1
|
Noya SB, Sengupta A, Yue Z, Weljie A, Sehgal A. Balancing brain metabolic states during sickness and recovery sleep. Eur J Neurosci 2024; 60:6605-6616. [PMID: 39542871 PMCID: PMC11612838 DOI: 10.1111/ejn.16588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/20/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Sickness sleep and rebound following sleep deprivation share humoral signals including the rise of cytokines, in particular interleukins. Nevertheless, they represent unique physiological states with unique brain firing patterns and involvement of specific circuitry. Here, we performed untargeted metabolomics of mouse cortex and hippocampus to uncover changes with sickness and rebound sleep as compared with normal daily sleep. We found that the three settings are biochemically unique with larger differences in the cortex than in the hippocampus. Both sickness and rebound sleep shared an increase in tryptophan. Surprisingly, these two sleep conditions showed opposite modulation of the methionine-homocysteine cycle and differences in terms of the energetic signature, with sickness impinging on glycolysis intermediates whilst rebound increased the triphosphorylated form of nucleotides. These findings indicate that rebound following sleep deprivation stimulates an energy rich setting in the brain that is devoid during sickness sleep.
Collapse
Affiliation(s)
- Sara B. Noya
- Howard Hughes Medical InstituteUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Chronobiology and Sleep Institute, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational TherapeuticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Translational Medicine and TherapeuticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zhifeng Yue
- Chronobiology and Sleep Institute, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Aalim Weljie
- Department of Systems Pharmacology and Translational TherapeuticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Translational Medicine and TherapeuticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Amita Sehgal
- Howard Hughes Medical InstituteUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Chronobiology and Sleep Institute, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Luo Y, Meng X, Cui L, Wang S. Circadian Regulation of Lipid Metabolism during Pregnancy. Int J Mol Sci 2024; 25:11491. [PMID: 39519044 PMCID: PMC11545986 DOI: 10.3390/ijms252111491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A cluster of metabolic changes occur to provide energy for fetal growth and development during pregnancy. There is a burgeoning body of research highlighting the pivotal role of circadian rhythms in the pathogenesis of metabolic disorders and lipid homeostasis in mammals. Perturbations of the circadian system and lipid metabolism during gestation might be responsible for a variety of adverse reproductive outcomes comprising miscarriage, gestational diabetes mellitus, and preeclampsia. Growing studies have confirmed that resynchronizing circadian rhythms might alleviate metabolic disturbance. However, there is no clear evidence regarding the specific mechanisms by which the diurnal rhythm regulates lipid metabolism during pregnancy. In this review, we summarize previous knowledge on the strong interaction among the circadian clock, lipid metabolism, and pregnancy. Analyzing the circadian clock genes will improve our understanding of how circadian rhythms are implicated in complex lipid metabolic disorders during pregnancy. Exploring the potential of resynchronizing these circadian rhythms to disrupt abnormal lipid metabolism could also result in a breakthrough in reducing adverse pregnancy outcomes.
Collapse
Affiliation(s)
| | | | - Liyuan Cui
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China; (Y.L.); (X.M.)
| | - Songcun Wang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China; (Y.L.); (X.M.)
| |
Collapse
|
3
|
McHill AW, Melanson EL, Wright KP, Depner CM. Circadian misalignment disrupts biomarkers of cardiovascular disease risk and promotes a hypercoagulable state. Eur J Neurosci 2024; 60:5450-5466. [PMID: 39053917 DOI: 10.1111/ejn.16468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
The circadian system regulates 24-h time-of-day patterns of cardiovascular physiology, with circadian misalignment resulting in adverse cardiovascular risk. Although many proteins in the coagulation-fibrinolysis axis show 24-h time-of-day patterns, it is not understood if these temporal patterns are regulated by circadian or behavioral (e.g., sleep and food intake) cycles, or how circadian misalignment influences these patterns. Thus, we utilized a night shiftwork protocol to analyze circadian versus behavioral cycle regulation of 238 plasma proteins linked to cardiovascular physiology. Six healthy men aged 26.2 ± 5.6 years (mean ± SD) completed the protocol involving two baseline days with 8-h nighttime sleep opportunities (circadian alignment), a transition to shiftwork day, followed by 2 days of simulated night shiftwork with 8-h daytime sleep opportunities (circadian misalignment). Plasma was collected for proteomics every 4 h across 24 h during baseline and during daytime sleep and the second night shift. Cosinor analyses identified proteins with circadian or behavioral cycle-regulated 24-h time-of-day patterns. Five proteins were circadian regulated (plasminogen activator inhibitor-1, angiopoietin-2, insulin-like growth factor binding protein-4, follistatin-related protein-3, and endoplasmic reticulum resident protein-29). No cardiovascular-related proteins showed regulation by behavioral cycles. Within the coagulation pathway, circadian misalignment decreased tissue factor pathway inhibitor, increased tissue factor, and induced a 24-h time-of-day pattern in coagulation factor VII (all FDR < 0.10). Such changes in protein abundance are consistent with changes observed in hypercoagulable states. Our analyses identify circadian regulation of proteins involved in cardiovascular physiology and indicate that acute circadian misalignment could promote a hypercoagulable state, possibly contributing to elevated cardiovascular disease risk among shift workers.
Collapse
Affiliation(s)
- Andrew W McHill
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, Portland, Oregon, USA
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Edward L Melanson
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kenneth P Wright
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Christopher M Depner
- Department of Health and Kinesiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
4
|
McManus D, Patton AP, Smyllie NJ, Chin JW, Hastings MH. PERfect Day: reversible and dose-dependent control of circadian time-keeping in the mouse suprachiasmatic nucleus by translational switching of PERIOD2 protein expression. Eur J Neurosci 2024; 60:5537-5552. [PMID: 39300693 PMCID: PMC7617102 DOI: 10.1111/ejn.16537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
The biological clock of the suprachiasmatic nucleus (SCN) orchestrates circadian (approximately daily) rhythms of behaviour and physiology that underpin health. SCN cell-autonomous time-keeping revolves around a transcriptional/translational feedback loop (TTFL) within which PERIOD (PER1,2) and CRYPTOCHROME (CRY1,2) proteins heterodimerise and suppress trans-activation of their encoding genes (Per1,2; Cry1,2). To explore its contribution to SCN time-keeping, we used adeno-associated virus-mediated translational switching to express PER2 (tsPER2) in organotypic SCN slices carrying bioluminescent TTFL circadian reporters. Translational switching requires provision of the non-canonical amino acid, alkyne lysine (AlkK), for protein expression. Correspondingly, AlkK, but not vehicle, induced constitutive expression of tsPER2 in SCN neurons and reversibly and dose-dependently suppressed pPer1-driven transcription in PER-deficient (Per1,2-null) SCN, illustrating the potency of PER2 in negative regulation within the TTFL. Constitutive expression of tsPER2, however, failed to initiate circadian oscillations in arrhythmic PER-deficient SCN. In rhythmic, PER-competent SCN, AlkK dose-dependently reduced the amplitude of PER2-reported oscillations as inhibition by tsPER2 progressively damped the TTFL. tsPER2 also dose-dependently lengthened the period of the SCN TTFL and neuronal calcium rhythms. Following wash-out of AlkK to remove tsPER2, the SCN regained TTFL amplitude and period. Furthermore, SCN retained their pre-washout phase: the removal of tsPER2 did not phase-shift the TTFL. Given that constitutive tsCRY1 can regulate TTFL amplitude and period, but also reset TTFL phase and initiate rhythms in CRY-deficient SCN, these results reveal overlapping and distinct properties of PER2 and CRY1 within the SCN, and emphasise the utility of translational switching to explore the functions of circadian proteins.
Collapse
Affiliation(s)
- David McManus
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew P Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicola J Smyllie
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason W Chin
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Michael H Hastings
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
5
|
Buijink MR, van Weeghel M, Harms A, Murli DS, Meijer JH, Hankemeier T, Michel S, Kervezee L. Loss of temporal coherence in the circadian metabolome across multiple tissues during ageing in mice. Eur J Neurosci 2024; 60:3843-3857. [PMID: 38802069 DOI: 10.1111/ejn.16428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
Circadian clock function declines with ageing, which can aggravate ageing-related diseases such as type 2 diabetes and neurodegenerative disorders. Understanding age-related changes in the circadian system at a systemic level can contribute to the development of strategies to promote healthy ageing. The goal of this study was to investigate the impact of ageing on 24-h rhythms in amine metabolites across four tissues in young (2 months of age) and old (22-25 months of age) mice using a targeted metabolomics approach. Liver, plasma, the suprachiasmatic nucleus (SCN; the location of the central circadian clock in the hypothalamus) and the paraventricular nucleus (PVN; a downstream target of the SCN) were collected from young and old mice every 4 h during a 24-h period (n = 6-7 mice per group). Differential rhythmicity analysis revealed that ageing impacts 24-h rhythms in the amine metabolome in a tissue-specific manner. Most profound changes were observed in the liver, in which rhythmicity was lost in 60% of the metabolites in aged mice. Furthermore, we found strong correlations in metabolite levels between the liver and plasma and between the SCN and the PVN in young mice. These correlations were almost completely abolished in old mice. These results indicate that ageing is accompanied by a severe loss of the circadian coordination between tissues and by disturbed rhythmicity of metabolic processes. The tissue-specific impact of ageing may help to differentiate mechanisms of ageing-related disorders in the brain versus peripheral tissues and thereby contribute to the development of potential therapies for these disorders.
