- Endocrinology Unit, Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Juán Luján
- General Surgery Service, Hospital Quirón salud, Murcia, Spain
| | - Belén Rivero-Gutierrez
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Fermín Sánchez de Medina
- Department of Pharmacology, Centro de Investigación Biomédica en Red (CIBERed), Ibs Granada, Universidad de Granada, Granada, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology 2, Centro de Investigación Biomédica en Red, Enfermedades Hepáticas y Digestivas (CIBERehd), Ibs Granada, Instituto de Nutrición y Tecnología de los Alimentos (INYTA) José Mataix, Universidad de Granada, Granada, Spain
| | - Mikael Ryden
- Endocrinology Unit, Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Frank A. J. L. Scheer
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Marta Garaulet
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca-Universidad de Murcia (UMU), University Clinical Hospital, Murcia, Spain
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
5
|
Meyer-Lindemann U, Moggio A, Dutsch A, Kessler T, Sager HB. The Impact of Exercise on Immunity, Metabolism, and Atherosclerosis. Int J Mol Sci 2023; 24:3394. [PMID: 36834808 PMCID: PMC9967592 DOI: 10.3390/ijms24043394] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Physical exercise represents an effective preventive and therapeutic strategy beneficially modifying the course of multiple diseases. The protective mechanisms of exercise are manifold; primarily, they are elicited by alterations in metabolic and inflammatory pathways. Exercise intensity and duration strongly influence the provoked response. This narrative review aims to provide comprehensive up-to-date insights into the beneficial effects of physical exercise by illustrating the impact of moderate and vigorous exercise on innate and adaptive immunity. Specifically, we describe qualitative and quantitative changes in different leukocyte subsets while distinguishing between acute and chronic exercise effects. Further, we elaborate on how exercise modifies the progression of atherosclerosis, the leading cause of death worldwide, representing a prime example of a disease triggered by metabolic and inflammatory pathways. Here, we describe how exercise counteracts causal contributors and thereby improves outcomes. In addition, we identify gaps that still need to be addressed in the future.
Collapse
Affiliation(s)
- Ulrike Meyer-Lindemann
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
| | - Alexander Dutsch
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK e.V. (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
6
|
Khazaal AQ, Haque N, Krager CR, Krager SL, Chambers C, Wilber A, Tischkau SA. Aryl hydrocarbon receptor affects circadian-regulated lipolysis through an E-Box-dependent mechanism. Mol Cell Endocrinol 2023; 559:111809. [PMID: 36283500 PMCID: PMC10509633 DOI: 10.1016/j.mce.2022.111809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
An internal circadian clock regulates timing of systemic energy homeostasis. The central clock in the hypothalamic suprachiasmatic nucleus (SCN) directs local clocks in peripheral tissues such as liver, muscle, and adipose tissue to synchronize metabolism with food intake and rest/activity cycles. Aryl hydrocarbon receptor (AhR) interacts with the molecular circadian clockworks. Activation of AhR dampens rhythmic expression of core clock genes, which may lead to metabolic dysfunction. Given the importance of appropriately-timed adipose tissue function to regulation of energy homeostasis, this study focused on mechanisms by which AhR may influence clock-controlled adipose tissue activity. We hypothesized that AhR activation in adipose tissue would impair lipolysis by dampening adipose rhythms, leading to a decreased lipolysis rate during fasting, and subsequently, altered serum glucose concentrations. Levels of clock gene and lipolysis gene transcripts in mouse mesenchymal stem cells (BMSCs) differentiated into mature adipocytes were suppressed by the AhR agonist β-napthoflavone (BNF), in an AhR dependent manner. BNF altered rhythms of core clock gene and lipolysis gene transcripts in C57bl6/J mice. BNF reduced serum free fatty acids, glycerol and liver glycogen. Chromatin immunoprecipitation indicated that BNF increased binding of AhR to E-Box elements in clock gene and lipolysis gene promoters. These data establish a link between AhR activation and impaired lipolysis, specifically by altering adipose tissue rhythmicity. In response to the decreased available energy from impaired lipolysis, the body increases glycogenolysis, thereby degrading more glycogen to provide necessary energy.
Collapse
Affiliation(s)
- Ali Qasim Khazaal
- Biotechnology Department, College of Science, University of Baghdad, Baghdad, Iraq; Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Nazmul Haque
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Callie R Krager
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Stacey L Krager
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Christopher Chambers
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Shelley A Tischkau
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA; Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.
| |
Collapse
|
7
|
The circadian transcription factor ARNTL2 is regulated by weight-loss interventions in human white adipose tissue and inhibits adipogenesis. Cell Death Dis 2022; 8:443. [PMID: 36329012 PMCID: PMC9633602 DOI: 10.1038/s41420-022-01239-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Misalignment of physiological circadian rhythms promotes obesity which is characterized by white adipose tissue (WAT) expansion. Differentiation of Adipose stem/progenitor cells (ASCs) contributes to WAT increase but the importance of the cellular clock in this process is incompletely understood. In the present study, we reveal the role of the circadian transcription factor Aryl hydrocarbon receptor nuclear translocator-like 2 (ARNTL2) in human ASCs, isolated from subcutaneous (s)WAT samples of patients undergoing routine elective plastic abdominal surgery. We show that circadian synchronization by serum-shock or stimulation with adipogenic stimuli leads to a different expression pattern of ARNTL2 relative to its well-studied paralogue ARNTL1. We demonstrate that ARNTL2 mRNA is downregulated in ASCs upon weight-loss (WL) whereas ARNTL2 protein is rapidly induced in the course of adipogenic differentiation and highly abundant in adipocytes. ARNTL2 protein is maintained in ASCs cooperatively by mechanistic Target of Rapamycin (mTOR) and Mitogen-activated Protein Kinase (MAPK) signalling pathways while ARNTL2 functions as an inhibitor on both circuits, leading to a feedback mechanism. Consistently, ectopic overexpression of ARNTL2 repressed adipogenesis by facilitating the degradation of ARNTL1, inhibition of Kruppel-Like Factor 15 (KLF15) gene expression and down-regulation of the MAPK-CCAAT/enhancer-binding protein β (C/EBPβ) axis. Western blot analysis of sWAT samples from normal-weight, obese and WL donors revealed that ARNTL2 protein was solely elevated by WL compared to ARNTL1 which underscores unique functions of both transcription factors. In conclusion, our study reveals ARNTL2 to be a WL-regulated inhibitor of adipogenesis which might provide opportunities to develop strategies to ameliorate obesity.
Collapse
|
8
|
Meriin AB, Zaarur N, Roy D, Kandror KV. Egr1 plays a major role in the transcriptional response of white adipocytes to insulin and environmental cues. Front Cell Dev Biol 2022; 10:1003030. [PMID: 36246998 PMCID: PMC9554007 DOI: 10.3389/fcell.2022.1003030] [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: 07/25/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
It is believed that insulin regulates metabolic functions of white adipose tissue primarily at the post-translational level via the PI3K-Akt-mediated pathway. Still, changes in transcription also play an important role in the response of white adipocytes to insulin and environmental signals. One transcription factor that is dramatically and rapidly induced in adipocytes by insulin and nutrients is called Early Growth Response 1, or Egr1. Among other functions, it directly binds to promoters of leptin and ATGL stimulating the former and inhibiting the latter. Furthermore, expression of Egr1 in adipocytes demonstrates cell autonomous circadian pattern suggesting that Egr1 not only mediates the effect of insulin and nutrients on lipolysis and leptin production but also, coordinates insulin action with endogenous circadian rhythms of adipose tissue.
Collapse
Affiliation(s)
- A. B. Meriin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - N. Zaarur
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - D. Roy
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - K. V. Kandror
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
- *Correspondence: K. V. Kandror,
| |
Collapse
|
9
|
The Shades of Grey in Adipose Tissue Reprogramming. Biosci Rep 2022; 42:230844. [PMID: 35211733 PMCID: PMC8905306 DOI: 10.1042/bsr20212358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
The adipose tissue (AT) has a major role in contributing to obesity-related pathologies through regulating systemic immunometabolism. The pathogenicity of the AT is underpinned by its remarkable plasticity to be reprogrammed during obesity, in the perspectives of tissue morphology, extracellular matrix (ECM) composition, angiogenesis, immunometabolic homoeostasis and circadian rhythmicity. Dysregulation in these features escalates the pathogenesis conferred by this endometabolic organ. Intriguingly, the potential to be reprogrammed appears to be an Achilles’ heel of the obese AT that can be targeted for the management of obesity and its associated comorbidities. Here, we provide an overview of the reprogramming processes of white AT (WAT), with a focus on their dynamics and pleiotropic actions over local and systemic homoeostases, followed by a discussion of potential strategies favouring therapeutic reprogramming. The potential involvement of AT remodelling in the pathogenesis of COVID-19 is also discussed.
Collapse
|
10
|
Shiraishi R, Morita S, Goto Y, Mizoguchi Y, Nakamura W, Nakamura TJ. Diurnal variations of triglyceride accumulation in mouse and bovine adipocyte-derived cell lines. Anim Sci J 2022; 93:e13802. [PMID: 36562279 DOI: 10.1111/asj.13802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
Several studies have suggested a strong interaction between the circadian clock and lipid metabolism in mammals. The circadian clock is driven by endogenous cyclic gene expression patterns, commonly referred to as clock genes, and transcription-translation negative feedback loops. Clock genes regulate the transcription of some lipid metabolism-related genes; however, the relationship between the circadian clock and triglyceride (TG) accumulation at the cellular level remains unclear. Here, we evaluated rhythms of intracellular TG accumulation levels as well as the expression of clock genes and lipid metabolism-related genes for 54 h in mouse and bovine adipose-derived cell cultures. To the best of our knowledge, this study represents the first report demonstrating that TG accumulation exhibits diurnal variations, with the pattern differing among cell types. Furthermore, we found that expression of clock genes and corresponding lipid metabolism-related genes exhibited circadian rhythms. Our results suggest that the cellular clock regulates lipid metabolism-related genes to relate circadian rhythms of TG accumulation in each cell type. We anticipate that the amount of fat stored depends on the timing of the supply of glucose-the precursor of fat. The findings of this study will contribute to the advancement of chrono-nutrition.
Collapse
Affiliation(s)
- Rena Shiraishi
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Satomi Morita
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Yoshikuni Goto
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Yasushi Mizoguchi
- Laboratory of Animal Genetics, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Wataru Nakamura
- Department of Oral-Chrono Physiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| |
Collapse
|
11
|
García-Aviles JE, Méndez-Hernández R, Guzmán-Ruiz MA, Cruz M, Guerrero-Vargas NN, Velázquez-Moctezuma J, Hurtado-Alvarado G. Metabolic Disturbances Induced by Sleep Restriction as Potential Triggers for Alzheimer's Disease. Front Integr Neurosci 2021; 15:722523. [PMID: 34539357 PMCID: PMC8447653 DOI: 10.3389/fnint.2021.722523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/26/2021] [Indexed: 01/15/2023] Open
Abstract
Sleep has a major role in learning, memory consolidation, and metabolic function. Although it is known that sleep restriction increases the accumulation of amyloid β peptide (Aβ) and the risk to develop Alzheimer's disease (AD), the mechanism behind these effects remains unknown. In this review, we discuss how chronic sleep restriction induces metabolic and cognitive impairments that could result in the development of AD in late life. Here, we integrate evidence regarding mechanisms whereby metabolic signaling becomes disturbed after short or chronic sleep restriction in the context of cognitive impairment, particularly in the accumulation of Aβ in the brain. We also discuss the role of the blood-brain barrier in sleep restriction with an emphasis on the transport of metabolic signals into the brain and Aβ clearance. This review presents the unexplored possibility that the alteration of peripheral metabolic signals induced by sleep restriction, especially insulin resistance, is responsible for cognitive deficit and, subsequently, implicated in AD development.