Collapse
Affiliation(s)
- M Renate Buijink
- Laboratory for Neurophysiology, Department of Cellular and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel van Weeghel
- Laboratory for Neurophysiology, Department of Cellular and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Amy Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Devika S Murli
- Laboratory for Neurophysiology, Department of Cellular and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johanna H Meijer
- Laboratory for Neurophysiology, Department of Cellular and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Stephan Michel
- Laboratory for Neurophysiology, Department of Cellular and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Laura Kervezee
- Laboratory for Neurophysiology, Department of Cellular and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Pivovarova-Ramich O, Malin SK. Editorial: Circadian rhythm in obesity. Front Endocrinol (Lausanne) 2024; 15:1387889. [PMID: 38577573 PMCID: PMC10993243 DOI: 10.3389/fendo.2024.1387889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 04/06/2024] Open
Affiliation(s)
- Olga Pivovarova-Ramich
- Research Group Molecular Nutritional Medicine and Department of Human Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Endocrinology and Metabolism, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Steven K. Malin
- Department of Kinesiology and Health, Rutgers University, New Brunswick, NJ, United States
- Division of Endocrinology, Metabolism and Nutrition, Rutgers University, New Brunswick, NJ, United States
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, United States
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, NJ, United States
| |
Collapse
|
7
|
Lee CH, Murrell CE, Chu A, Pan X. Circadian Regulation of Apolipoproteins in the Brain: Implications in Lipid Metabolism and Disease. Int J Mol Sci 2023; 24:17415. [PMID: 38139244 PMCID: PMC10743770 DOI: 10.3390/ijms242417415] [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: 11/02/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
The circadian rhythm is a 24 h internal clock within the body that regulates various factors, including sleep, body temperature, and hormone secretion. Circadian rhythm disruption is an important risk factor for many diseases including neurodegenerative illnesses. The central and peripheral oscillators' circadian clock network controls the circadian rhythm in mammals. The clock genes govern the central clock in the suprachiasmatic nucleus (SCN) of the brain. One function of the circadian clock is regulating lipid metabolism. However, investigations of the circadian regulation of lipid metabolism-associated apolipoprotein genes in the brain are lacking. This review summarizes the rhythmic expression of clock genes and lipid metabolism-associated apolipoprotein genes within the SCN in Mus musculus. Nine of the twenty apolipoprotein genes identified from searching the published database (SCNseq and CircaDB) are highly expressed in the SCN. Most apolipoprotein genes (ApoE, ApoC1, apoA1, ApoH, ApoM, and Cln) show rhythmic expression in the brain in mice and thus might be regulated by the master clock. Therefore, this review summarizes studies on lipid-associated apolipoprotein genes in the SCN and other brain locations, to understand how apolipoproteins associated with perturbed cerebral lipid metabolism cause multiple brain diseases and disorders. This review describes recent advancements in research, explores current questions, and identifies directions for future research.
Collapse
Affiliation(s)
- Chaeeun Hannah Lee
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Charlotte Ellzabeth Murrell
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Alexander Chu
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| |
Collapse
|
8
|
Alvarez-García L, Sánchez-García FJ, Vázquez-Pichardo M, Moreno-Altamirano MM. Chikungunya Virus, Metabolism, and Circadian Rhythmicity Interplay in Phagocytic Cells. Metabolites 2023; 13:1143. [PMID: 37999239 PMCID: PMC10672914 DOI: 10.3390/metabo13111143] [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: 10/13/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/25/2023] Open
Abstract
Chikungunya virus (CHIKV) is transmitted to humans by mosquitoes of the genus Aedes, causing the chikungunya fever disease, associated with inflammation and severe articular incapacitating pain. There has been a worldwide reemergence of chikungunya and the number of cases increased to 271,006 in 2022 in the Americas alone. The replication of CHIKV takes place in several cell types, including phagocytic cells. Monocytes and macrophages are susceptible to infection by CHIKV; at the same time, they provide protection as components of the innate immune system. However, in host-pathogen interactions, CHIKV might have the ability to alter the function of immune cells, partly by rewiring the tricarboxylic acid cycle. Some viral evasion mechanisms depend on the metabolic reprogramming of immune cells, and the cell metabolism is intertwined with circadian rhythmicity; thus, a circadian immunovirometabolism axis may influence viral pathogenicity. Therefore, analyzing the interplay between viral infection, circadian rhythmicity, and cellular metabolic reprogramming in human macrophages could shed some light on the new field of immunovirometabolism and eventually contribute to the development of novel drugs and therapeutic approaches based on circadian rhythmicity and metabolic reprogramming.
Collapse
Affiliation(s)
- Linamary Alvarez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del IPN, Prolongación de Carpio y Plan de Ayala s/n, Col. Casco de Santo Tomás, Mexico City 11340, Mexico; (L.A.-G.); (F.J.S.-G.); (M.V.-P.)
| | - F. Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del IPN, Prolongación de Carpio y Plan de Ayala s/n, Col. Casco de Santo Tomás, Mexico City 11340, Mexico; (L.A.-G.); (F.J.S.-G.); (M.V.-P.)
| | - Mauricio Vázquez-Pichardo
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del IPN, Prolongación de Carpio y Plan de Ayala s/n, Col. Casco de Santo Tomás, Mexico City 11340, Mexico; (L.A.-G.); (F.J.S.-G.); (M.V.-P.)
- Laboratorio de Arbovirus, Departamento de Virología, Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE), Secretaría de Salud, Francisco de P. Miranda 177, Col. Lomas de Plateros, Mexico City 01480, Mexico
| | - M. Maximina Moreno-Altamirano
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas del IPN, Prolongación de Carpio y Plan de Ayala s/n, Col. Casco de Santo Tomás, Mexico City 11340, Mexico; (L.A.-G.); (F.J.S.-G.); (M.V.-P.)
| |
Collapse
|
9
|
de Assis LVM, Demir M, Oster H. Nonalcoholic Steatohepatitis Disrupts Diurnal Liver Transcriptome Rhythms in Mice. Cell Mol Gastroenterol Hepatol 2023; 16:341-354. [PMID: 37270062 PMCID: PMC10444956 DOI: 10.1016/j.jcmgh.2023.05.008] [Citation(s) in RCA: 1] [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: 01/25/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND & AIMS The liver ensures organismal homeostasis through modulation of physiological functions over the course of the day. How liver diseases such as nonalcoholic steatohepatitis (NASH) affect daily transcriptome rhythms in the liver remains elusive. METHODS To start closing this gap, we evaluated the impact of NASH on the diurnal regulation of the liver transcriptome in mice. In addition, we investigated how stringent consideration of circadian rhythmicity affects the outcomes of NASH transcriptome analyses. RESULTS Comparative rhythm analysis of the liver transcriptome from diet-induced NASH and control mice showed an almost 3-hour phase advance in global gene expression rhythms. Rhythmically expressed genes associated with DNA repair and cell-cycle regulation showed increased overall expression and circadian amplitude. In contrast, lipid and glucose metabolism-associated genes showed loss of circadian amplitude, reduced overall expression, and phase advances in NASH livers. Comparison of NASH-induced liver transcriptome responses between published studies showed little overlap (12%) in differentially expressed genes (DEGs). However, by controlling for sampling time and using circadian analytical tools, a 7-fold increase in DEG detection was achieved compared with methods without time control. CONCLUSIONS NASH had a strong effect on circadian liver transcriptome rhythms with phase- and amplitude-specific effects for key metabolic and cell repair pathways, respectively. Accounting for circadian rhythms in NASH transcriptome studies markedly improves DEG detection and enhances reproducibility.
Collapse
Affiliation(s)
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany; Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain Behavior and Metabolism, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
10
|
Hohor S, Mandanach C, Maftei A, Zugravu CA, Oțelea MR. Impaired Melatonin Secretion, Oxidative Stress and Metabolic Syndrome in Night Shift Work. Antioxidants (Basel) 2023; 12:antiox12040959. [PMID: 37107334 PMCID: PMC10135726 DOI: 10.3390/antiox12040959] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/08/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic syndrome has been associated in many studies with working in shifts. Even if the mechanistic details are not fully understood, forced sleep deprivation and exposure to light, as happens during night shifts, or irregular schedules with late or very early onset of the working program, lead to a sleep-wake rhythm misalignment, metabolic dysregulation and oxidative stress. The cyclic melatonin secretion is regulated by the hypothalamic suprachiasmatic nuclei and light exposure. At a central level, melatonin promotes sleep and inhibits wake-signals. Beside this role, melatonin acts as an antioxidant and influences the functionality of the cardiovascular system and of different metabolic processes. This review presents data about the influence of night shifts on melatonin secretion and oxidative stress. Assembling data from epidemiological, experimental and clinical studies contributes to a better understanding of the pathological links between chronodisruption and the metabolic syndrome related to working in shifts.
Collapse
Affiliation(s)
- Sorina Hohor
- Doctoral School, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Cristina Mandanach
- Doctoral School, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Andreea Maftei
- Doctoral School, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- "Dr. Carol Davila" Central Military Emergency University Hospital, 134 Calea Plevnei, Sector 1, 010242 Bucharest, Romania
| | - Corina Aurelia Zugravu
- Department of Hygiene and Ecology, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Marina Ruxandra Oțelea
- Clinical Department 5, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| |
Collapse
|
11
|
Pivovarova-Ramich O, Zimmermann HG, Paul F. Multiple sclerosis and circadian rhythms: Can diet act as a treatment? Acta Physiol (Oxf) 2023; 237:e13939. [PMID: 36700353 DOI: 10.1111/apha.13939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/15/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory and neurodegenerative disease of the central nervous system (CNS) with increasing incidence and prevalence. MS is associated with inflammatory and metabolic disturbances that, as preliminary human and animal data suggest, might be mediated by disruption of circadian rhythmicity. Nutrition habits can influence the risk for MS, and dietary interventions may be effective in modulating MS disease course. Chronotherapeutic approaches such as time-restricted eating (TRE) may benefit people with MS by stabilizing the circadian clock and restoring immunological and metabolic rhythms, thus potentially counteracting disease progression. This review provides a summary of selected studies on dietary intervention in MS, circadian rhythms, and their disruption in MS, including clock gene variations, circadian hormones, and retino-hypothalamic tract changes. Furthermore, we present studies that reported diurnal variations in MS, which might result from circadian disruption. And lastly, we suggest how chrononutritive approaches like TRE might counteract MS disease activity.