Collapse
Affiliation(s)
- Jesús Enrique García-Aviles
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Cruz
- Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Hospital de Especialidades, Unidad de Investigación Médica en Bioquímica, Mexico City, Mexico
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Javier Velázquez-Moctezuma
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Gabriela Hurtado-Alvarado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
12
|
Tuvia N, Pivovarova-Ramich O, Murahovschi V, Lück S, Grudziecki A, Ost AC, Kruse M, Nikiforova VJ, Osterhoff M, Gottmann P, Gögebakan Ö, Sticht C, Gretz N, Schupp M, Schürmann A, Rudovich N, Pfeiffer AFH, Kramer A. Insulin Directly Regulates the Circadian Clock in Adipose Tissue. Diabetes 2021; 70:1985-1999. [PMID: 34226282 DOI: 10.2337/db20-0910] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 06/15/2021] [Indexed: 11/13/2022]
Abstract
Adipose tissue (AT) is a key metabolic organ which functions are rhythmically regulated by an endogenous circadian clock. Feeding is a "zeitgeber" aligning the clock in AT with the external time, but mechanisms of this regulation remain largely unclear. We tested the hypothesis that postprandial changes of the hormone insulin directly entrain circadian clocks in AT and investigated a transcriptional-dependent mechanism of this regulation. We analyzed gene expression in subcutaneous AT (SAT) of obese subjects collected before and after the hyperinsulinemic-euglycemic clamp or control saline infusion (SC). The expressions of core clock genes PER2, PER3, and NR1D1 in SAT were differentially changed upon insulin and saline infusion, suggesting insulin-dependent clock regulation. In human stem cell-derived adipocytes, mouse 3T3-L1 cells, and AT explants from mPer2Luc knockin mice, insulin induced a transient increase of the Per2 mRNA and protein expression, leading to the phase shift of circadian oscillations, with similar effects for Per1 Insulin effects were dependent on the region between -64 and -43 in the Per2 promoter but not on CRE and E-box elements. Our results demonstrate that insulin directly regulates circadian clocks in AT and isolated adipocytes, thus representing a primary mechanism of feeding-induced AT clock entrainment.
Collapse
Affiliation(s)
- Neta Tuvia
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Laboratory of Chronobiology, Berlin, Germany
| | - Olga Pivovarova-Ramich
- Reseach Group Molecular Nutritional Medicine, Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Veronica Murahovschi
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Sarah Lück
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Laboratory of Chronobiology, Berlin, Germany
| | - Astrid Grudziecki
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Laboratory of Chronobiology, Berlin, Germany
| | - Anne-Catrin Ost
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Michael Kruse
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Victoria J Nikiforova
- Department of Molecular Physiology, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Martin Osterhoff
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Pascal Gottmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Özlem Gögebakan
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Schupp
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Berlin, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Natalia Rudovich
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Spital Bülach, Bülach, Switzerland
| | - Andreas F H Pfeiffer
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Achim Kramer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Laboratory of Chronobiology, Berlin, Germany
| |
Collapse
|
13
|
Hernández-González T, González-Barrio R, Escobar C, Madrid JA, Periago MJ, Collado MC, Scheer FAJL, Garaulet M. Timing of chocolate intake affects hunger, substrate oxidation, and microbiota: A randomized controlled trial. FASEB J 2021; 35:e21649. [PMID: 34164846 DOI: 10.1096/fj.202002770rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 11/11/2022]
Abstract
Eating chocolate in the morning or in the evening/at night, may differentially affect energy balance and impact body weight due to changes in energy intake, substrate oxidation, microbiota (composition/function), and circadian-related variables. In a randomized controlled trial, postmenopausal females (n = 19) had 100 g of chocolate in the morning (MC), in the evening/at night (EC), or no chocolate (N) for 2 weeks and ate any other food ad libitum. Our results show that 14 days of chocolate intake did not increase body weight. Chocolate consumption decreased hunger and desire for sweets (P < .005), and reduced ad libitum energy intake by ~300 kcal/day during MC and ~150 kcal/day during EC (P = .01), but did not fully compensate for the extra energy contribution of chocolate (542 kcal/day). EC increased physical activity by +6.9%, heat dissipation after meals +1.3%, and carbohydrate oxidation by +35.3% (P < .05). MC reduced fasting glucose (4.4%) and waist circumference (-1.7%) and increased lipid oxidation (+25.6%). Principal component analyses showed that both timings of chocolate intake resulted in differential microbiota profiles and function (P < .05). Heat map of wrist temperature and sleep records showed that EC induced more regular timing of sleep episodes with lower variability of sleep onset among days than MC (60 min vs 78 min; P = .028). In conclusion, having chocolate in the morning or in the evening/night results in differential effects on hunger and appetite, substrate oxidation, fasting glucose, microbiota (composition and function), and sleep and temperature rhythms. Results highlight that the "when" we eat is a relevant factor to consider in energy balance and metabolism.
Collapse
Affiliation(s)
- Teresa Hernández-González
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital, Murcia, Spain
| | - Rocío González-Barrio
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital, Murcia, Spain.,Department of Food Technology, Food Science and Nutrition, Faculty of Veterinary Sciences, Regional Campus of Internacional Excellence, University of Murcia, Murcia, Spain
| | - Carolina Escobar
- Department of Anatomy, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Juan Antonio Madrid
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital, Murcia, Spain
| | - Maria Jesús Periago
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital, Murcia, Spain.,Department of Food Technology, Food Science and Nutrition, Faculty of Veterinary Sciences, Regional Campus of Internacional Excellence, University of Murcia, Murcia, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Spain
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Marta Garaulet
- Department of Physiology, Regional Campus of International Excellence, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital, Murcia, Spain.,Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Gopalakrishnan S, Kannan NN. Only time will tell: the interplay between circadian clock and metabolism. Chronobiol Int 2020; 38:149-167. [PMID: 33345624 DOI: 10.1080/07420528.2020.1842436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In most organisms ranging from cyanobacteria to humans, the endogenous timekeeping system temporally coordinates the behavioral, physiological, and metabolic processes with a periodicity close to 24 h. The timing of these daily rhythms is orchestrated by the synchronized oscillations of both the central pacemaker in the brain and the peripheral clocks located across multiple organs and tissues. A growing body of evidence suggests that the central circadian clock and peripheral clocks residing in the metabolically active tissues are incredibly well coordinated to confer coherent metabolic homeostasis. The interplay between nutrient metabolism and circadian rhythms can occur at various levels supported by the molecular clock network, multiple systemic mechanisms, and the neuroendocrine signaling pathways. While studies suggest the reciprocal regulation between circadian clock and metabolism, it is important to understand the precise mechanisms and the underlying pathways involved in the cross-talk among circadian oscillators and diverse metabolic networks. In addition to the internal synchronization of the metabolic rhythms, feeding time is considered as a potential external synchronization cue that fine tunes the timing of the circadian rhythms in metabolic peripheral clocks. A deeper understanding of how the timing of food intake and the diet composition drive the tissue-specific metabolic rhythms across the body is concomitantly important to develop novel therapeutic strategies for the metabolic disorders arising from circadian misalignment. This review summarizes the recent advancements in the circadian clock regulation of nutrient metabolism and discusses the current understanding of the metabolic feedback signals that link energy metabolism with the circadian clock.
Collapse
Affiliation(s)
- Swetha Gopalakrishnan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER) , Thiruvananthapuram, India
| | - Nisha N Kannan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER) , Thiruvananthapuram, India
| |
Collapse
|
15
|
Kato H, Ogasawara J, Takakura H, Shirato K, Sakurai T, Kizaki T, Izawa T. Exercise Training-Enhanced Lipolytic Potency to Catecholamine Depends on the Time of the Day. Int J Mol Sci 2020; 21:ijms21186920. [PMID: 32967199 PMCID: PMC7554872 DOI: 10.3390/ijms21186920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 12/22/2022] Open
Abstract
Exercise training is well known to enhance adipocyte lipolysis in response to hormone challenge. However, the existence of a relationship between the timing of exercise training and its effect on adipocyte lipolysis is unknown. To clarify this issue, Wistar rats were run on a treadmill for 9 weeks in either the early part (E-EX) or late part of the active phase (L-EX). L-EX rats exhibited greater isoproterenol-stimulated lipolysis expressed as fold induction over basal lipolysis, with greater protein expression levels of hormone-sensitive lipase (HSL) phosphorylated at Ser 660 compared to E-EX rats. Furthermore, we discovered that Brain and muscle Arnt-like (BMAL)1 protein can associate directly with several protein kinase A (PKA) regulatory units (RIα, RIβ, and RIIβ) of protein kinase, its anchoring protein (AKAP)150, and HSL, and that the association of BMAL1 with the regulatory subunits of PKA, AKAP150, and HSL was greater in L-EX than in E-EX rats. In contrast, comparison between E-EX and their counterpart sedentary control rats showed a greater co-immunoprecipitation only between BMAL1 and ATGL. Thus, both E-EX and L-EX showed an enhanced lipolytic response to isoproterenol, but the mechanisms underlying exercise training-enhanced lipolytic response to isoproterenol were different in each group.