Collapse
Affiliation(s)
- Olga Pivovarova-Ramich
- Research Group Molecular Nutritional Medicine, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Hanna Gwendolyn Zimmermann
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Einstein Center Digital Future, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max-Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
12
|
Disruption of the Expression of the Placental Clock and Melatonin Genes in Preeclampsia. Int J Mol Sci 2023; 24:ijms24032363. [PMID: 36768691 PMCID: PMC9917141 DOI: 10.3390/ijms24032363] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Circadian rhythms have been described in numerous tissues of living organisms and are necessary for homeostasis. The understanding of their role in normal and pathological pregnancy is only just emerging. It has been established that clock genes are expressed in the placenta of animals and humans, but the rhythmicity of placenta immune cells is not known. Macrophages from healthy placenta of women at term were isolated and the expression of clock genes BMAL1, CLOCK, PER2, CRY2, and NR1D1 was assessed by qRT-PCR every 4 h over 24 h. Raw data were treated with cosinor analysis to evaluate the significance of the oscillations. Placental macrophages exhibited significant circadian expression of clock genes but one third of placental macrophages lost clock gene rhythmicity; the clock gene oscillations were restored by co-culture with trophoblasts. We wondered if melatonin, a key hormone regulating circadian rhythm, was involved in the oscillations of placental cells. We showed that macrophages and trophoblasts produced melatonin and expressed MT2 receptor. In women who developed preeclampsia during pregnancy, circadian oscillations of placental macrophages were lost and could not be rescued by coculture with trophoblasts from healthy women. Moreover, production and oscillations of melatonin were altered in preeclamptic macrophages. For the first time to our knowledge, this study shows circadian rhythms and melatonin production by placental macrophages. It also shows that preeclampsia is associated with a disruption of the circadian rhythm of placental cells. These results represent a new scientific breakthrough that may contribute to the prevention and treatment of obstetrical pathologies.
Collapse
|
13
|
Wishart DS, Rout M, Lee BL, Berjanskii M, LeVatte M, Lipfert M. Practical Aspects of NMR-Based Metabolomics. Handb Exp Pharmacol 2023; 277:1-41. [PMID: 36271165 DOI: 10.1007/164_2022_613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
While NMR-based metabolomics is only about 20 years old, NMR has been a key part of metabolic and metabolism studies for >40 years. Historically, metabolic researchers used NMR because of its high level of reproducibility, superb instrument stability, facile sample preparation protocols, inherently quantitative character, non-destructive nature, and amenability to automation. In this chapter, we provide a short history of NMR-based metabolomics. We then provide a detailed description of some of the practical aspects of performing NMR-based metabolomics studies including sample preparation, pulse sequence selection, and spectral acquisition and processing. The two different approaches to metabolomics data analysis, targeted vs. untargeted, are briefly outlined. We also describe several software packages to help users process NMR spectra obtained via these two different approaches. We then give several examples of useful or interesting applications of NMR-based metabolomics, ranging from applications to drug toxicology, to identifying inborn errors of metabolism to analyzing the contents of biofluids from dairy cattle. Throughout this chapter, we will highlight the strengths and limitations of NMR-based metabolomics. Additionally, we will conclude with descriptions of recent advances in NMR hardware, methodology, and software and speculate about where NMR-based metabolomics is going in the next 5-10 years.
Collapse
Affiliation(s)
- David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada.
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada.
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Manoj Rout
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Brian L Lee
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Mark Berjanskii
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Marcia LeVatte
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Matthias Lipfert
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
- Reference Standard Management & NMR QC, Lonza Group AG, Visp, Switzerland
| |
Collapse
|
14
|
Jiang S, Chen L, Huang ZL, Chen CR. Role of the paraventricular nucleus of the hypothalamus in sleep–wake regulation. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The paraventricular nucleus of the hypothalamus (PVH) acts as a cohesive functional unit that regulates neuroendocrine and autonomic function, complex behavior, and negative emotions after stress. However, how the PVH integrates arousal with these biological functions has only recently been explored. Clinical reports, combined with neurotoxic lesioning, immunochemistry, neuronal activity recordings, and the polysomnographic analyses of genetically modified animals, have revealed that the PVH is important for the control of wakefulness. Here, we review emerging anatomical and neural mechanisms for sleep–wake regulation in the PVH to support its essential role in the promotion and maintenance of wakefulness.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Lu Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| | - Chang-Rui Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College Fudan University, Shanghai 200030, China
| |
Collapse
|
15
|
Yong YN, Henry CJ, Haldar S. Is There a Utility of Chrono-Specific Diets in Improving Cardiometabolic Health? Mol Nutr Food Res 2022; 66:e2200043. [PMID: 35856629 DOI: 10.1002/mnfr.202200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/31/2022] [Indexed: 11/10/2022]
Abstract
Modern lifestyle is generally associated with the consumption of three main meals per day, one of which is typically in the evening or at night. It is also well established that consumption of meals in the later part of the day, notably in the evenings, is associated with circadian desynchrony, which in turn increases the risk of non-communicable diseases, particularly cardiometabolic diseases. While it is not feasible to avoid food consumption during the evenings altogether, there is an opportunity to provide chrono-specific, diet-based solutions to mitigate some of these risks. To date, there has been substantial progress in the understanding of chrononutrition, with evidence derived mainly from in vitro and in vivo animal studies. Some of these approaches include the manipulation of the quality and quantity of certain nutrients to be consumed at specific times of the day, as well as incorporating certain dietary components (macronutrients, micronutrients, or non-nutrient bioactives, including polyphenols) with the ability to modulate circadian rhythmicity. However, robust human studies are generally lacking. In this review, the study has consolidated and critically appraised the current evidence base, with an aim to translate these findings to improve cardiometabolic health and provides recommendations to move this field forward.
Collapse
Affiliation(s)
- Yi Ning Yong
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117599, Singapore
| | - Sumanto Haldar
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, 117599, Singapore
| |
Collapse
|
16
|
Colom-Pellicer M, Rodríguez RM, Soliz-Rueda JR, de Assis LVM, Navarro-Masip È, Quesada-Vázquez S, Escoté X, Oster H, Mulero M, Aragonès G. Proanthocyanidins Restore the Metabolic Diurnal Rhythm of Subcutaneous White Adipose Tissue According to Time-Of-Day Consumption. Nutrients 2022; 14:2246. [PMID: 35684049 PMCID: PMC9182881 DOI: 10.3390/nu14112246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Consumption of grape seed proanthocyanidin extract (GSPE) has beneficial effects on the functionality of white adipose tissue (WAT). However, although WAT metabolism shows a clear diurnal rhythm, whether GSPE consumption could affect WAT rhythmicity in a time-dependent manner has not been studied. Ninety-six male Fischer rats were fed standard (STD, two groups) or cafeteria (CAF, four groups) diet for 9 weeks (n = 16 each group). From week 6 on, CAF diet animals were supplemented with vehicle or 25 mg GSPE/kg of body weight either at the beginning of the light/rest phase (ZT0) or at the beginning of the dark/active phase (ZT12). The two STD groups were also supplemented with vehicle at ZT0 or ZT12. In week 9, animals were sacrificed at 6 h intervals (n = 4) to analyze the diurnal rhythms of subcutaneous WAT metabolites by nuclear magnetic resonance spectrometry. A total of 45 metabolites were detected, 19 of which presented diurnal rhythms in the STD groups. Although most metabolites became arrhythmic under CAF diet, GSPE consumption at ZT12, but not at ZT0, restored the rhythmicity of 12 metabolites including compounds involved in alanine, aspartate, and glutamate metabolism. These results demonstrate that timed GSPE supplementation may restore, at least partially, the functional dynamics of WAT when it is consumed at the beginning of the active phase. This study opens an innovative strategy for time-dependent polyphenol treatment in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Marina Colom-Pellicer
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Romina M. Rodríguez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Jorge R. Soliz-Rueda
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Leonardo Vinícius Monteiro de Assis
- Center of Brain, Behavior and Metabolism, Institute of Neurobiology, University of Lübeck, Marie Curie Street, 23562 Lübeck, Germany; (L.V.M.d.A.); (H.O.)
| | - Èlia Navarro-Masip
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Sergio Quesada-Vázquez
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (X.E.)
| | - Xavier Escoté
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (X.E.)
| | - Henrik Oster
- Center of Brain, Behavior and Metabolism, Institute of Neurobiology, University of Lübeck, Marie Curie Street, 23562 Lübeck, Germany; (L.V.M.d.A.); (H.O.)
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| |
Collapse
|
17
|
Hamzeiy H, Ferretti D, Robles MS, Cox J. Perseus plugin "Metis" for metabolic-pathway-centered quantitative multi-omics data analysis for static and time-series experimental designs. CELL REPORTS METHODS 2022; 2:100198. [PMID: 35497496 PMCID: PMC9046241 DOI: 10.1016/j.crmeth.2022.100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 01/14/2022] [Accepted: 03/28/2022] [Indexed: 11/22/2022]
Abstract
We introduce Metis, a new plugin for the Perseus software aimed at analyzing quantitative multi-omics data based on metabolic pathways. Data from different omics types are connected through reactions of a genome-scale metabolic-pathway reconstruction. Metabolite concentrations connect through the reactants, while transcript, protein, and protein post-translational modification (PTM) data are associated through the enzymes catalyzing the reactions. Supported experimental designs include static comparative studies and time-series data. As an example for the latter, we combine circadian mouse liver multi-omics data and study the contribution of cycles of phosphoproteome and metabolome to enzyme activity regulation. Our analysis resulted in 52 pairs of cycling phosphosites and metabolites connected through a reaction. The time lags between phosphorylation and metabolite peak show non-uniform behavior, indicating a major contribution of phosphorylation in the modulation of enzymatic activity.