Collapse
Affiliation(s)
- Hisashi Kato
- Organization for Research Initiatives and Development, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
| | - Junetsu Ogasawara
- Department of Health Science, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan;
| | - Hisashi Takakura
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
| | - Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University of School Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan; (K.S.); (T.S.); (T.K.)
| | - Takuya Sakurai
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University of School Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan; (K.S.); (T.S.); (T.K.)
| | - Takako Kizaki
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University of School Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan; (K.S.); (T.S.); (T.K.)
| | - Tetsuya Izawa
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan
- Correspondence: ; Tel.: +81-424-65-6721; Fax: +81-424-65-6729
| |
Collapse
|
16
|
Finger AM, Dibner C, Kramer A. Coupled network of the circadian clocks: a driving force of rhythmic physiology. FEBS Lett 2020; 594:2734-2769. [PMID: 32750151 DOI: 10.1002/1873-3468.13898] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/06/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022]
Abstract
The circadian system is composed of coupled endogenous oscillators that allow living beings, including humans, to anticipate and adapt to daily changes in their environment. In mammals, circadian clocks form a hierarchically organized network with a 'master clock' located in the suprachiasmatic nucleus of the hypothalamus, which ensures entrainment of subsidiary oscillators to environmental cycles. Robust rhythmicity of body clocks is indispensable for temporally coordinating organ functions, and the disruption or misalignment of circadian rhythms caused for instance by modern lifestyle is strongly associated with various widespread diseases. This review aims to provide a comprehensive overview of our current knowledge about the molecular architecture and system-level organization of mammalian circadian oscillators. Furthermore, we discuss the regulatory roles of peripheral clocks for cell and organ physiology and their implication in the temporal coordination of metabolism in human health and disease. Finally, we summarize methods for assessing circadian rhythmicity in humans.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Charna Dibner
- Division of Endocrinology, Diabetes, Nutrition, and Patient Education, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland.,Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Achim Kramer
- Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
17
|
Kessler K, Hornemann S, Rudovich N, Weber D, Grune T, Kramer A, Pfeiffer AFH, Pivovarova-Ramich O. Saliva Samples as A Tool to Study the Effect of Meal Timing on Metabolic And Inflammatory Biomarkers. Nutrients 2020; 12:nu12020340. [PMID: 32013045 PMCID: PMC7071228 DOI: 10.3390/nu12020340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/20/2020] [Accepted: 01/25/2020] [Indexed: 12/13/2022] Open
Abstract
Meal timing affects metabolic regulation in humans. Most studies use blood samples for their investigations. Saliva, although easily available and non-invasive, seems to be rarely used for chrononutritional studies. In this pilot study, we tested if saliva samples could be used to study the effect of timing of carbohydrate and fat intake on metabolic rhythms. In this cross-over trial, 29 nonobese men were randomized to two isocaloric 4-week diets: (1) carbohydrate-rich meals until 13:30 and high-fat meals between 16:30 and 22:00 or (2) the inverse order of meals. Stimulated saliva samples were collected every 4 h for 24 h at the end of each intervention, and levels of hormones and inflammatory biomarkers were assessed in saliva and blood. Cortisol, melatonin, resistin, adiponectin, interleukin-6 and MCP-1 demonstrated distinct diurnal variations, mirroring daytime reports in blood and showing significant correlations with blood levels. The rhythm patterns were similar for both diets, indicating that timing of carbohydrate and fat intake has a minimal effect on metabolic and inflammatory biomarkers in saliva. Our study revealed that saliva is a promising tool for the non-invasive assessment of metabolic rhythms in chrononutritional studies, but standardisation of sample collection is needed in out-of-lab studies.
Collapse
Affiliation(s)
- Katharina Kessler
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (K.K.); (S.H.); (N.R.); (A.F.H.P.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany;
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University of Medicine, 12203 Berlin, Germany
- Biomineral Research Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (K.K.); (S.H.); (N.R.); (A.F.H.P.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany;
| | - Natalia Rudovich
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (K.K.); (S.H.); (N.R.); (A.F.H.P.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany;
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University of Medicine, 12203 Berlin, Germany
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Spital Bülach, 8180 Bülach, Switzerland
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany;
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
| | - Tilman Grune
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany;
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany;
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
- German Center for Cardiovascular Research (DZHK), 10785 Berlin, Germany
- Institute of Nutrition, University of Potsdam, 14558 Nuthetal, Germany
| | - Achim Kramer
- Laboratory of Chronobiology, Institute for Medical Immunology, Charité University of Medicine, 10117 Berlin, Germany;
| | - Andreas F. H. Pfeiffer
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (K.K.); (S.H.); (N.R.); (A.F.H.P.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany;
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University of Medicine, 12203 Berlin, Germany
| | - Olga Pivovarova-Ramich
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (K.K.); (S.H.); (N.R.); (A.F.H.P.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany;
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University of Medicine, 12203 Berlin, Germany
- Reseach Group Molecular Nutritional Medicine, Dept. of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany
- Correspondence:
| |
Collapse
|
18
|
Sinturel F, Petrenko V, Dibner C. Circadian Clocks Make Metabolism Run. J Mol Biol 2020; 432:3680-3699. [PMID: 31996313 DOI: 10.1016/j.jmb.2020.01.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Most organisms adapt to the 24-h cycle of the Earth's rotation by anticipating the time of the day through light-dark cycles. The internal time-keeping system of the circadian clocks has been developed to ensure this anticipation. The circadian system governs the rhythmicity of nearly all physiological and behavioral processes in mammals. In this review, we summarize current knowledge stemming from rodent and human studies on the tight interconnection between the circadian system and metabolism in the body. In particular, we highlight recent advances emphasizing the roles of the peripheral clocks located in the metabolic organs in regulating glucose, lipid, and protein homeostasis at the organismal and cellular levels. Experimental disruption of circadian system in rodents is associated with various metabolic disturbance phenotypes. Similarly, perturbation of the clockwork in humans is linked to the development of metabolic diseases. We discuss recent studies that reveal roles of the circadian system in the temporal coordination of metabolism under physiological conditions and in the development of human pathologies.
Collapse
Affiliation(s)
- Flore Sinturel
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland.
| | - Volodymyr Petrenko
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland.
| |
Collapse
|
19
|
Dibner C. The importance of being rhythmic: Living in harmony with your body clocks. Acta Physiol (Oxf) 2020; 228:e13281. [PMID: 30980501 DOI: 10.1111/apha.13281] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 02/06/2023]
Abstract
Circadian rhythms have developed in all light-sensitive organisms, including humans, as a fundamental anticipatory mechanism that enables proactive adaptation to environmental changes. The circadian system is organized in a highly hierarchical manner, with clocks operative in most cells of the body ensuring the temporal coordination of physiological processes. Circadian misalignment, stemming from modern life style, draws increasing attention due to its tight association with the development of metabolic, cardiovascular, inflammatory and mental diseases as well as cancer. This review highlights recent findings emphasizing the role of the circadian system in the temporal orchestration of physiology, with a particular focus on implications of circadian misalignment in human pathologies.
Collapse
Affiliation(s)
- Charna Dibner
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Medicine University Hospital of Geneva Geneva Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine University of Geneva Geneva Switzerland
- Diabetes Center, Faculty of Medicine University of Geneva Geneva Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3) Geneva Switzerland
| |
Collapse
|
20
|
Bellos I, Fitrou G, Pergialiotis V, Perrea DN, Daskalakis G. Serum levels of adipokines in gestational diabetes: a systematic review. J Endocrinol Invest 2019; 42:621-631. [PMID: 30392100 DOI: 10.1007/s40618-018-0973-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To determine the difference of serum levels of 10 adipokines (apelin, chemerin, fatty acid-binding protein-4, fibroblast growth factor-21, monocyte chemoattractant protein-1, nesfatin-1, omentin-1, resistin, vaspin, and visfatin) among women with gestational diabetes and healthy pregnant controls. MATERIALS AND METHODS Literature search was conducted using the Medline (1966-2018), Scopus (2004-2018), Cochrane Central Register of Controlled Trials (CENTRAL) (1999-2018), Clinicaltrials.gov (2008-2018) and Google Scholar (2004-2018) databases, along with the reference list of the included studies. RESULTS Ninety-one studies were included in the present review, with a total number of 11,074 pregnant women. A meta-analysis was not conducted due to the high inter-study heterogeneity. Current evidence suggests that fatty acid-binding protein-4 levels are significantly increased in pregnancies complicated with gestational diabetes, while no association of serum apelin and monocyte chemoattractant protein-1 with the disease can be supported. Data regarding the rest adipokines are conflicting, since the available studies did not unanimously indicate a significant change of their levels in gestational diabetes. CONCLUSIONS The findings of the present systematic review suggest the promising role of fatty acid-binding protein-4 in the prediction of gestational diabetes, while inconsistent evidence exists regarding the rest novel adipokines. Future cohorts are needed to assess their predictive efficacy and fully elucidate their contribution in the disease.
Collapse
Affiliation(s)
- I Bellos
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, Athens University Medical School, National and Kapodistrian University of Athens, 15Β, Ag. Thoma str., 115 27, Athens, Greece.
| | - G Fitrou
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, Athens University Medical School, National and Kapodistrian University of Athens, 15Β, Ag. Thoma str., 115 27, Athens, Greece
| | - V Pergialiotis
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, Athens University Medical School, National and Kapodistrian University of Athens, 15Β, Ag. Thoma str., 115 27, Athens, Greece
| | - D N Perrea
- Laboratory of Experimental Surgery and Surgical Research N.S. Christeas, Athens University Medical School, National and Kapodistrian University of Athens, 15Β, Ag. Thoma str., 115 27, Athens, Greece
| | - G Daskalakis
- First Department of Obstetrics and Gynecology, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
21
|
Dollet L, Zierath JR. Interplay between diet, exercise and the molecular circadian clock in orchestrating metabolic adaptations of adipose tissue. J Physiol 2019; 597:1439-1450. [PMID: 30615204 DOI: 10.1113/jp276488] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
Disruption of circadian rhythmicity induced by prolonged light exposure, altered sleep patterns and shift work is associated with the development of obesity and related metabolic disorders, including type 2 diabetes and cardiovascular diseases. White and brown adipose tissue activity shows circadian rhythmicity, with daily variations in the regulation of metabolic processes such as lipolysis, glucose and lipid uptake, and adipokine secretion. The role of the circadian clock in the regulation of energy homeostasis has raised interest in clock-related strategies to mitigate metabolic disturbances associated with type 2 diabetes, including 'resynchronizing' metabolism through diet or targeting a particular time of a day to potentiate the effect of a pharmacological or physiological treatment. Exercise is an effective intervention to prevent insulin resistance and type 2 diabetes. Beyond its effect on skeletal muscle, exercise training also has a profound effect on adipose tissue. Adipose tissue partly mediates the beneficial effect of exercise on glucose and energy homeostasis, via its metabolic and endocrine function. The interaction between zeitgeber time and diet or exercise is likely to influence the metabolic response of adipose tissue and therefore impact the whole-body phenotype. Understanding the impact of circadian clock systems on human physiology and how this is regulated by exercise in a tissue-specific manner will yield new insights for the management of metabolic disorders.
Collapse
Affiliation(s)
- Lucile Dollet
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Abstract
The epidemic of Type 2 diabetes mellitus necessitates development of novel therapeutic and preventative strategies to attenuate expansion of this debilitating disease. Evidence links the circadian system to various aspects of diabetes pathophysiology and treatment. The aim of this review will be to outline the rationale for therapeutic targeting of the circadian system in the treatment and prevention of Type 2 diabetes mellitus and consequent metabolic comorbidities.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic School of Medicine, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
23
|
Lambert JM, Anderson AK, Cowart LA. Sphingolipids in adipose tissue: What's tipping the scale? Adv Biol Regul 2018; 70:19-30. [PMID: 30473005 PMCID: PMC11129658 DOI: 10.1016/j.jbior.2018.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/13/2018] [Accepted: 10/14/2018] [Indexed: 01/06/2023]
Abstract
Adipose tissue lies at the heart of obesity, mediating its many effects upon the rest of the body, with its unique capacity to expand and regenerate, throughout the lifespan of the organism. Adipose is appreciated as an endocrine organ, with its myriad adipokines that elicit both physiological and pathological outcomes. Sphingolipids, bioactive signaling molecules, affect many aspects of obesity and the metabolic syndrome. While sphingolipids are appreciated in the context of these diseases in other tissues, there are many discoveries yet to be uncovered in the adipose tissue. This review focuses on the effects of sphingolipids on various aspects of adipose function and dysfunction. The processes of adipogenesis, metabolism and thermogenesis, in addition to inflammation and insulin resistance are intimately linked to sphingolipids as discussed below.
Collapse
Affiliation(s)
- Johana M Lambert
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Andrea K Anderson
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, VA, USA.
| |
Collapse
|
24
|
Petrenko V, Philippe J, Dibner C. Time zones of pancreatic islet metabolism. Diabetes Obes Metab 2018; 20 Suppl 2:116-126. [PMID: 30230177 DOI: 10.1111/dom.13383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/04/2018] [Accepted: 05/23/2018] [Indexed: 12/28/2022]
Abstract
Most living beings possess an intrinsic system of circadian oscillators, allowing anticipation of the Earth's rotation around its own axis. The mammalian circadian timing system orchestrates nearly all aspects of physiology and behaviour. Together with systemic signals originating from the central clock that resides in the hypothalamic suprachiasmatic nucleus, peripheral oscillators orchestrate tissue-specific fluctuations in gene transcription and translation, and posttranslational modifications, driving overt rhythms in physiology and behaviour. There is accumulating evidence of a reciprocal connection between the circadian oscillator and most aspects of physiology and metabolism, in particular as the circadian system plays a critical role in orchestrating body glucose homeostasis. Recent reports imply that circadian clocks operative in the endocrine pancreas regulate insulin secretion, and that islet clock perturbation in rodents leads to the development of overt type 2 diabetes. While whole islet clocks have been extensively studied during the last years, the heterogeneity of islet cell oscillators and the interplay between α- and β-cellular clocks for orchestrating glucagon and insulin secretion have only recently gained attention. Here, we review recent findings on the molecular makeup of the circadian clocks operative in pancreatic islet cells in rodents and in humans, and focus on the physiologically relevant synchronizers that are resetting these time-keepers. Moreover, the implication of islet clock functional outputs in the temporal coordination of metabolism in health and disease will be highlighted.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Charna Dibner
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| |
Collapse
|
25
|
Noshiro M, Kawamoto T, Nakashima A, Ozaki N, Ueno T, Saeki M, Honda K, Fujimoto K, Kato Y. Deficiency of the basic helix-loop-helix transcription factor DEC1 prevents obesity induced by a high-fat diet in mice. Genes Cells 2018; 23:658-669. [PMID: 29968353 DOI: 10.1111/gtc.12607] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/14/2018] [Accepted: 05/29/2018] [Indexed: 01/04/2023]
Abstract
Obesity is a major public health problem in developed countries resulting from increased food intake and decreased energy consumption and usually associated with abnormal lipid metabolism. Here, we show that DEC1, a basic helix-loop-helix transcription factor, plays an important role in the regulation of lipid consumption in mouse brown adipose tissue (BAT), which is the major site of thermogenesis. Homozygous Dec1 deletion attenuated high-fat-diet-induced obesity, adipocyte hypertrophy, fat volume and hepatic steatosis. Furthermore, DEC1 deficiency increased body temperature during daytime and enhanced the expression of uncoupler protein 1, a key factor of thermogenesis, and various lipolysis-related genes in interscapular BAT. In vitro experiments suggested that DEC1 suppresses the expression of various lipolysis-related genes induced by the heterodimer of peroxisome proliferator-activated receptor γ and retinoid X receptor α (RXRα) through direct binding to RXRα. These observations suggest that enhanced lipolysis in BAT caused by DEC1 deficiency leads to an increase in lipid consumption, thereby decreasing lipid accumulation in adipose tissues and the liver. Thus, DEC1 may serve as an energy-saving factor that suppresses lipid consumption, which may be relevant to managing obesity.
Collapse
Affiliation(s)
- Mitsuhide Noshiro
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takeshi Kawamoto
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Writing Center, Hiroshima University, Higashi-Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Noritsugu Ozaki
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Toshinori Ueno
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Masayumi Saeki
- Health Examination Center, Chugoku Rousai Hospital, Kure, Japan
| | - Kiyomasa Honda
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Katsumi Fujimoto
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Kato
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
26
|
Froy O, Garaulet M. The Circadian Clock in White and Brown Adipose Tissue: Mechanistic, Endocrine, and Clinical Aspects. Endocr Rev 2018; 39:261-273. [PMID: 29490014 PMCID: PMC6456924 DOI: 10.1210/er.2017-00193] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 02/22/2018] [Indexed: 12/19/2022]
Abstract
Obesity is a major risk factor for the development of illnesses, such as insulin resistance and hypertension, and has become a serious public health problem. Mammals have developed a circadian clock located in the hypothalamic suprachiasmatic nuclei (SCN) that responds to the environmental light-dark cycle. Clocks similar to the one located in the SCN are found in peripheral tissues, such as the kidney, liver, and adipose tissue. The circadian clock regulates metabolism and energy homeostasis in peripheral tissues by mediating activity and/or expression of key metabolic enzymes and transport systems. Knockouts or mutations in clock genes that lead to disruption of cellular rhythmicity have provided evidence to the tight link between the circadian clock and metabolism. In addition, key proteins play a dual role in regulating the core clock mechanism, as well as adipose tissue metabolism, and link circadian rhythms with lipogenesis and lipolysis. Adipose tissues are distinguished as white, brown, and beige (or brite), each with unique metabolic characteristics. Recently, the role of the circadian clock in regulating the differentiation into the different adipose tissues has been investigated. In this review, the role of clock proteins and the downstream signaling pathways in white, brown, and brite adipose tissue function and differentiation will be reviewed. In addition, chronodisruption and metabolic disorders and clinical aspects of circadian adiposity will be addressed.
Collapse
Affiliation(s)
- Oren Froy
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Marta Garaulet
- Department of Physiology, University of Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| |
Collapse
|
27
|
Dierickx P, Van Laake LW, Geijsen N. Circadian clocks: from stem cells to tissue homeostasis and regeneration. EMBO Rep 2018; 19:18-28. [PMID: 29258993 PMCID: PMC5757216 DOI: 10.15252/embr.201745130] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/30/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023] Open
Abstract
The circadian clock is an evolutionarily conserved timekeeper that adapts body physiology to diurnal cycles of around 24 h by influencing a wide variety of processes such as sleep-to-wake transitions, feeding and fasting patterns, body temperature, and hormone regulation. The molecular clock machinery comprises a pathway that is driven by rhythmic docking of the transcription factors BMAL1 and CLOCK on clock-controlled output genes, which results in tissue-specific oscillatory gene expression programs. Genetic as well as environmental perturbation of the circadian clock has been implicated in various diseases ranging from sleep to metabolic disorders and cancer development. Here, we review the origination of circadian rhythms in stem cells and their function in differentiated cells and organs. We describe how clocks influence stem cell maintenance and organ physiology, as well as how rhythmicity affects lineage commitment, tissue regeneration, and aging.
Collapse
Affiliation(s)
- Pieterjan Dierickx
- Hubrecht Institute-KNAW and University Medical Center, Utrecht, The Netherlands
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Linda W Van Laake
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Niels Geijsen
- Hubrecht Institute-KNAW and University Medical Center, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
28
|
Plano SA, Casiraghi LP, García Moro P, Paladino N, Golombek DA, Chiesa JJ. Circadian and Metabolic Effects of Light: Implications in Weight Homeostasis and Health. Front Neurol 2017; 8:558. [PMID: 29097992 PMCID: PMC5653694 DOI: 10.3389/fneur.2017.00558] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022] Open
Abstract
Daily interactions between the hypothalamic circadian clock at the suprachiasmatic nucleus (SCN) and peripheral circadian oscillators regulate physiology and metabolism to set temporal variations in homeostatic regulation. Phase coherence of these circadian oscillators is achieved by the entrainment of the SCN to the environmental 24-h light:dark (LD) cycle, coupled through downstream neural, neuroendocrine, and autonomic outputs. The SCN coordinate activity and feeding rhythms, thus setting the timing of food intake, energy expenditure, thermogenesis, and active and basal metabolism. In this work, we will discuss evidences exploring the impact of different photic entrainment conditions on energy metabolism. The steady-state interaction between the LD cycle and the SCN is essential for health and wellbeing, as its chronic misalignment disrupts the circadian organization at different levels. For instance, in nocturnal rodents, non-24 h protocols (i.e., LD cycles of different durations, or chronic jet-lag simulations) might generate forced desynchronization of oscillators from the behavioral to the metabolic level. Even seemingly subtle photic manipulations, as the exposure to a “dim light” scotophase, might lead to similar alterations. The daily amount of light integrated by the clock (i.e., the photophase duration) strongly regulates energy metabolism in photoperiodic species. Removing LD cycles under either constant light or darkness, which are routine protocols in chronobiology, can also affect metabolism, and the same happens with disrupted LD cycles (like shiftwork of jetlag) and artificial light at night in humans. A profound knowledge of the photic and metabolic inputs to the clock, as well as its endocrine and autonomic outputs to peripheral oscillators driving energy metabolism, will help us to understand and alleviate circadian health alterations including cardiometabolic diseases, diabetes, and obesity.
Collapse
Affiliation(s)
- Santiago A Plano
- Chronophysiology Laboratory, Institute for Biomedical Research (BIOMED - CONICET), School of Medical Sciences, Universidad Católica Argentina (UCA), Buenos Aires, Argentina.,Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Leandro P Casiraghi
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Paula García Moro
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Natalia Paladino
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Diego A Golombek
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Juan J Chiesa
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| |
Collapse
|
29
|
Freese J, Klement RJ, Ruiz-Núñez B, Schwarz S, Lötzerich H. The sedentary (r)evolution: Have we lost our metabolic flexibility? F1000Res 2017; 6:1787. [PMID: 29225776 DOI: 10.12688/f1000research.12724.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
During the course of evolution, up until the agricultural revolution, environmental fluctuations forced the human species to develop a flexible metabolism in order to adapt its energy needs to various climate, seasonal and vegetation conditions. Metabolic flexibility safeguarded human survival independent of food availability. In modern times, humans switched their primal lifestyle towards a constant availability of energy-dense, yet often nutrient-deficient, foods, persistent psycho-emotional stressors and a lack of exercise. As a result, humans progressively gain metabolic disorders, such as the metabolic syndrome, type 2 diabetes, non-alcoholic fatty liver disease, certain types of cancer, cardiovascular disease and Alzheimer´s disease, wherever the sedentary lifestyle spreads in the world. For more than 2.5 million years, our capability to store fat for times of food shortage was an outstanding survival advantage. Nowadays, the same survival strategy in a completely altered surrounding is responsible for a constant accumulation of body fat. In this article, we argue that the metabolic disease epidemic is largely based on a deficit in metabolic flexibility. We hypothesize that the modern energetic inflexibility, typically displayed by symptoms of neuroglycopenia, can be reversed by re-cultivating suppressed metabolic programs, which became obsolete in an affluent environment, particularly the ability to easily switch to ketone body and fat oxidation. In a simplified model, the basic metabolic programs of humans' primal hunter-gatherer lifestyle are opposed to the current sedentary lifestyle. Those metabolic programs, which are chronically neglected in modern surroundings, are identified and conclusions for the prevention of chronic metabolic diseases are drawn.