Collapse
Affiliation(s)
- Hamid Hamzeiy
- Computational Systems Biochemistry Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Daniela Ferretti
- Computational Systems Biochemistry Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Maria S. Robles
- Institute of Medical Psychology, Faculty of Medicine, LMU, Munich, Germany
| | - Jürgen Cox
- Computational Systems Biochemistry Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| |
Collapse
|
18
|
Xu L, Lin J, Liu Y, Hua B, Cheng Q, Lin C, Yan Z, Wang Y, Sun N, Qian R, Lu C. CLOCK regulates Drp1 mRNA stability and mitochondrial homeostasis by interacting with PUF60. Cell Rep 2022; 39:110635. [PMID: 35417690 DOI: 10.1016/j.celrep.2022.110635] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 01/26/2022] [Accepted: 03/16/2022] [Indexed: 12/16/2022] Open
Abstract
Circadian genes such as Clock, Bmal1, Cryptochrome1/2, and Period1/2/3 constitute the precise circadian system. ClockΔ19 is a commonly used mouse model harboring a circadian clock gene mutation, which lacks the EXON-19-encoded 51 amino acids. Previous reports have shown that ClockΔ19 mice have severe metabolic abnormalities. Here, we report that the mitochondria of ClockΔ19 mice exhibit excessive fission and dysfunction. We also demonstrate that CLOCK binds to the RNA-binding protein PUF60 through its EXON 19. Further, we find that PUF60 directly maintains mitochondrial homeostasis through regulating Drp1 mRNA stability, while the association with CLOCK can competitively inhibit this function. In ClockΔ19 mice, CLOCKΔ19 releases PUF60, leading to enhanced Drp1 mRNA stability and persistent mitochondrial fission. Our results reveal a direct post-transcriptional role of CLOCK in regulating mitochondrial homeostasis via Drp1 mRNA stability and that the loss of EXON 19 of CLOCK in ClockΔ19 mice leads to severe mitochondrial homeostasis disorders.
Collapse
Affiliation(s)
- Lirong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jiaxin Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yutong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Bingxuan Hua
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qianyun Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Changpo Lin
- Institute of Vascular Surgery, Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yaping Wang
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ning Sun
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214122, China.
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
19
|
Park J, Kim J, Yun Y, Han D, Kim K, Hong J, Cho S. Daily injection of melatonin inhibits insulin resistance induced by chronic mealtime shift. Physiol Rep 2022; 10:e15227. [PMID: 35343087 PMCID: PMC8958345 DOI: 10.14814/phy2.15227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 06/14/2023] Open
Abstract
Shift work disorders have become an emerging concern worldwide. Shift disorders encompass a wide range of illnesses that have yet to be identified. The study focused on the relationship between shift work disorders and insulin resistance. Previously, it was reported that advancing the usual mealtime of mice triggered insulin resistance. Here, the hypothesis that chronic mealtime shifts induce oxidative damage leading to chronic diseases such as type 2 diabetes was tested. It was found that mealtime shift causes imbalances between anti-oxidative capacity and reactive oxygen species (ROS) levels, indicating increased oxidative damage during the light/rest phase. This study further demonstrated that daily supplementation of antioxidants at the appropriate time of day inhibited insulin resistance caused by chronic mealtime shifts, suggesting significant and chronic health implications for shift workers. In conclusion, it was confirmed that increased ROS levels caused by mealtime shift induce insulin resistance, which is inhibited by the antioxidant melatonin.
Collapse
Affiliation(s)
- Jihyun Park
- Department of NeuroscienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Jichul Kim
- Department of Life & Nanopharmaceutical ScienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Yejin Yun
- Department of Biomedical ScienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Dong‐Hee Han
- Department of NeuroscienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
- Present address:
Ildong Pharmaceutical Co. LtdSeocho‐guSeoulRepublic of Korea
| | - Kyungjin Kim
- Department of Brain ScienceDGISTDaeguSouth Korea
| | - Jongki Hong
- College of PharmacyKyung Hee UniversitySeoulSouth Korea
| | - Sehyung Cho
- Department of NeuroscienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
- Department of PhysiologyKyung Hee University School of MedicineSeoulSouth Korea
| |
Collapse
|
20
|
Yildirim E, Curtis R, Hwangbo DS. Roles of peripheral clocks: lessons from the fly. FEBS Lett 2022; 596:263-293. [PMID: 34862983 PMCID: PMC8844272 DOI: 10.1002/1873-3468.14251] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023]
Abstract
To adapt to and anticipate rhythmic changes in the environment such as daily light-dark and temperature cycles, internal timekeeping mechanisms called biological clocks evolved in a diverse set of organisms, from unicellular bacteria to humans. These biological clocks play critical roles in organisms' fitness and survival by temporally aligning physiological and behavioral processes to the external cues. The central clock is located in a small subset of neurons in the brain and drives daily activity rhythms, whereas most peripheral tissues harbor their own clock systems, which generate metabolic and physiological rhythms. Since the discovery of Drosophila melanogaster clock mutants in the early 1970s, the fruit fly has become an extensively studied model organism to investigate the mechanism and functions of circadian clocks. In this review, we primarily focus on D. melanogaster to survey key discoveries and progresses made over the past two decades in our understanding of peripheral clocks. We discuss physiological roles and molecular mechanisms of peripheral clocks in several different peripheral tissues of the fly.
Collapse
Affiliation(s)
| | - Rachel Curtis
- Department of Biology, University of Louisville, Louisville, KY, USA
| | - Dae-Sung Hwangbo
- Department of Biology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
21
|
Artati A, Prehn C, Lutter D, Dyar KA. Untargeted and Targeted Circadian Metabolomics Using Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) and Flow Injection-Electrospray Ionization-Tandem Mass Spectrometry (FIA-ESI-MS/MS). Methods Mol Biol 2022; 2482:311-327. [PMID: 35610436 DOI: 10.1007/978-1-0716-2249-0_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A diverse array of 24-h oscillating hormones and metabolites direct and reflect circadian clock function. Circadian metabolomics uses advanced high-throughput analytical chemistry techniques to comprehensively profile these small molecules (<1.5 kDa) across 24 h in cells, media, body fluids, breath, tissues, and subcellular compartments. The goals of circadian metabolomics experiments are often multifaceted. These include identifying and tracking rhythmic metabolic inputs and outputs of central and peripheral circadian clocks, quantifying endogenous free-running period, monitoring relative phase alignment between clocks, and mapping pathophysiological consequences of clock disruption or misalignment. Depending on the particular experimental question, samples are collected under free-running or entrained conditions. Here we describe both untargeted and targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) and flow injection-electrospray ionization-tandem mass spectrometry (FIA-ESI-MS/MS) based assays we have used for circadian metabolomics studies. We discuss tissue homogenization, chemical derivatization, measurement, and tips for data processing, normalization, scaling, how to handle outliers, and imputation of missing values.
Collapse
Affiliation(s)
- Anna Artati
- Metabolomics and Proteomics Core Facility, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core Facility, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Dominik Lutter
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Kenneth Allen Dyar
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Metabolic Physiology, Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
22
|
Vallianatou T, Lin W, Bèchet NB, Correia MSP, Shanbhag NC, Lundgaard I, Globisch D. Differential regulation of oxidative stress, microbiota-derived, and energy metabolites in the mouse brain during sleep. J Cereb Blood Flow Metab 2021; 41:3324-3338. [PMID: 34293940 PMCID: PMC8669215 DOI: 10.1177/0271678x211033358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/25/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022]
Abstract
Sleep has evolved as a universal core function to allow for restorative biological processes. Detailed knowledge of metabolic changes necessary for the sleep state in the brain is missing. Herein, we have performed an in-depth metabolic analysis of four mouse brain regions and uncovered region-specific circadian variations. Metabolites linked to oxidative stress were altered during sleep including acylcarnitines, hydroxylated fatty acids, phenolic compounds, and thiol-containing metabolites. These findings provide molecular evidence of a significant metabolic shift of the brain energy metabolism. Specific alterations were observed for brain metabolites that have previously not been associated with a circadian function including the microbiome-derived metabolite ergothioneine that suggests a regulatory function. The pseudopeptide β-citryl-glutamate has been linked to brain development and we have now discovered a previously unknown regioisomer. These metabolites altered by the circadian rhythm represent the foundation for hypothesis-driven studies of the underlying metabolic processes and their function.
Collapse
Affiliation(s)
- Theodosia Vallianatou
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Weifeng Lin
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Nicholas B Bèchet
- Department of Experimental Medical Science, Lund University, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund University, Lund, Sweden
| | - Mario SP Correia
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Nagesh C Shanbhag
- Department of Experimental Medical Science, Lund University, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund University, Lund, Sweden
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund University, Lund, Sweden
| | - Daniel Globisch
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Role of circadian rhythm and impact of circadian rhythm disturbance on the metabolism and disease. J Cardiovasc Pharmacol 2021; 79:254-263. [PMID: 34840256 DOI: 10.1097/fjc.0000000000001178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/23/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Molecular circadian clocks exist in almost all cells of the organism and operate for approximately 24 h, maintain the normal physiological and behavioral body processes and regulate metabolism of many cells related to a variety of disease states. Circadian rhythms regulate metabolism, mainly including neurotransmitters, hormones, amino acids and lipids. Circadian misalignment is related to metabolic syndromes, such as obesity, diabetes and hypertension, which have reached an alarming level in modern society. We reviewed the mechanism of the circadian clock and the interaction between circadian rhythm and metabolism, as well as circadian rhythm disturbance on the metabolism of hypertension, obesity and diabetes. Finally, we discuss how to use the circadian rhythm to prevent diseases. Thus, this review is a micro to macro discussion from the perspective of circadian rhythm and aims to provide basic ideas for circadian rhythm research and disease therapies.
Collapse
|
24
|
Abstract
A molecular circadian clock exists not only in the brain, but also in most cells of the body. Research over the past two decades has demonstrated that it directs daily rhythmicity of nearly every aspect of metabolism. It also consolidates sleep-wake behavior each day into an activity/feeding period and a sleep/fasting period. Otherwise, sleep-wake states are mostly controlled by hypothalamic and thalamic regulatory circuits in the brain that direct overall brain state. Recent evidence suggests that hypothalamic control of appetite and metabolism may be concomitant with sleep-wake regulation, and even share the same control centers. Thus, circadian control of metabolic pathways might be overlaid by sleep-wake control of the same pathways, providing a flexible and redundant system to modify metabolism according to both activity and environment.