Collapse
Affiliation(s)
- Jens Freese
- Institute of Outdoor Sports and Environmental Science, German Sports University Cologne, Cologne, 50933, Germany
| | - Rainer Johannes Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Schweinfurt, 97422, Germany
| | - Begoña Ruiz-Núñez
- Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713, Netherlands
| | - Sebastian Schwarz
- University College Physiotherapy Thim van der Laan,, Landquart, 7302, Switzerland
| | - Helmut Lötzerich
- Institute of Outdoor Sports and Environmental Science, German Sports University Cologne, Cologne, 50933, Germany
| |
Collapse
|
30
|
Freese J, Klement RJ, Ruiz-Núñez B, Schwarz S, Lötzerich H. The sedentary (r)evolution: Have we lost our metabolic flexibility? F1000Res 2017; 6:1787. [PMID: 29225776 PMCID: PMC5710317 DOI: 10.12688/f1000research.12724.2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2018] [Indexed: 12/18/2022] Open
Abstract
During the course of evolution, up until the agricultural revolution, environmental fluctuations forced the human species to develop a flexible metabolism in order to adapt its energy needs to various climate, seasonal and vegetation conditions. Metabolic flexibility safeguarded human survival independent of food availability. In modern times, humans switched their primal lifestyle towards a constant availability of energy-dense, yet often nutrient-deficient, foods, persistent psycho-emotional stressors and a lack of exercise. As a result, humans progressively gain metabolic disorders, such as the metabolic syndrome, type 2 diabetes, non-alcoholic fatty liver disease, certain types of cancer, cardiovascular disease and Alzheimer´s disease, wherever the sedentary lifestyle spreads in the world. For more than 2.5 million years, our capability to store fat for times of food shortage was an outstanding survival advantage. Nowadays, the same survival strategy in a completely altered surrounding is responsible for a constant accumulation of body fat. In this article, we argue that the metabolic disease epidemic is largely based on a deficit in metabolic flexibility. We hypothesize that the modern energetic inflexibility, typically displayed by symptoms of neuroglycopenia, can be reversed by re-cultivating suppressed metabolic programs, which became obsolete in an affluent environment, particularly the ability to easily switch to ketone body and fat oxidation. In a simplified model, the basic metabolic programs of humans’ primal hunter-gatherer lifestyle are opposed to the current sedentary lifestyle. Those metabolic programs, which are chronically neglected in modern surroundings, are identified and conclusions for the prevention of chronic metabolic diseases are drawn.
Collapse
Affiliation(s)
- Jens Freese
- Institute of Outdoor Sports and Environmental Science, German Sports University Cologne, Cologne, 50933, Germany
| | - Rainer Johannes Klement
- Department of Radiotherapy and Radiation Oncology, Leopoldina Hospital Schweinfurt, Schweinfurt, 97422, Germany
| | - Begoña Ruiz-Núñez
- Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713, Netherlands
| | - Sebastian Schwarz
- University College Physiotherapy Thim van der Laan,, Landquart, 7302, Switzerland
| | - Helmut Lötzerich
- Institute of Outdoor Sports and Environmental Science, German Sports University Cologne, Cologne, 50933, Germany
| |
Collapse
|
31
|
Zubidat AE, Haim A. Artificial light-at-night - a novel lifestyle risk factor for metabolic disorder and cancer morbidity. J Basic Clin Physiol Pharmacol 2017; 28:295-313. [PMID: 28682785 DOI: 10.1515/jbcpp-2016-0116] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 03/02/2017] [Indexed: 06/07/2023]
Abstract
Both obesity and breast cancer are already recognized worldwide as the most common syndromes in our modern society. Currently, there is accumulating evidence from epidemiological and experimental studies suggesting that these syndromes are closely associated with circadian disruption. It has been suggested that melatonin (MLT) and the circadian clock genes both play an important role in the development of these syndromes. However, we still poorly understand the molecular mechanism underlying the association between circadian disruption and the modern health syndromes. One promising candidate is epigenetic modifications of various genes, including clock genes, circadian-related genes, oncogenes, and metabolic genes. DNA methylation is the most prominent epigenetic signaling tool for gene expression regulation induced by environmental exposures, such as artificial light-at-night (ALAN). In this review, we first provide an overview on the molecular feedback loops that generate the circadian regulation and how circadian disruption by ALAN can impose adverse impacts on public health, particularly metabolic disorders and breast cancer development. We then focus on the relation between ALAN-induced circadian disruption and both global DNA methylation and specific loci methylation in relation to obesity and breast cancer morbidities. DNA hypo-methylation and DNA hyper-methylation, are suggested as the most studied epigenetic tools for the activation and silencing of genes that regulate metabolic and monostatic responses. Finally, we discuss the potential clinical and therapeutic roles of MLT suppression and DNA methylation patterns as novel biomarkers for the early detection of metabolic disorders and breast cancer development.
Collapse
|
32
|
Catabolic and anabolic faces of insulin resistance and their disorders: a new insight into circadian control of metabolic disorders leading to diabetes. Future Sci OA 2017; 3:FSO201. [PMID: 28884000 PMCID: PMC5583684 DOI: 10.4155/fsoa-2017-0015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/23/2017] [Indexed: 12/18/2022] Open
Abstract
Maintenance of glucose homeostasis during circadian behavioral cycles is critical. The processes controlling the switch between predominant lipolysis/fatty oxidation during fasting and predominant lipid storage/glucose oxidation following feeding are determined principally by insulin. Chronic elevated threshold of insulin resistance (IR) is a key pathological feature of obesity, Type 2 diabetes, sepsis and cancer cachexia; however, a temporal reduced threshold of IR is widely met in fasting/hibernation, pregnancy, antibacterial immunity, exercise and stress. Paradoxically, some of these cases are associated with catabolic metabolism, whereas others are related to anabolic pathways. This article considers the possible causes of circadian disorders in glucose and lipid metabolism that act as a driving force for obesity-promoted development of Type 2 diabetes. This is intended to provide improved insight into the pathogenesis of chronic circadian disorders that increase the risk of diabetes, and consider new targets for its metabolic and drug correction. Insulin resistance (IR) is a common adaptive mechanism, acting under opposite anabolic and catabolic conditions. However, chronic IR is a key pathological feature of obesity, Type 2 diabetes and cancer cachexia, whereas a temporal IR is widely seen in fasting, pregnancy, exercise and stress. Therefore, it is important to understand when this transient IR-mechanism shifts to chronic IR-associated diseases. What factors result in the switch between the anabolic and catabolic conditions and what defect(s) in this switch is associated with chronic IR induction? The present opinion article aimed to address these questions to the metabolic changes typical for circadian regulation in lean, obese and diabetic patients. Graphical abstract: Early circadian IR disorders caused by overweight and obesity are associated with increased risk for diabetes via formation of a vicious cycle between lipid anabolic and catabolic programs thus distorting insulin and lipid levels in day/night period.
Collapse
|
33
|
Gachon F, Loizides-Mangold U, Petrenko V, Dibner C. Glucose Homeostasis: Regulation by Peripheral Circadian Clocks in Rodents and Humans. Endocrinology 2017; 158:1074-1084. [PMID: 28324069 DOI: 10.1210/en.2017-00218] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/10/2017] [Indexed: 12/15/2022]
Abstract
Most organisms, including humans, have developed an intrinsic system of circadian oscillators, allowing the anticipation of events related to the rotation of Earth around its own axis. The mammalian circadian timing system orchestrates nearly all aspects of physiology and behavior. Together with systemic signals, emanating from the central clock that resides in the hypothalamus, peripheral oscillators orchestrate tissue-specific fluctuations in gene expression, protein synthesis, and posttranslational modifications, driving overt rhythms in physiology and behavior. There is increasing evidence on the essential roles of the peripheral oscillators, operative in metabolically active organs in the regulation of body glucose homeostasis. Here, we review some recent findings on the molecular and cellular makeup of the circadian timing system and its implications in the temporal coordination of metabolism in health and disease.
Collapse
Affiliation(s)
- Frédéric Gachon
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Ursula Loizides-Mangold
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Diabetes Center, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Volodymyr Petrenko
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Diabetes Center, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Charna Dibner
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Diabetes Center, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
34
|
Rynders CA, Bergouignan A, Kealey E, Bessesen DH. Ability to adjust nocturnal fat oxidation in response to overfeeding predicts 5-year weight gain in adults. Obesity (Silver Spring) 2017; 25:873-880. [PMID: 28440048 PMCID: PMC5407418 DOI: 10.1002/oby.21807] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/29/2017] [Accepted: 01/30/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To determine whether metabolic responses to short-term overfeeding predict longitudinal changes in body weight. METHODS Twenty-four-hour energy expenditure (EE) and substrate utilization were measured at baseline in a room calorimeter following 3 days of eucaloric and hypercaloric feeding (40% excess) in a sample of lean adults (n: 34; age: 28 ± 2 y; BMI: 22 ± 3 kg/m2 ). Body mass and fat mass (dual-energy x-ray absorptiometry) were measured annually for 5 years. Regression analyses examined whether changes in EE and fuel use with overfeeding predicted body weight and composition changes over 5 years. RESULTS Overfeeding increased EE and reduced fat oxidation when examined over the 24-hour, waking, and nocturnal periods. Absolute change in body mass over 5 years was 3.0 ± 0.6 kg (average rate of change = 0.7 ± 0.1 kg/y, P < 0.001). Lower nocturnal (but not 24-hour or waking) fat oxidation (r = -0.42, P = 0.01) and EE (r = -0.33, P = 0.05) with overfeeding were the strongest predictors of 5-year weight gain. When adjusted for covariates, changes in nocturnal fat oxidation and EE with overfeeding predicted 41% of the variance in weight change (P = 0.02). CONCLUSIONS Failure to maintain fat oxidation at night following a period of overfeeding appears to be associated with a metabolic phenotype favoring weight gain.
Collapse
Affiliation(s)
- Corey A Rynders
- Division of Geriatric Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Audrey Bergouignan
- Anschutz Health & Wellness Center at the University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elizabeth Kealey
- Anschutz Health & Wellness Center at the University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel H Bessesen
- Anschutz Health & Wellness Center at the University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Endocrinology, Denver Health Medical Center, Denver, Colorado, USA
| |
Collapse
|
35
|
Boltze J, Nitzsche F, Jolkkonen J, Weise G, Pösel C, Nitzsche B, Wagner DC. Concise Review: Increasing the Validity of Cerebrovascular Disease Models and Experimental Methods for Translational Stem Cell Research. Stem Cells 2017; 35:1141-1153. [DOI: 10.1002/stem.2595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Boltze
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Cell Technology; Lübeck Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| | - Franziska Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pennsylvania USA
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern Finland; Kuopio Finland
| | - Gesa Weise
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Neurology; University of Leipzig; Germany
| | - Claudia Pösel
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Björn Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Nuclear Medicine; University Hospital Leipzig; Germany
| | - Daniel-Christoph Wagner
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Institute of Pathology, University Medical Center Mainz; Germany
| |
Collapse
|
36
|
Petrenko V, Gosmain Y, Dibner C. High-Resolution Recording of the Circadian Oscillator in Primary Mouse α- and β-Cell Culture. Front Endocrinol (Lausanne) 2017; 8:68. [PMID: 28439257 PMCID: PMC5383706 DOI: 10.3389/fendo.2017.00068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/24/2017] [Indexed: 01/14/2023] Open
Abstract
Circadian clocks have been developed in evolution as an anticipatory mechanism allowing for adaptation to the constantly changing light environment due to rotation of the Earth. This mechanism is functional in all light-sensitive organisms. There is a considerable body of evidence on the tight connection between the circadian clock and most aspects of physiology and metabolism. Clocks, operative in the pancreatic islets, have caught particular attention in the last years due to recent reports on their critical roles in regulation of insulin secretion and etiology of type 2 diabetes. While β-cell clocks have been extensively studied during the last years, α-cell clocks and their role in islet function and orchestration of glucose metabolism stayed unexplored, largely due to the difficulty to isolate α-cells, which represents a considerable technical challenge. Here, we provide a detailed description of an experimental approach for the isolation of separate mouse α- and β-cell population, culture of isolated primary α- and β-cells, and their subsequent long-term high-resolution circadian bioluminescence recording. For this purpose, a triple reporter ProGlucagon-Venus/RIP-Cherry/Per2:Luciferase mouse line was established, carrying specific fluorescent reporters for α- and β-cells, and luciferase reporter for monitoring the molecular clockwork. Flow cytometry fluorescence-activated cell sorting allowed separating pure α- and β-cell populations from isolated islets. Experimental conditions, developed by us for the culture of functional primary mouse α- and β-cells for at least 10 days, will be highlighted. Importantly, temporal analysis of freshly isolated α- and β-cells around-the-clock revealed preserved rhythmicity of core clock genes expression. Finally, we describe the setting to assess circadian rhythm in cultured α- and β-cells synchronized in vitro. The here-described methodology allows to analyze the functional properties of primary α- and β-cells under physiological or pathophysiological conditions and to assess the islet cellular clock properties.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Endocrinology, Diabetes, Hypertension and Nutrition Division, Department of Specialties of Medicine, University Hospital of Geneva, Geneva, Switzerland
- Faculty of Medicine, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Yvan Gosmain
- Diabetes Center of the Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Molecular Diabetes Laboratory, Endocrinology, Diabetes, Hypertension and Nutrition Division, Faculty of Medicine, Department of Specialties of Medicine, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Endocrinology, Diabetes, Hypertension and Nutrition Division, Department of Specialties of Medicine, University Hospital of Geneva, Geneva, Switzerland
- Faculty of Medicine, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva, Geneva, Switzerland
- *Correspondence: Charna Dibner,
| |
Collapse
|
37
|
Koch CE, Bartlang MS, Kiehn JT, Lucke L, Naujokat N, Helfrich-Förster C, Reber SO, Oster H. Time-of-day-dependent adaptation of the HPA axis to predictable social defeat stress. J Endocrinol 2016; 231:209-221. [PMID: 27660201 DOI: 10.1530/joe-16-0163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/22/2016] [Indexed: 11/08/2022]
Abstract
In modern societies, the risk of developing a whole array of affective and somatic disorders is associated with the prevalence of frequent psychosocial stress. Therefore, a better understanding of adaptive stress responses and their underlying molecular mechanisms is of high clinical interest. In response to an acute stressor, each organism can either show passive freezing or active fight-or-flight behaviour, with activation of sympathetic nervous system and the hypothalamus-pituitary-adrenal (HPA) axis providing the necessary energy for the latter by releasing catecholamines and glucocorticoids (GC). Recent data suggest that stress responses are also regulated by the endogenous circadian clock. In consequence, the timing of stress may critically affect adaptive responses to and/or pathological effects of repetitive stressor exposure. In this article, we characterize the impact of predictable social defeat stress during daytime versus nighttime on bodyweight development and HPA axis activity in mice. While 19 days of social daytime stress led to a transient reduction in bodyweight without altering HPA axis activity at the predicted time of stressor exposure, more detrimental effects were seen in anticipation of nighttime stress. Repeated nighttime stressor exposure led to alterations in food metabolization and reduced HPA axis activity with lower circulating adrenocorticotropic hormone (ACTH) and GC concentrations at the time of predicted stressor exposure. Our data reveal a circadian gating of stress adaptation to predictable social defeat stress at the level of the HPA axis with impact on metabolic homeostasis underpinning the importance of timing for the body's adaptability to repetitive stress.
Collapse
Affiliation(s)
- C E Koch
- University of LübeckChronophysiology Group, Medical Department 1, Lübeck, Germany
| | - M S Bartlang
- University of WürzburgBiocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Würzburg, Germany
| | - J T Kiehn
- University of LübeckChronophysiology Group, Medical Department 1, Lübeck, Germany
| | - L Lucke
- Department of Behavioral and Molecular NeurobiologyUniversity of Regensburg, Regensburg, Germany
| | - N Naujokat
- University of LübeckChronophysiology Group, Medical Department 1, Lübeck, Germany
| | - C Helfrich-Förster
- University of WürzburgBiocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Würzburg, Germany
| | - S O Reber
- Department of Behavioral and Molecular NeurobiologyUniversity of Regensburg, Regensburg, Germany
| | - H Oster
- University of LübeckChronophysiology Group, Medical Department 1, Lübeck, Germany
| |
Collapse
|
38
|
Petrenko V, Saini C, Perrin L, Dibner C. Parallel Measurement of Circadian Clock Gene Expression and Hormone Secretion in Human Primary Cell Cultures. J Vis Exp 2016. [PMID: 27911383 DOI: 10.3791/54673] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Circadian clocks are functional in all light-sensitive organisms, allowing for an adaptation to the external world by anticipating daily environmental changes. Considerable progress in our understanding of the tight connection between the circadian clock and most aspects of physiology has been made in the field over the last decade. However, unraveling the molecular basis that underlies the function of the circadian oscillator in humans stays of highest technical challenge. Here, we provide a detailed description of an experimental approach for long-term (2-5 days) bioluminescence recording and outflow medium collection in cultured human primary cells. For this purpose, we have transduced primary cells with a lentiviral luciferase reporter that is under control of a core clock gene promoter, which allows for the parallel assessment of hormone secretion and circadian bioluminescence. Furthermore, we describe the conditions for disrupting the circadian clock in primary human cells by transfecting siRNA targeting CLOCK. Our results on the circadian regulation of insulin secretion by human pancreatic islets, and myokine secretion by human skeletal muscle cells, are presented here to illustrate the application of this methodology. These settings can be used to study the molecular makeup of human peripheral clocks and to analyze their functional impact on primary cells under physiological or pathophysiological conditions.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Department of Medical Specialties, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Diabetes Center, University of Geneva Medical School, Institute of Genetics and Genomics in Geneva (iGE3)
| | - Camille Saini
- Population Epidemiology Unit (UEP), Community Medicine, Geneva University Hospital
| | - Laurent Perrin
- Department of Medical Specialties, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Diabetes Center, University of Geneva Medical School, Institute of Genetics and Genomics in Geneva (iGE3)
| | - Charna Dibner
- Department of Medical Specialties, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Diabetes Center, University of Geneva Medical School, Institute of Genetics and Genomics in Geneva (iGE3);
| |
Collapse
|
39
|
Ren R, Chen Z, Zhao X, Sun T, Zhang Y, Chen J, Lu S, Ma W. A possible regulatory link between Twist 1 and PPARγ gene regulation in 3T3-L1 adipocytes. Lipids Health Dis 2016; 15:189. [PMID: 27825360 PMCID: PMC5101646 DOI: 10.1186/s12944-016-0361-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 11/02/2016] [Indexed: 12/13/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor γ (PPARγ) is a critical gene that regulates the function of adipocytes. Therefore, studies on the molecular regulation mechanism of PPARγ are important to understand the function of adipose tissue. Twist 1 is another important functional gene in adipose tissue, and hundreds of genes are regulated by Twist 1. The aim of this study was to investigate the regulation of Twist 1 and PPARγ expression in 3T3-L1 mature adipocytes. Methods We induced differentiation in 3T3-L1 preadipocytes and examined alterations in Twist 1 and PPARγ expression. We used the PPARγ agonist pioglitazone and the PPARγ antagonist T0070907 to investigate the effect of PPARγ on Twist 1 expression. In addition, we utilized retroviral interference and overexpression of Twist 1 to determine the effects of Twist 1 on PPARγ expression. Results The expression levels of Twist 1 and PPARγ were induced during differentiation in 3T3-L1 adipocytes. Application of either a PPARγ agonist (pioglitazone) or antagonist (T0070907) influenced Twist 1 expression, with up-regulation of Twist 1 under pioglitazone (1 μM, 24 h) and down-regulation of Twist 1 under T0070907 (100 μM, 24 h) exposure. Furthermore, the retroviral interference of Twist 1 decreased the protein and mRNA expression of PPARγ, while Twist 1 overexpression had the opposite effect. Conclusions There was a possible regulatory link between Twist 1 and PPARγ in 3T3-L1 mature adipocytes. This regulatory link enhanced the regulation of PPARγ and may be a functional mechanism of Twist 1 regulation of adipocyte physiology and pathology.
Collapse
Affiliation(s)
- Rui Ren
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China
| | - Zhufeng Chen
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China
| | - Xia Zhao
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China
| | - Tao Sun
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China
| | - Yuchao Zhang
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, 266071, People's Republic of China
| | - Jie Chen
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China
| | - Sumei Lu
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China.
| | - Wanshan Ma
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, People's Republic of China
| |
Collapse
|
40
|
Lv Y, Zhang SY, Liang X, Zhang H, Xu Z, Liu B, Xu MJ, Jiang C, Shang J, Wang X. Adrenomedullin 2 Enhances Beiging in White Adipose Tissue Directly in an Adipocyte-autonomous Manner and Indirectly through Activation of M2 Macrophages. J Biol Chem 2016; 291:23390-23402. [PMID: 27621315 DOI: 10.1074/jbc.m116.735563] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Indexed: 12/15/2022] Open
Abstract
Adrenomedullin 2 (ADM2) is an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family. Our previous studies showed that overexpression of ADM2 in mice reduced obesity and insulin resistance by increasing thermogenesis in brown adipose tissue. However, the effects of ADM2 in another type of thermogenic adipocyte, beige adipocytes, remain to be understood. The plasma ADM2 levels were inversely correlated with obesity in humans, and adipo-ADM2-transgenic (tg) mice displayed resistance to high-fat diet-induced obesity with increased energy expenditure. Beiging of subcutaneous white adipose tissues (WAT) was more noticeably induced in high-fat diet-fed transgenic mice with adipocyte-ADM2 overexpression (adipo-ADM2-tg mice) than in WT animals. ADM2 treatment in primary rat subcutaneous adipocytes induced beiging with up-regulation of UCP1 and beiging-related marker genes and increased mitochondrial uncoupling respiration, which was mainly mediated by activation of the calcitonin receptor-like receptor (CRLR)·receptor activity-modifying protein 1 (RAMP1) complex and PKA and p38 MAPK signaling pathways. Importantly, this adipocyte-autonomous beiging effect by ADM2 was translatable to human primary adipocytes. In addition, M2 macrophage activation also contributed to the beiging effects of ADM2 through catecholamine secretion. Therefore, our study reveals that ADM2 enhances subcutaneous WAT beiging via a direct effect by activating the CRLR·RAMP1-cAMP/PKA and p38 MAPK pathways in white adipocytes and via an indirect effect by stimulating alternative M2 polarization in macrophages. Through both mechanisms, beiging of WAT by ADM2 results in increased energy expenditure and reduced obesity, suggesting ADM2 as a novel anti-obesity target.