Collapse
|
25
|
Feng C, Liu W, Chen H, Dong W, Yang J. Effect of dark environment on intestinal flora and expression of genes related to liver metabolism in zebrafish (Danio rerio). Comp Biochem Physiol C Toxicol Pharmacol 2021; 249:109100. [PMID: 34174412 DOI: 10.1016/j.cbpc.2021.109100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023]
Abstract
To explore the effects of dark environment on intestinal flora and expression of genes related to liver metabolism in zebrafish, a total of 60 zebrafish were fed for 21 days (24 h dark treatments or 14/10 h light/dark cycle), and the influence of dark environment on gut microbes and liver gene expression was studied using sequencing analysis of intestinal flora and liver. The results showed that the body weight of fish was significantly increased in the dark group than that in the control group (P < 0.05). Compared with the control group, dark environment treatment changed the composition of dominant flora, increased the abundance of unconventional bacteria and reduced probiotics in the intestine of zebrafish. Of these, the ratio of Bacteroidetes to Firmicutes in the intestine was reduced. The genome expression of the liver showed significant changes, and liver metabolites were also affected. Meanwhile, dark environment decreased gene expression associated with changes in blood glucose, lipid metabolism and immunization. Dark environment also caused liver steatosis as observed by histological study. This study shows that dark environment treatment has an important impact on liver metabolism and intestinal microbes in zebrafish.
Collapse
Affiliation(s)
- Chi Feng
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China
| | - Wuyun Liu
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China; School of Animal Science, Mongolian State University of Agriculture, Bayangol, Ulaanbaatar, Mongolia
| | - Hao Chen
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China
| | - Wu Dong
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China
| | - Jingfeng Yang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China.
| |
Collapse
|
26
|
Xu L, Liu Y, Cheng Q, Shen Y, Yuan Y, Jiang X, Li X, Guo D, Jiang J, Lin C. Bmal1 Downregulation Worsens Critical Limb Ischemia by Promoting Inflammation and Impairing Angiogenesis. Front Cardiovasc Med 2021; 8:712903. [PMID: 34447794 PMCID: PMC8384109 DOI: 10.3389/fcvm.2021.712903] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Critical limb ischemia (CLI) is the most advanced clinical stage of peripheral vascular disease with high mobility and mortality. CLI patients suffer from lower extremity rest pain, ulceration, and gangrene caused by insufficient blood and oxygen supply. Seeking for effective biomarkers and therapeutic targets is of great significance for improving the life quality of CLI patients. The circadian clock has been reported to be involved in the progression of kinds of cardiovascular diseases. Whether and how circadian genes play a role in CLI remains unknown. In this study, by collecting femoral artery and muscle specimens of CLI patients who underwent amputation, we confirmed that the circadian gene Bmal1 is downregulated in the CLI femoral artery and ischemic distal lower limb muscle. Furthermore, we verified that Bmal1 affects CLI by regulating lipid metabolism, inflammation, and angiogenesis. A hindlimb ischemia model performed in wild-type and Bmal1−/− mice confirmed that Bmal1 disruption would lead to impaired angiogenesis. In vitro experiments indicated that the decreased expression of Bmal1 would increase ox-LDL uptake and impair endothelial cell functions, including proliferation, migration, and tube formation. As for mechanisms, Bmal1 represses inflammation by inhibiting lipid uptake and by activating IL-10 transcription and promotes angiogenesis by transcriptionally regulating VEGF expression. In conclusion, we provide evidence that the circadian gene Bmal1 plays an important role in CLI by inhibiting inflammation and promoting angiogenesis. Thus, Bmal1 may be an effective biomarker and a potential therapeutic target in CLI.
Collapse
Affiliation(s)
- Lirong Xu
- Department of Pathology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Physiology and Pathophysiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Yutong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Qianyun Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Yuan
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolang Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Li
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daqiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junhao Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changpo Lin
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Abstract
The objective of chronotherapy is to optimize medical treatments taking into account the body's circadian rhythms. Chronotherapy is referred to and practiced in two different ways: (1) to alter the sleep-wake rhythms of patients to improve the sequels of several pathologies; (2) to take into account the circadian rhythms of patients to improve therapeutics. Even minor dysfunction of the biological clock can greatly affect sleep/wake physiology causing excessive diurnal somnolence, increase in sleep onset latency, phase delays or advances in sleep onset, frequent night awakenings, reduced sleep efficiency, delayed and shortened rapid eye movement sleep, or increased periodic leg movements. Chronotherapy aims to restore the proper circadian pattern of the sleep-wake cycle, through adequate sleep hygiene, timed light exposure, and the use of chronobiotic medications, such as melatonin, that affect the output phase of circadian rhythms, thus controlling the clock. Concerning the second use of chronotherapy, therapeutic outcomes as diverse as the survival after open-heart surgery or the efficacy and tolerance to chemotherapy vary according to the time of day. However, humans are heterogeneous concerning the timing of their internal clocks. Not only different chronotypes exist but also the endogenous human circadian period (τ) is not a stable trait as it depends on many internal and external factors. If any scheduled therapeutic intervention is going to be optimized, a tool is needed for simple diagnostic and objectively measurement of an individual's internal time at any given time. Methodologic advances like the use of single-sample gene expression and metabolomics are discussed.
Collapse
Affiliation(s)
- Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Gregory M Brown
- Department of Psychiatry, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
28
|
Grima-Reyes M, Martinez-Turtos A, Abramovich I, Gottlieb E, Chiche J, Ricci JE. Physiological impact of in vivo stable isotope tracing on cancer metabolism. Mol Metab 2021; 53:101294. [PMID: 34256164 PMCID: PMC8358691 DOI: 10.1016/j.molmet.2021.101294] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/30/2021] [Accepted: 07/08/2021] [Indexed: 11/25/2022] Open
Abstract
Background There is growing interest in the analysis of tumor metabolism to identify cancer-specific metabolic vulnerabilities and therapeutic targets. Finding of such candidate metabolic pathways mainly relies on the highly sensitive identification and quantitation of numerous metabolites and metabolic fluxes using metabolomics and isotope tracing analyses. However, nutritional requirements and metabolic routes used by cancer cells cultivated in vitro do not always reflect the metabolic demands of malignant cells within the tumor milieu. Therefore, to understand how the metabolism of tumor cells in its physiological environment differs from that of normal cells, these analyses must be performed in vivo. Scope of Review This review covers the physiological impact of the exogenous administration of a stable isotope tracer into cancer animal models. We discuss specific aspects of in vivo isotope tracing protocols based on discrete bolus injections of a labeled metabolite: the tracer administration per se and the fasting period prior to it. In addition, we illustrate the complex physiological scenarios that arise when studying tumor metabolism – by isotopic labeling in animal models fed with a specific amino acid restricted diet. Finally, we provide strategies to minimize these limitations. Major Conclusions There is growing evidence that metabolic dependencies in cancers are influenced by tissue environment, cancer lineage, and genetic events. An increasing number of studies describe discrepancies in tumor metabolic dependencies when studied in in vitro settings or in vivo models, including cancer patients. Therefore, in-depth in vivo profiling of tumor metabolic routes within the appropriate pathophysiological environment will be key to identify relevant alterations that contribute to cancer onset and progression. In vivo isotope tracing is the state-of-the-art approach to study tumor metabolism. In vivo tracer administration challenges the physiological metabolism of mice. Interorgan conversion of the tracer might confound tumor labeling patterns. Mouse fasting before in vivo tracing impacts on systemic and tumor metabolism. Optimization is key to minimize physiological alterations linked to in vivo tracing.
Collapse
Affiliation(s)
- Manuel Grima-Reyes
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France
| | - Adriana Martinez-Turtos
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Eyal Gottlieb
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Johanna Chiche
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France
| | - Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice, France; Equipe labellisée LIGUE Contre le Cancer, Nice, France.
| |
Collapse
|
29
|
Metabolic control of daily locomotor activity mediated by tachykinin in Drosophila. Commun Biol 2021; 4:693. [PMID: 34099879 PMCID: PMC8184744 DOI: 10.1038/s42003-021-02219-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 05/14/2021] [Indexed: 12/20/2022] Open
Abstract
Metabolism influences locomotor behaviors, but the understanding of neural curcuit control for that is limited. Under standard light-dark cycles, Drosophila exhibits bimodal morning (M) and evening (E) locomotor activities that are controlled by clock neurons. Here, we showed that a high-nutrient diet progressively extended M activity but not E activity. Drosophila tachykinin (DTk) and Tachykinin-like receptor at 86C (TkR86C)-mediated signaling was required for the extension of M activity. DTk neurons were anatomically and functionally connected to the posterior dorsal neuron 1s (DN1ps) in the clock neuronal network. The activation of DTk neurons reduced intracellular Ca2+ levels in DN1ps suggesting an inhibitory connection. The contacts between DN1ps and DTk neurons increased gradually over time in flies fed a high-sucrose diet, consistent with the locomotor behavior. DN1ps have been implicated in integrating environmental sensory inputs (e.g., light and temperature) to control daily locomotor behavior. This study revealed that DN1ps also coordinated nutrient information through DTk signaling to shape daily locomotor behavior. Lee and colleagues report the effect of a high-sucrose diet on Drosophila locomotor activity via DTk-TkR86C neuropeptide signalling. This signalling pattern appears to involve a circadian element, with pacemaker neuron involvement having a possible time-of-day effect on locomotor behaviour.
Collapse
|
30
|
Subramanian P, Jayapalan JJ, Abdul-Rahman PS. Inevitable interlinks between biological clock and metabolism in mammals. BIOL RHYTHM RES 2021. [DOI: 10.1080/09291016.2021.1928386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Perumal Subramanian
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram Tamil Nadu, India
| | - Jaime Jacqueline Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, Department of Molecular Medicine, University of Malaya, Kuala Lumpur Malaysia
| | - Puteri Shafinaz Abdul-Rahman
- Department of Molecular Medicine, Faculty of Medicine, Department of Molecular Medicine, University of Malaya, Kuala Lumpur Malaysia
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
31
|
Lee Y, Fong SY, Shon J, Zhang SL, Brooks R, Lahens NF, Chen D, Dang CV, Field JM, Sehgal A. Time-of-day specificity of anticancer drugs may be mediated by circadian regulation of the cell cycle. SCIENCE ADVANCES 2021; 7:eabd2645. [PMID: 33579708 PMCID: PMC7880601 DOI: 10.1126/sciadv.abd2645] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/24/2020] [Indexed: 05/04/2023]
Abstract
Circadian rhythms are an integral part of physiology, underscoring their relevance for the treatment of disease. We conducted cell-based high-throughput screening to investigate time-of-day influences on the activity of known antitumor agents and found that many compounds exhibit daily rhythms of cytotoxicity concomitant with previously reported oscillations of target genes. Rhythmic action of HSP90 inhibitors was mediated by specific isoforms of HSP90, genetic perturbation of which affected the cell cycle. Furthermore, clock mutants affected the cell cycle in parallel with abrogating rhythms of cytotoxicity, and pharmacological inhibition of the cell cycle also eliminated rhythmic drug effects. An HSP90 inhibitor reduced growth rate of a mouse melanoma in a time-of-day-specific manner, but efficacy was impaired in clock-deficient tumors. These results provide a powerful rationale for appropriate daily timing of anticancer drugs and suggest circadian regulation of the cell cycle within the tumor as an underlying mechanism.