Collapse
Affiliation(s)
- Ying Lv
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Song-Yang Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Xianyi Liang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Heng Zhang
- the Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhi Xu
- the Department of General Surgery, Peking University Third Hospital, Beijing 100191, China, and
| | - Bo Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Ming-Jiang Xu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Changtao Jiang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China,
| | - Jin Shang
- the Department of Cardiometabolic Disease, Merck Research Laboratories, Merck & Co, Inc., Kenilworth, New Jersey 07033
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
41
|
Transcription Profile in Sporadic Multiple Symmetric Lipomatosis Reveals Differential Expression at the Level of Adipose Tissue-Derived Stem Cells. Plast Reconstr Surg 2016; 137:1181-1190. [PMID: 27018673 DOI: 10.1097/prs.0000000000002013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The cause of the rare fat distribution disorder multiple symmetric lipomatosis is unknown. Independent reports suggest a higher proliferative activity, hormone resistance, and involvement of mitochondrial function in the disease. METHODS The authors performed morphologic comparison of affected and unaffected tissues in five unrelated patients and generated adipose-derived stem cell cultures from the tissue samples and characterized them as a possible cellular model of multiple symmetric lipomatosis evolution. The authors investigated proliferative activity and the expression of genes relevant to disease processes. RESULTS There was no difference in the morphologic appearance and the surface marker profile. Stem cells from lipomatous tissue showed significantly higher proliferative activity. Polymerase chain reaction arrays showed marked changes in genes associated with proliferation, hormonal regulation, and mitochondria. The authors show that multiple symmetric lipomatosis tissue is morphologically and histologically different from regular subcutaneous fat. CONCLUSIONS This study indicates an involvement of mesenchymal stem cells in the pathogenesis of multiple symmetric lipomatosis and that the evolution of multiple symmetric lipomatosis tissue is a process driven by an inherent defect of the respective cell clone(s). Further molecular genetics and functional analysis will be required to unravel the pathogenetic mechanism underlying the derailment in fat cell metabolism and proliferation. Here, the authors show for the first time that adipose-derived stem cells exhibit many characteristics previously described for native multiple symmetric lipomatosis fat tissue and propose that they are therefore an excellent tool for further functional investigations in multiple symmetric lipomatosis and other disorders of the fat tissue. CLINICAL QUESTION/LEVEL OF EVIDENCE Risk, V.
Collapse
|
42
|
When to eat? The influence of circadian rhythms on metabolic health: are animal studies providing the evidence? Nutr Res Rev 2016; 29:180-193. [PMID: 27364352 DOI: 10.1017/s095442241600010x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As obesity and metabolic diseases rise, there is need to investigate physiological and behavioural aspects associated with their development. Circadian rhythms have a profound influence on metabolic processes, as they prepare the body to optimise energy use and storage. Moreover, food-related signals confer temporal order to organs involved in metabolic regulation. Therefore food intake should be synchronised with the suprachiasmatic nucleus (SCN) to elaborate efficient responses to environmental challenges. Human studies suggest that a loss of synchrony between mealtime and the SCN promotes obesity and metabolic disturbances. Animal research using different paradigms has been performed to characterise the effects of timing of food intake on metabolic profiles. Therefore the purpose of the present review is to critically examine the evidence of animal studies, to provide a state of the art on metabolic findings and to assess whether the paradigms used in rodent models give the evidence to support a 'best time' for food intake. First we analyse and compare the current findings of studies where mealtime has been shifted out of phase from the light-dark cycle. Then, we analyse studies restricting meal times to different moments within the active period. So far animal studies correlate well with human studies, demonstrating that restricting food intake to the active phase limits metabolic disturbances produced by high-energy diets and that eating during the inactive/sleep phase leads to a worse metabolic outcome. Based on the latter we discuss the missing elements and possible mechanisms leading to the metabolic consequences, as these are still lacking.
Collapse
|
43
|
Harfmann BD, Schroder EA, Kachman MT, Hodge BA, Zhang X, Esser KA. Muscle-specific loss of Bmal1 leads to disrupted tissue glucose metabolism and systemic glucose homeostasis. Skelet Muscle 2016; 6:12. [PMID: 27486508 PMCID: PMC4969979 DOI: 10.1186/s13395-016-0082-x] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 02/05/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Diabetes is the seventh leading cause of death in the USA, and disruption of circadian rhythms is gaining recognition as a contributing factor to disease prevalence. This disease is characterized by hyperglycemia and glucose intolerance and symptoms caused by failure to produce and/or respond to insulin. The skeletal muscle is a key insulin-sensitive metabolic tissue, taking up ~80 % of postprandial glucose. To address the role of the skeletal muscle molecular clock to insulin sensitivity and glucose tolerance, we generated an inducible skeletal muscle-specific Bmal1 (-/-) mouse (iMSBmal1 (-/-)). RESULTS Progressive changes in body composition (decreases in percent fat) were seen in the iMSBmal1 (-/-) mice from 3 to 12 weeks post-treatment as well as glucose intolerance and non-fasting hyperglycemia. Ex vivo analysis of glucose uptake revealed that the extensor digitorum longus (EDL) muscles did not respond to either insulin or 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) stimulation. RT-PCR and Western blot analyses demonstrated a significant decrease in mRNA expression and protein content of the muscle glucose transporter (Glut4). We also found that both mRNA expression and activity of two key rate-limiting enzymes of glycolysis, hexokinase 2 (Hk2) and phosphofructokinase 1 (Pfk1), were significantly reduced in the iMSBmal1 (-/-) muscle. Lastly, results from metabolomics analyses provided evidence of decreased glycolytic flux and uncovered decreases in some tricarboxylic acid (TCA) intermediates with increases in amino acid levels in the iMSBmal1 (-/-) muscle. These findings suggest that the muscle is relying predominantly on fat as a fuel with increased protein breakdown to support the TCA cycle. CONCLUSIONS These data support a fundamental role for Bmal1, the endogenous circadian clock, in glucose metabolism in the skeletal muscle. Our findings have implicated altered molecular clock dictating significant changes in altered substrate metabolism in the absence of feeding or activity changes. The changes in body composition in our model also highlight the important role that changes in skeletal muscle carbohydrate, and fat metabolism can play in systemic metabolism.
Collapse
Affiliation(s)
- Brianna D. Harfmann
- Center for Muscle Biology, University of Kentucky, Lexington, KY USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Elizabeth A. Schroder
- Center for Muscle Biology, University of Kentucky, Lexington, KY USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Maureen T. Kachman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI USA
| | - Brian A. Hodge
- Center for Muscle Biology, University of Kentucky, Lexington, KY USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
- Present address: Department of Physiology and Functional Genomics, Myology Institute, University of Florida, Gainesville, FL 32610-0274 USA
| | - Xiping Zhang
- Center for Muscle Biology, University of Kentucky, Lexington, KY USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
- Present address: Department of Physiology and Functional Genomics, Myology Institute, University of Florida, Gainesville, FL 32610-0274 USA
| | - Karyn A. Esser
- Center for Muscle Biology, University of Kentucky, Lexington, KY USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
- Present address: Department of Physiology and Functional Genomics, Myology Institute, University of Florida, Gainesville, FL 32610-0274 USA
| |
Collapse
|
44
|
Smith J, Fahrenkrug J, Jørgensen HL, Christoffersen C, Goetze JP. Diurnal gene expression of lipolytic natriuretic peptide receptors in white adipose tissue. Endocr Connect 2015; 4:206-14. [PMID: 26286623 PMCID: PMC4566841 DOI: 10.1530/ec-15-0074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/18/2015] [Indexed: 01/01/2023]
Abstract
Disruption of the circadian rhythm can lead to obesity and cardiovascular disease. In white adipose tissue, activation of the natriuretic peptide receptors (NPRs) stimulates lipolysis. We have previously shown that natriuretic peptides are expressed in a circadian manner in the heart, but the temporal expression profile of their cognate receptors has not been examined in white adipose tissue. We therefore collected peri-renal white adipose tissue and serum from WT mice. Tissue mRNA contents of NPRs - NPR-A and NPR-C, the clock genes Per1 and Bmal1, and transcripts involved in lipid metabolism were quantified at 4-h intervals: in the diurnal study, mice were exposed to a period of 12 h light followed by 12 h darkness (n=52). In the circadian study, mice were kept in darkness for 24 h (n=47). Concomitant serum concentrations of free fatty acids, glycerol, triglycerides (TGs), and insulin were measured. Per1 and Bmal1 mRNA contents showed reciprocal circadian profiles (P<0.0001). NPR-A mRNA contents followed a temporal pattern (P=0.01), peaking in the dark (active) period. In contrast, NPR-C mRNA was expressed in an antiphase manner with nadir in the active period (P=0.007). TG concentrations in serum peaked in the active dark period (P=0.003). In conclusion, NPR-A and NPR-C gene expression is associated with the expression of clock genes in white adipose tissue. The reciprocal expression may thus contribute to regulate lipolysis and energy homeostasis in a diurnal manner.