Collapse
Affiliation(s)
- Yool Lee
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shi Yi Fong
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joy Shon
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shirley L Zhang
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebekah Brooks
- Cell and Molecular Biology Graduate Group (CAMB), University of Pennsylvania, Philadelphia, PA 19104, USA
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dechun Chen
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chi Van Dang
- Ludwig Institute for Cancer Research, New York, NY 10017, USA
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jeffrey M Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Schuppelius B, Peters B, Ottawa A, Pivovarova-Ramich O. Time Restricted Eating: A Dietary Strategy to Prevent and Treat Metabolic Disturbances. Front Endocrinol (Lausanne) 2021; 12:683140. [PMID: 34456861 PMCID: PMC8387818 DOI: 10.3389/fendo.2021.683140] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Time-restricted eating (TRE), a dietary approach limiting the daily eating window, has attracted increasing attention in media and research. The eating behavior in our modern society is often characterized by prolonged and erratic daily eating patterns, which might be associated with increased risk of obesity, diabetes, and cardiovascular diseases. In contrast, recent evidence suggests that TRE might support weight loss, improve cardiometabolic health, and overall wellbeing, but the data are controversial. The present work reviews how TRE affects glucose and lipid metabolism based on clinical trials published until June 2021. A range of trials demonstrated that TRE intervention lowered fasting and postprandial glucose levels in response to a standard meal or oral glucose tolerance test, as well as mean 24-h glucose and glycemic excursions assessed using continuous glucose monitoring. In addition, fasting insulin decreases and improvement of insulin sensitivity were demonstrated. These changes were often accompanied by the decrease of blood triglyceride and cholesterol levels. However, a number of studies found that TRE had either adverse or no effects on glycemic and lipid traits, which might be explained by the different study designs (i.e., fasting/eating duration, daytime of eating, changes of calorie intake, duration of intervention) and study subject cohorts (metabolic status, age, gender, chronotype, etc.). To summarize, TRE represents an attractive and easy-to-adapt dietary strategy for the prevention and therapy of glucose and lipid metabolic disturbances. However, carefully controlled future TRE studies are needed to confirm these effects to understand the underlying mechanisms and assess the applicability of personalized interventions.
Collapse
Affiliation(s)
- Bettina Schuppelius
- Research Group Molecular Nutritional Medicine, Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Beeke Peters
- Research Group Molecular Nutritional Medicine, Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Human Nutrition and Food Science, Faculty of Agriculture and Food Sciences, Christian-Albrecht-University Kiel, Kiel, Germany
| | - Agnieszka Ottawa
- Research Group Molecular Nutritional Medicine, Department of Molecular Toxicology, 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, Berlin, Germany
| | - Olga Pivovarova-Ramich
- Research Group Molecular Nutritional Medicine, Department of Molecular Toxicology, 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, Berlin, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- *Correspondence: Olga Pivovarova-Ramich,
| |
Collapse
|
33
|
Cervela-Cardona L, Yoshida T, Zhang Y, Okada M, Fernie A, Mas P. Circadian Control of Metabolism by the Clock Component TOC1. FRONTIERS IN PLANT SCIENCE 2021; 12:683516. [PMID: 34194455 PMCID: PMC8238050 DOI: 10.3389/fpls.2021.683516] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/18/2021] [Indexed: 05/11/2023]
Abstract
Photosynthesis in chloroplasts during the day and mitochondrial respiration during the night execute nearly opposing reactions that are coordinated with the internal cellular status and the external conditions. Here, we describe a mechanism by which the Arabidopsis clock component TIMING OF CAB EXPRESSION1 (TOC1) contributes to the diurnal regulation of metabolism. Proper expression of TOC1 is important for sustaining cellular energy and for the diel and circadian oscillations of sugars, amino acids and tricarboxylic acid (TCA) cycle intermediates. TOC1 binds to the promoter of the TCA-related gene FUMARASE 2 to repress its expression at night, which results in decreased fumarate accumulation in TOC1 over-expressing plants and increased in toc1-2 mutant. Genetic interaction studies confirmed that over-expression of FUMARASE 2 in TOC1 over-expressing plants alleviates the molecular and physiological energy-deprivation phenotypes of TOC1 over-expressing plants. Thus, we propose that the tandem TOC1-FUMARASE 2 is one of the mechanisms that contribute to the regulation of plant metabolism during the day and night.
Collapse
Affiliation(s)
- Luis Cervela-Cardona
- Centre for Research in Agricultural Genomics, CSIC-IRTA-UAB-UB, Barcelona, Spain
| | - Takuya Yoshida
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Potsdam, Germany
| | - Youjun Zhang
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Potsdam, Germany
- Center of Plant Systems Biology and Plant Biotechnology, Plovdiv, Bulgaria
| | - Masaaki Okada
- Centre for Research in Agricultural Genomics, CSIC-IRTA-UAB-UB, Barcelona, Spain
| | - Alisdair Fernie
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Potsdam, Germany
- Center of Plant Systems Biology and Plant Biotechnology, Plovdiv, Bulgaria
| | - Paloma Mas
- Centre for Research in Agricultural Genomics, CSIC-IRTA-UAB-UB, Barcelona, Spain
- Consejo Superior de Investigaciones Científicas, Barcelona, Spain
- *Correspondence: Paloma Mas,
| |
Collapse
|
34
|
Maric A, Mas P. Chromatin Dynamics and Transcriptional Control of Circadian Rhythms in Arabidopsis. Genes (Basel) 2020; 11:E1170. [PMID: 33036236 PMCID: PMC7601625 DOI: 10.3390/genes11101170] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
Circadian rhythms pervade nearly all aspects of plant growth, physiology, and development. Generation of the rhythms relies on an endogenous timing system or circadian clock that generates 24-hour oscillations in multiple rhythmic outputs. At its bases, the plant circadian function relies on dynamic interactive networks of clock components that regulate each other to generate rhythms at specific phases during the day and night. From the initial discovery more than 13 years ago of a parallelism between the oscillations in chromatin status and the transcriptional rhythms of an Arabidopsis clock gene, a number of studies have later expanded considerably our view on the circadian epigenome and transcriptome landscapes. Here, we describe the most recent identification of chromatin-related factors that are able to directly interact with Arabidopsis clock proteins to shape the transcriptional waveforms of circadian gene expression and clock outputs. We discuss how changes in chromatin marks associate with transcript initiation, elongation, and the rhythms of nascent RNAs, and speculate on future interesting research directions in the field.
Collapse
Affiliation(s)
- Aida Maric
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
| | - Paloma Mas
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, 08193 Barcelona, Spain;
- Consejo Superior de Investigaciones Científicas (CSIC), 08028 Barcelona, Spain
| |
Collapse
|
35
|
Brami-Cherrier K, Lewis RG, Cervantes M, Liu Y, Tognini P, Baldi P, Sassone-Corsi P, Borrelli E. Cocaine-mediated circadian reprogramming in the striatum through dopamine D2R and PPARγ activation. Nat Commun 2020; 11:4448. [PMID: 32895370 PMCID: PMC7477550 DOI: 10.1038/s41467-020-18200-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Substance abuse disorders are linked to alteration of circadian rhythms, although the molecular and neuronal pathways implicated have not been fully elucidated. Addictive drugs, such as cocaine, induce a rapid increase of dopamine levels in the brain. Here, we show that acute administration of cocaine triggers reprogramming in circadian gene expression in the striatum, an area involved in psychomotor and rewarding effects of drugs. This process involves the activation of peroxisome protein activator receptor gamma (PPARγ), a nuclear receptor involved in inflammatory responses. PPARγ reprogramming is altered in mice with cell-specific ablation of the dopamine D2 receptor (D2R) in the striatal medium spiny neurons (MSNs) (iMSN-D2RKO). Administration of a specific PPARγ agonist in iMSN-D2RKO mice elicits substantial rescue of cocaine-dependent control of circadian genes. These findings have potential implications for development of strategies to treat substance abuse disorders. Drugs of abuse have been shown to perturb circadian rhythms. Here, the authors show in mice that cocaine exposure modulates circadian gene expression in the striatum through a previously unappreciated pathway that involves dopamine D2 receptors and the nuclear receptor PPARγ.
Collapse
Affiliation(s)
- Karen Brami-Cherrier
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA
| | - Robert G Lewis
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA
| | - Marlene Cervantes
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA
| | - Yu Liu
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Paola Tognini
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA.
| | - Emiliana Borrelli
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
36
|
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: 53] [Impact Index Per Article: 13.3] [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
|
37
|
Satyamitra MM, Cassatt DR, Hollingsworth BA, Price PW, Rios CI, Taliaferro LP, Winters TA, DiCarlo AL. Metabolomics in Radiation Biodosimetry: Current Approaches and Advances. Metabolites 2020; 10:metabo10080328. [PMID: 32796693 PMCID: PMC7465152 DOI: 10.3390/metabo10080328] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Triage and medical intervention strategies for unanticipated exposure during a radiation incident benefit from the early, rapid and accurate assessment of dose level. Radiation exposure results in complex and persistent molecular and cellular responses that ultimately alter the levels of many biological markers, including the metabolomic phenotype. Metabolomics is an emerging field that promises the determination of radiation exposure by the qualitative and quantitative measurements of small molecules in a biological sample. This review highlights the current role of metabolomics in assessing radiation injury, as well as considerations for the diverse range of bioanalytical and sampling technologies that are being used to detect these changes. The authors also address the influence of the physiological status of an individual, the animal models studied, the technology and analysis employed in interrogating response to the radiation insult, and variables that factor into discovery and development of robust biomarker signatures. Furthermore, available databases for these studies have been reviewed, and existing regulatory guidance for metabolomics are discussed, with the ultimate goal of providing both context for this area of radiation research and the consideration of pathways for continued development.
Collapse
Affiliation(s)
- Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
- Correspondence: ; Tel.: +1-240-669-5432
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Brynn A. Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Paul W. Price
- Office of Regulatory Affairs, Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA;
| | - Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| |
Collapse
|
38
|
Guo D, Zhu Y, Wang H, Wang G, Wang C, Ren H. E3 ubiquitin ligase HRD1 modulates the circadian clock through regulation of BMAL1 stability. Exp Ther Med 2020; 20:2639-2648. [PMID: 32765757 PMCID: PMC7401958 DOI: 10.3892/etm.2020.8988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/29/2020] [Indexed: 12/20/2022] Open
Abstract
Circadian rhythm serves an essential role in numerous physiological functions. Circadian oscillations are organized by circadian clock components at the molecular level. The precision of the circadian clock is controlled by transcriptional-translational negative feedback loops, as well as post-translational modifications of clock proteins, including ubiquitination; however, the influence of E3 ligases on clock protein ubiquitination requires further investigation. The results of co-immunoprecipitation and immunofluorescent localization, indicated that the endoplasmic reticulum transmembrane E3 ubiquitin ligase HRD1, encoded by the synoviolin 1 gene, interacted with brain and muscle ARNT-like 1 (BMAL1) and enhanced BMAL1 protein ubiquitination. In addition, the results of western blotting and reverse transcription-quantitative PCR suggested that HRD1 promoted K48-associated polyubiquitination of BMAL1 and thus mediated its degradation via the ubiquitin-proteasome system. Furthermore, gene knockdown and gene overexpression assays revealed that HRD1-dependent degradation of BMAL1 protein regulated the expression of BMAL1 target genes and the amplitude of circadian oscillations in mammalian cells. The findings of the current study indicate that HRD1 may influence the regulation of circadian rhythm via modulation of BMAL1 stability.
Collapse
Affiliation(s)
- Dongkai Guo
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Yao Zhu
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Hongfeng Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Cheng Wang
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Haigang Ren
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China.,Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
39
|
Aon MA, Bernier M, Mitchell SJ, Di Germanio C, Mattison JA, Ehrlich MR, Colman RJ, Anderson RM, de Cabo R. Untangling Determinants of Enhanced Health and Lifespan through a Multi-omics Approach in Mice. Cell Metab 2020; 32:100-116.e4. [PMID: 32413334 PMCID: PMC8214079 DOI: 10.1016/j.cmet.2020.04.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/20/2019] [Accepted: 04/24/2020] [Indexed: 12/17/2022]
Abstract
The impact of chronic caloric restriction (CR) on health and survival is complex with poorly understood underlying molecular mechanisms. A recent study in mice addressing the diets used in nonhuman primate CR studies found that while diet composition did not impact longevity, fasting time and total calorie intake were determinant for increased survival. Here, integrated analysis of physiological and multi-omics data from ad libitum, meal-fed, or CR animals was used to gain insight into pathways associated with improved health and survival. We identified a potential involvement of the glycine-serine-threonine metabolic axis in longevity and related molecular mechanisms. Direct comparison of the different feeding strategies unveiled a pattern of shared pathways of improved health that included short-chain fatty acids and essential PUFA metabolism. These findings were recapitulated in the serum metabolome from nonhuman primates. We propose that the pathways identified might be targeted for their potential role in healthy aging.
Collapse
Affiliation(s)
- Miguel A Aon
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Laboratory of Cardiovascular Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Sarah J Mitchell
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Clara Di Germanio
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Margaux R Ehrlich
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ricki J Colman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Rozalyn M Anderson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53715, USA; Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
40
|
Malik DM, Paschos GK, Sehgal A, Weljie AM. Circadian and Sleep Metabolomics Across Species. J Mol Biol 2020; 432:3578-3610. [PMID: 32376454 PMCID: PMC7781158 DOI: 10.1016/j.jmb.2020.04.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Under normal circadian function, metabolic control is temporally coordinated across tissues and behaviors with a 24-h period. However, circadian disruption results in negative consequences for metabolic homeostasis including energy or redox imbalances. Yet, circadian disruption has become increasingly prevalent within today's society due to many factors including sleep loss. Metabolic consequences of both have been revealed by metabolomics analyses of circadian biology and sleep. Specifically, two primary analytical platforms, mass spectrometry and nuclear magnetic resonance spectroscopy, have been used to study molecular clock and sleep influences on overall metabolic rhythmicity. For example, human studies have demonstrated the prevalence of metabolic rhythms in human biology, as well as pan-metabolome consequences of sleep disruption. However, human studies are limited to peripheral metabolic readouts primarily through minimally invasive procedures. For further tissue- and organism-specific investigations, a number of model systems have been studied, based upon the conserved nature of both the molecular clock and sleep across species. Here we summarize human studies as well as key findings from metabolomics studies using mice, Drosophila, and zebrafish. While informative, a limitation in existing literature is a lack of interpretation regarding dynamic synthesis or catabolism within metabolite pools. To this extent, future work incorporating isotope tracers, specific metabolite reporters, and single-cell metabolomics may provide a means of exploring dynamic activity in pathways of interest.
Collapse
Affiliation(s)
- Dania M Malik
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Georgios K Paschos
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Penn Chronobiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Jiao X, Lu D, Pei X, Qi D, Huang S, Song Z, Gu J, Li Z. Type 1 diabetes mellitus impairs diurnal oscillations in murine extraorbital lacrimal glands. Ocul Surf 2020; 18:438-452. [PMID: 32360784 DOI: 10.1016/j.jtos.2020.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE People with diabetes are at high risk of lacrimal gland dysfunction, but the underlying mechanism is not well understood. In this study, we determined how type 1 diabetes mellitus (T1DM) influences circadian homeostasis of the murine extraorbital lacrimal glands (ELGs). METHODS A T1DM animal model was established by systemic streptozotocin injection in C57BL/6J mice. After 5 weeks, ELGs were collected at 3-h intervals over a 24-h circadian cycle. Total extracted RNA was subjected to high-throughput RNA sequencing, and rhythmic transcriptional data were evaluated using the Jonckheere-Terpstra-Kendall algorithm, Kyoto Encyclopedia of Genes and Genomes pathway analysis, Phase Set Enrichment Analysis, and time series cluster analysis to determine the phase, rhythmicity, and unique signature of the transcripts over temporally coordinated expression. Additionally, mass, cell size, histology, and tear secretion of the ELGs were evaluated. RESULTS T1DM globally altered the composition of the ELG transcriptome. Specifically, T1DM significantly reprogrammed the circadian transcriptomic profiles of normal ELGs and reorganized core clock machinery. Unique temporal and clustering enrichment pathways were also rewired by T1DM. Finally, normal daily rhythms of mass, cell size, and tear secretion of mouse ELGs were significantly impaired by streptozotocin-induced diabetes. CONCLUSIONS T1DM significantly reprograms the diurnal oscillations of the lacrimal glands and impairs their structure and tear secretion. This information may reveal potential targets for improving lacrimal gland dysfunction in patients with diabetes.
Collapse
Affiliation(s)
- Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jianqin Gu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.
| |
Collapse
|
42
|
Ulgherait M, Chen A, McAllister SF, Kim HX, Delventhal R, Wayne CR, Garcia CJ, Recinos Y, Oliva M, Canman JC, Picard M, Owusu-Ansah E, Shirasu-Hiza M. Circadian regulation of mitochondrial uncoupling and lifespan. Nat Commun 2020; 11:1927. [PMID: 32317636 PMCID: PMC7174288 DOI: 10.1038/s41467-020-15617-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/11/2020] [Indexed: 12/24/2022] Open
Abstract
Because old age is associated with defects in circadian rhythm, loss of circadian regulation is thought to be pathogenic and contribute to mortality. We show instead that loss of specific circadian clock components Period (Per) and Timeless (Tim) in male Drosophila significantly extends lifespan. This lifespan extension is not mediated by canonical diet-restriction longevity pathways but is due to altered cellular respiration via increased mitochondrial uncoupling. Lifespan extension of per mutants depends on mitochondrial uncoupling in the intestine. Moreover, upregulated uncoupling protein UCP4C in intestinal stem cells and enteroblasts is sufficient to extend lifespan and preserve proliferative homeostasis in the gut with age. Consistent with inducing a metabolic state that prevents overproliferation, mitochondrial uncoupling drugs also extend lifespan and inhibit intestinal stem cell overproliferation due to aging or even tumorigenesis. These results demonstrate that circadian-regulated intestinal mitochondrial uncoupling controls longevity in Drosophila and suggest a new potential anti-aging therapeutic target.
Collapse
Affiliation(s)
- Matt Ulgherait
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Anna Chen
- Columbia College, New York, NY, 10027, USA
| | | | - Han X Kim
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Rebecca Delventhal
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Charlotte R Wayne
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Christian J Garcia
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Yocelyn Recinos
- Department of Systems Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | | | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Martin Picard
- Departments of Psychiatry and Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| |
Collapse
|
43
|
Melatonin Orchestrates Lipid Homeostasis through the Hepatointestinal Circadian Clock and Microbiota during Constant Light Exposure. Cells 2020; 9:cells9020489. [PMID: 32093272 PMCID: PMC7072737 DOI: 10.3390/cells9020489] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
Misalignment between natural light rhythm and modern life activities induces disruption of the circadian rhythm. It is mainly evident that light at night (LAN) interferes with the human endocrine system and contributes to the increasing rates of obesity and lipid metabolic disease. Maintaining hepatointestinal circadian homeostasis is vital for improving lipid homeostasis. Melatonin is a chronobiotic substance that plays a main role in stabilizing bodily rhythm and has shown beneficial effects in protecting against obesity. Based on the dual effect of circadian rhythm regulation and antiobesity, we tested the effect of melatonin in mice under constant light exposure. Exposure to 24-h constant light (LL) increased weight and insulin resistance compared with those of the control group (12-h light–12-h dark cycle, LD), and simultaneous supplementation in the melatonin group (LLM) ameliorated this phenotype. Constant light exposure disturbed the expression pattern of a series of transcripts, including lipid metabolism, circadian regulation and nuclear receptors in the liver. Melatonin also showed beneficial effects in improving lipid metabolism and circadian rhythm homeostasis. Furthermore, the LL group had increased absorption and digestion of lipids in the intestine as evidenced by the elevated influx of lipids in the duodenum and decrease in the efflux of lipids in the jejunum. More interestingly, melatonin ameliorated the gut microbiota dysbiosis and improved lipid efflux from the intestine. Thus, these findings offer a novel clue regarding the obesity-promoting effect attributed to LAN and suggest a possibility for obesity therapy by melatonin in which melatonin could ameliorate rhythm disorder and intestinal dysbiosis.