Collapse
Affiliation(s)
- Julie Smith
- Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark
| | - Henrik L Jørgensen
- Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark Department of Clinical Biochemistry (KB3014) Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark Department of Technology Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark Department of Clinical Biochemistry Faculty of Health Sciences, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark Department of Biomedical Sciences Copenhagen University, Copenhagen, Denmark Department of Clinical Medicine Aarhus University, Aarhus, Denmark
| |
Collapse
|
45
|
Hodge BA, Wen Y, Riley LA, Zhang X, England JH, Harfmann BD, Schroder EA, Esser KA. The endogenous molecular clock orchestrates the temporal separation of substrate metabolism in skeletal muscle. Skelet Muscle 2015; 5:17. [PMID: 26000164 PMCID: PMC4440511 DOI: 10.1186/s13395-015-0039-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/13/2015] [Indexed: 01/04/2023] Open
Abstract
Background Skeletal muscle is a major contributor to whole-body metabolism as it serves as a depot for both glucose and amino acids, and is a highly metabolically active tissue. Within skeletal muscle exists an intrinsic molecular clock mechanism that regulates the timing of physiological processes. A key function of the clock is to regulate the timing of metabolic processes to anticipate time of day changes in environmental conditions. The purpose of this study was to identify metabolic genes that are expressed in a circadian manner and determine if these genes are regulated downstream of the intrinsic molecular clock by assaying gene expression in an inducible skeletal muscle-specific Bmal1 knockout mouse model (iMS-Bmal1−/−). Methods We used circadian statistics to analyze a publicly available, high-resolution time-course skeletal muscle expression dataset. Gene ontology analysis was utilized to identify enriched biological processes in the skeletal muscle circadian transcriptome. We generated a tamoxifen-inducible skeletal muscle-specific Bmal1 knockout mouse model and performed a time-course microarray experiment to identify gene expression changes downstream of the molecular clock. Wheel activity monitoring was used to assess circadian behavioral rhythms in iMS-Bmal1−/− and control iMS-Bmal1+/+ mice. Results The skeletal muscle circadian transcriptome was highly enriched for metabolic processes. Acrophase analysis of circadian metabolic genes revealed a temporal separation of genes involved in substrate utilization and storage over a 24-h period. A number of circadian metabolic genes were differentially expressed in the skeletal muscle of the iMS-Bmal1−/− mice. The iMS-Bmal1−/− mice displayed circadian behavioral rhythms indistinguishable from iMS-Bmal1+/+ mice. We also observed a gene signature indicative of a fast to slow fiber-type shift and a more oxidative skeletal muscle in the iMS-Bmal1−/− model. Conclusions These data provide evidence that the intrinsic molecular clock in skeletal muscle temporally regulates genes involved in the utilization and storage of substrates independent of circadian activity. Disruption of this mechanism caused by phase shifts (that is, social jetlag) or night eating may ultimately diminish skeletal muscle’s ability to efficiently maintain metabolic homeostasis over a 24-h period. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0039-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brian A Hodge
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Lance A Riley
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Xiping Zhang
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Jonathan H England
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Brianna D Harfmann
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Elizabeth A Schroder
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| | - Karyn A Esser
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536 USA ; Center for Muscle Biology, University of Kentucky, 800 Rose Street, Lexington, KY 40536 USA
| |
Collapse
|
46
|
Abstract
Most living beings, including humans, must adapt to rhythmically occurring daily changes in their environment that are generated by the Earth's rotation. In the course of evolution, these organisms have acquired an internal circadian timing system that can anticipate environmental oscillations and thereby govern their rhythmic physiology in a proactive manner. In mammals, the circadian timing system coordinates virtually all physiological processes encompassing vigilance states, metabolism, endocrine functions and cardiovascular activity. Research performed during the past two decades has established that almost every cell in the body possesses its own circadian timekeeper. The resulting clock network is organized in a hierarchical manner. A master pacemaker, located in the suprachiasmatic nucleus (SCN) of the hypothalamus, is synchronized every day to the photoperiod. In turn, the SCN determines the phase of the cellular clocks in peripheral organs through a wide variety of signalling pathways dependent on feeding cycles, body temperature rhythms, oscillating bloodborne signals and, in some organs, inputs of the peripheral nervous system. A major purpose of circadian clocks in peripheral tissues is the temporal orchestration of key metabolic processes, including food processing (metabolism and xenobiotic detoxification). Here, we review some recent findings regarding the molecular and cellular composition of the circadian timing system and discuss its implications for the temporal coordination of metabolism in health and disease. We focus primarily on metabolic disorders such as obesity and type 2 diabetes, although circadian misalignments (shiftwork or 'social jet lag') have also been associated with the aetiology of human malignancies.
Collapse
Affiliation(s)
- C Dibner
- Department of Endocrinology, Diabetes, Nutrition and Hypertension, University Hospital of Geneva, Geneva, Switzerland
| | | |
Collapse
|
47
|
Plikus MV, Van Spyk EN, Pham K, Geyfman M, Kumar V, Takahashi JS, Andersen B. The circadian clock in skin: implications for adult stem cells, tissue regeneration, cancer, aging, and immunity. J Biol Rhythms 2015; 30:163-82. [PMID: 25589491 DOI: 10.1177/0748730414563537] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Historically, work on peripheral circadian clocks has been focused on organs and tissues that have prominent metabolic functions, such as the liver, fat, and muscle. In recent years, skin has emerged as a model for studying circadian clock regulation of cell proliferation, stem cell functions, tissue regeneration, aging, and carcinogenesis. Morphologically, skin is complex, containing multiple cell types and structures, and there is evidence for a functional circadian clock in most, if not all, of its cell types. Despite the complexity, skin stem cell populations are well defined, experimentally tractable, and exhibit prominent daily cell proliferation cycles. Hair follicle stem cells also participate in recurrent, long-lasting cycles of regeneration: the hair growth cycles. Among other advantages of skin is a broad repertoire of available genetic tools enabling the creation of cell type-specific circadian mutants. Also, due to the accessibility of skin, in vivo imaging techniques can be readily applied to study the circadian clock and its outputs in real time, even at the single-cell level. Skin provides the first line of defense against many environmental and stress factors that exhibit dramatic diurnal variations such as solar ultraviolet (UV) radiation and temperature. Studies have already linked the circadian clock to the control of UVB-induced DNA damage and skin cancers. Due to the important role that skin plays in the defense against microorganisms, it also represents a promising model system to further explore the role of the clock in the regulation of the body's immune functions. To that end, recent studies have already linked the circadian clock to psoriasis, one of the most common immune-mediated skin disorders. Skin also provides opportunities to interrogate the clock regulation of tissue metabolism in the context of stem cells and regeneration. Furthermore, many animal species feature prominent seasonal hair molt cycles, offering an attractive model for investigating the role of the clock in seasonal organismal behaviors.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA Center for Complex Biological Systems, University of California, Irvine, Irvine, CA
| | - Elyse N Van Spyk
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA Department of Biological Chemistry, University of California, Irvine, Irvine, CA Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Kim Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA
| | | | - Vivek Kumar
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Bogi Andersen
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA Department of Biological Chemistry, University of California, Irvine, Irvine, CA Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA
| |
Collapse
|
48
|
Kolbe I, Dumbell R, Oster H. Circadian Clocks and the Interaction between Stress Axis and Adipose Function. Int J Endocrinol 2015; 2015:693204. [PMID: 26000016 PMCID: PMC4426660 DOI: 10.1155/2015/693204] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/03/2015] [Accepted: 04/03/2015] [Indexed: 01/21/2023] Open
Abstract
Many physiological processes and most endocrine functions show fluctuations over the course of the day. These so-called circadian rhythms are governed by an endogenous network of cellular clocks and serve as an adaptation to daily and, thus, predictable changes in the organism's environment. Circadian clocks have been described in several tissues of the stress axis and in adipose cells where they regulate the rhythmic and stimulated release of stress hormones, such as glucocorticoids, and various adipokine factors. Recent work suggests that both adipose and stress axis clock systems reciprocally influence each other and adrenal-adipose rhythms may be key players in the development and therapy of metabolic disorders. In this review, we summarize our current understanding of adrenal and adipose tissue rhythms and clocks and how they might interact to regulate energy homoeostasis and stress responses under physiological conditions. Potential chronotherapeutic strategies for the treatment of metabolic and stress disorders are discussed.
Collapse
Affiliation(s)
- Isa Kolbe
- Chronophysiology Group, Medical Department I, University of Lübeck, 23538 Lübeck, Germany
| | - Rebecca Dumbell
- Chronophysiology Group, Medical Department I, University of Lübeck, 23538 Lübeck, Germany
| | - Henrik Oster
- Chronophysiology Group, Medical Department I, University of Lübeck, 23538 Lübeck, Germany
- *Henrik Oster:
| |
Collapse
|
49
|
Harfmann BD, Schroder EA, Esser KA. Circadian rhythms, the molecular clock, and skeletal muscle. J Biol Rhythms 2014; 30:84-94. [PMID: 25512305 DOI: 10.1177/0748730414561638] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Circadian rhythms are the approximate 24-h biological cycles that function to prepare an organism for daily environmental changes. They are driven by the molecular clock, a transcriptional:translational feedback mechanism that in mammals involves the core clock genes Bmal1, Clock, Per1/2, and Cry1/2. The molecular clock is present in virtually all cells of an organism. The central clock in the suprachiasmatic nucleus (SCN) has been well studied, but the clocks in the peripheral tissues, such as heart and skeletal muscle, have just begun to be investigated. Skeletal muscle is one of the largest organs in the body, comprising approximately 45% of total body mass. More than 2300 genes in skeletal muscle are expressed in a circadian pattern, and these genes participate in a wide range of functions, including myogenesis, transcription, and metabolism. The circadian rhythms of skeletal muscle can be entrained both indirectly through light input to the SCN and directly through time of feeding and activity. It is critical for the skeletal muscle molecular clock not only to be entrained to the environment but also to be in synchrony with rhythms of other tissues. When circadian rhythms are disrupted, the observed effects on skeletal muscle include fiber-type shifts, altered sarcomeric structure, reduced mitochondrial respiration, and impaired muscle function. Furthermore, there are detrimental effects on metabolic health, including impaired glucose tolerance and insulin sensitivity, which skeletal muscle likely contributes to considering it is a key metabolic tissue. These data indicate a critical role for skeletal muscle circadian rhythms for both muscle and systems health. Future research is needed to determine the mechanisms of molecular clock function in skeletal muscle, identify the means by which skeletal muscle entrainment occurs, and provide a stringent comparison of circadian gene expression across the diverse tissue system of skeletal muscle.
Collapse
Affiliation(s)
- Brianna D Harfmann
- Center for Muscle Biology, Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Elizabeth A Schroder
- Center for Muscle Biology, Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Karyn A Esser
- Center for Muscle Biology, Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
50
|
Kalsbeek A, la Fleur S, Fliers E. Circadian control of glucose metabolism. Mol Metab 2014; 3:372-83. [PMID: 24944897 PMCID: PMC4060304 DOI: 10.1016/j.molmet.2014.03.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 01/15/2023] Open
Abstract
The incidence of obesity and type 2 diabetes mellitus (T2DM) has risen to epidemic proportions. The pathophysiology of T2DM is complex and involves insulin resistance, pancreatic β-cell dysfunction and visceral adiposity. It has been known for decades that a disruption of biological rhythms (which happens the most profoundly with shift work) increases the risk of developing obesity and T2DM. Recent evidence from basal studies has further sparked interest in the involvement of daily rhythms (and their disruption) in the development of obesity and T2DM. Most living organisms have molecular clocks in almost every tissue, which govern rhythmicity in many domains of physiology, such as rest/activity rhythms, feeding/fasting rhythms, and hormonal secretion. Here we present the latest research describing the specific role played by the molecular clock mechanism in the control of glucose metabolism and speculate on how disruption of these tissue clocks may lead to the disturbances in glucose homeostasis.
Collapse
Affiliation(s)
- Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands ; Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Susanne la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| |
Collapse
|