Collapse
|
44
|
Wang H, Wang X, Qi D, Sun M, Hou Q, Li Y, Jiang H. Establishment of the circadian metabolic phenotype strategy in spontaneously hypertensive rats: a dynamic metabolomics study. J Transl Med 2020; 18:38. [PMID: 31992312 PMCID: PMC6988197 DOI: 10.1186/s12967-020-02222-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Circadian rhythms play a fundamental role in the progression of cardiovascular events. Almost all cardiovascular diseases have a circadian misalignment usually characterized by changes in metabolites. This study aimed to dynamically monitor rhythmic biomarkers, to elucidate the metabolic pathways that are potentially under circadian control in spontaneously hypertensive rats (SHRs), and to eventually establish a circadian metabolic phenotype strategy based on metabolomics. METHODS In this study, an untargeted metabolomics technology was used to dynamically monitor changes in serum metabolites between SHR model group and WKY control group. Liquid chromatography-mass spectrometry (LC-MS) combined with multivariate statistical analysis was applied to identify markers of hypertension rhythm imbalance. The concentrations of amino acids and their metabolites identified as markers were quantified by a subsequent targeted metabolomics analysis. Overall, these approaches comprehensively explored the rhythm mechanism and established a circadian metabolic phenotype strategy. RESULTS The metabolic profile revealed a disorder in the diurnal metabolism pattern in SHRs. Moreover, multivariate statistical analysis revealed metabolic markers of rhythm homeostasis, such as arginine, proline, phenylalanine, citric acid, L-malic acid, succinic acid, etc., accompanied by an imbalance in hypertension. The key metabolic pathways related to rhythm imbalance in hypertension were found by enrichment analysis, including amino acid metabolism, and the tricarboxylic acid cycle (TCA). In addition, the quantitative analysis of amino acids and their metabolites showed that the changes in leucine, isoleucine, valine, taurine, serine, and glycine were the most obvious. CONCLUSIONS In summary, this study illustrated the relationship between metabolites and the pathways across time on hypertension. These results may provide a theoretical basis for personalized treatment programmes and timing for hypertension.
Collapse
Affiliation(s)
- Huanjun Wang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Xiaoming Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Dongmei Qi
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Mengjia Sun
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Qingqing Hou
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Yunlun Li
- TCM Clinical Research Base for Hypertension, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China.
| | - Haiqiang Jiang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China. .,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China. .,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
| |
Collapse
|
45
|
Pan X, Taylor MJ, Cohen E, Hanna N, Mota S. Circadian Clock, Time-Restricted Feeding and Reproduction. Int J Mol Sci 2020; 21:ijms21030831. [PMID: 32012883 PMCID: PMC7038040 DOI: 10.3390/ijms21030831] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022] Open
Abstract
The goal of this review was to seek a better understanding of the function and differential expression of circadian clock genes during the reproductive process. Through a discussion of how the circadian clock is involved in these steps, the identification of new clinical targets for sleep disorder-related diseases, such as reproductive failure, will be elucidated. Here, we focus on recent research findings regarding circadian clock regulation within the reproductive system, shedding new light on circadian rhythm-related problems in women. Discussions on the roles that circadian clock plays in these reproductive processes will help identify new clinical targets for such sleep disorder-related diseases.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
- Correspondence:
| | - Meredith J. Taylor
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Emma Cohen
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Nazeeh Hanna
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Department of Pediatrics, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| |
Collapse
|
46
|
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: 46] [Impact Index Per Article: 11.5] [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
|
47
|
Ai Z, Udalova IA. Transcriptional regulation of neutrophil differentiation and function during inflammation. J Leukoc Biol 2020; 107:419-430. [PMID: 31951039 DOI: 10.1002/jlb.1ru1219-504rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/30/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in innate immunity where they elicit powerful effector functions to eliminate invading pathogens and modulate the adaptive as well as the innate immune response. Neutrophil function must be tightly regulated during inflammation and infection to avoid additional tissue damage. Increasing evidence suggests that transcription factors (TFs) function as key regulators to modulate transcriptional output, thereby controlling cell fate decision and the inflammatory responses. However, the molecular mechanisms underlying neutrophil differentiation and function during inflammation remain largely uncharacterized. Here, we provide a comprehensive overview of TFs known to be crucial for neutrophil maturation and in the signaling pathways that control neutrophil differentiation and activation. We also outline how emerging genomic and single-cell technologies may facilitate further discovery of neutrophil transcriptional regulators.
Collapse
Affiliation(s)
- Zhichao Ai
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Pan X, Mota S, Zhang B. Circadian Clock Regulation on Lipid Metabolism and Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:53-66. [PMID: 32705594 PMCID: PMC8593891 DOI: 10.1007/978-981-15-6082-8_5] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The basic helix-loop-helix-PAS transcription factor (CLOCK, Circadian locomotor output cycles protein kaput) was discovered in 1994 as a circadian clock. Soon after its discovery, the circadian clock, Aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL, also call BMAL1), was shown to regulate adiposity and body weight by controlling on the brain hypothalamic suprachiasmatic nucleus (SCN). Farther, circadian clock genes were determined to exert several of lipid metabolic and diabetes effects, overall indicating that CLOCK and BMAL1 act as a central master circadian clock. A master circadian clock acts through the neurons and hormones, with expression in the intestine, liver, kidney, lung, heart, SCN of brain, and other various cell types of the organization. Among circadian clock genes, numerous metabolic syndromes are the most important in the regulation of food intake (via regulation of circadian clock genes or clock-controlled genes in peripheral tissue), which lead to a variation in plasma phospholipids and tissue phospholipids. Circadian clock genes affect the regulation of transporters and proteins included in the regulation of phospholipid metabolism. These genes have recently received increasing recognition because a pharmacological target of circadian clock genes may be of therapeutic worth to make better resistance against insulin, diabetes, obesity, metabolism syndrome, atherosclerosis, and brain diseases. In this book chapter, we focus on the regulation of circadian clock and summarize its phospholipid effect as well as discuss the chemical, physiology, and molecular value of circadian clock pathway regulation for the treatment of plasma lipids and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA.
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA
| | - Boyang Zhang
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA
| |
Collapse
|
49
|
Abstract
The neural mechanisms of sleep, a fundamental biological behavior from invertebrates to humans, have been a long-standing mystery and present an enormous challenge. Gradually, perspectives on the neurobiology of sleep have been more various with the technical innovations over the recent decades, and studies have now identified many specific neural circuits that selectively regulate the initiation and maintenance of wake, rapid eye movement (REM) sleep, and non-REM (NREM) sleep. The cholinergic system in basal forebrain (BF) that fire maximally during waking and REM sleep is one of the key neuromodulation systems related to waking and REM sleep. Here we outline the recent progress of the BF cholinergic system in sleep-wake cycle. The intricate local connectivity and multiple projections to other cortical and subcortical regions of the BF cholinergic system elaborately presented here form a conceptual framework for understanding the coordinating effects with the dissecting regions. This framework also provides evidences regarding the relationships between the general anesthesia and wakefulness/sleep cycle focusing on the neural circuitry of unconsciousness induced by anesthetic drugs.
Collapse
|
50
|
Iino K, Mitobe Y, Ikeda K, Takayama KI, Suzuki T, Kawabata H, Suzuki Y, Horie-Inoue K, Inoue S. RNA-binding protein NONO promotes breast cancer proliferation by post-transcriptional regulation of SKP2 and E2F8. Cancer Sci 2019; 111:148-159. [PMID: 31733123 PMCID: PMC6942431 DOI: 10.1111/cas.14240] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/19/2019] [Accepted: 11/05/2019] [Indexed: 12/18/2022] Open
Abstract
The majority of breast cancers are primarily hormone‐sensitive and can be managed by endocrine therapy, although therapy‐resistant or hormone‐refractory cancers need alternative treatments. Recently, increasing attention is being paid to RNA‐binding proteins (RBP) in cancer pathophysiology. The precise role of RBP in breast cancer, however, remains to be clarified. We herein show that an RBP non‐POU domain‐containing octamer binding (NONO) plays a critical role in the pathophysiology of breast cancers regardless of their hormone dependency. Clinicopathological and immunohistochemical study of 127 breast cancer cases showed that NONO is a significant independent prognostic factor for breast cancer patients. Notably, siRNA‐mediated NONO knockdown substantially repressed the proliferation of both hormone‐sensitive MCF‐7 and hormone‐refractory MB‐MDA‐231 breast cancer cells. Integrative analysis combined with expression microarray and RIP‐sequencing (RNA immunoprecipitation‐sequencing) showed that NONO post‐transcriptionally regulates the expression of cell proliferation‐related genes by binding to their mRNAs, as exemplified by S‐phase‐associated kinase 2 and E2F transcription factor 8. Overall, these results suggest that NONO is a key regulator for breast cancer proliferation through the pre‐mRNA splicing of cell proliferation‐related genes and could be a potential new diagnostic and therapeutic target for advanced disease.
Collapse
Affiliation(s)
- Kaori Iino
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Yuichi Mitobe
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Ken-Ichi Takayama
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hidetaka Kawabata
- Department of Breast and Endocrine Surgery, Toranomon Hospital, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan.,Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|