1
|
Du T, Liu S, Yu H, Hu T, Huang L, Gao L, Jia L, Hu J, Yu Y, Sun Q. Chronic sleep deprivation disturbs energy balance modulated by suprachiasmatic nucleus efferents in mice. BMC Biol 2024; 22:296. [PMID: 39710657 DOI: 10.1186/s12915-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Epidemiologic researches show that short sleep duration may affect feeding behaviors resulting in higher energy intake and increased risk of obesity, but the further mechanisms that can interpret the causality remain unclear. The circadian rhythm is fine-tuned by the suprachiasmatic nucleus (SCN) as the master clock, which is essential for driving rhythms in food intake and energy metabolism through neuronal projections to the arcuate nucleus (ARC) and paraventricular nucleus (PVN). RESULTS We showed that chronic SD-induced aberrant expressions of AgRP/NPY and POMC attributed to compromised JAK/STAT3 signals and reduced energy expenditure in the mice, which can be rescued with AAV-genetic overexpression of BMAL1 into SCN. The potential mechanism may be related to the disruptions of SCN efferent mediated by BMAL1. CONCLUSIONS Chronic SD impairs energy balance through directly dampening BMAL1 expression, probably in the transcription level, in the SCN, which in turn affects the neuron projections to ARC and PVN. Remarkably, we provide evidence that may explain the causal mechanisms associated with sleep curtailment and obesity in adolescents.
Collapse
Affiliation(s)
- Tingting Du
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Shuailing Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Honghong Yu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Tian Hu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lina Huang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lanyue Gao
- Experimental Center, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lihong Jia
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jiajin Hu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yang Yu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qi Sun
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
2
|
Vinciguerra D, Rajalakshmi P S, Yang J, Georgiou PG, Snell K, Pesenti T, Collins J, Tamboline M, Xu S, van Dam RM, Messina KMM, Hevener AL, Maynard HD. A Glucose-Responsive Glucagon-Micelle for the Prevention of Hypoglycemia. ACS CENTRAL SCIENCE 2024; 10:2036-2047. [PMID: 39634211 PMCID: PMC11613347 DOI: 10.1021/acscentsci.4c00937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 12/07/2024]
Abstract
While glucose-responsive insulin delivery systems are in widespread clinical use to treat insulin insufficiency, the on-demand supplementation of glucagon for acute hypoglycemia treatment remains understudied. A self-regulated glucagon release material is highly desired to mitigate the potential risks of severe insulin-induced hypoglycemia. Here, we describe a glucose-responsive polymeric nanosystem with glucagon covalently grafted to the end-group. Under normoglycemic conditions, phenylboronic acid units in the polymer chain reversibly bind glucose, triggering self-assembly of the conjugate into micelles. During hypoglycemia, however, the micelle disassembles into its original, unimeric state, revealing the active glucagon conjugate. The formulation showed a 5-fold increase in activity compared to native glucagon when tested in vitro. Glucagon-loaded micelles injected into mice prevented or reversed deep hypoglycemia when administered prior to or during an insulin challenge. Glucagon release was only observed at or below the counterregulatory threshold and not during normoglycemia or moderate hypoglycemia. The in vivo acute and chronic toxicity analysis, along with μPET/μCT imaging, established the biosafety profile of this formulation and demonstrated no organ accumulation. This proof-of-concept work is the first step toward development of a translational, stimuli-responsive glucagon delivery platform to control glycemia.
Collapse
Affiliation(s)
- Daniele Vinciguerra
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Rajalakshmi P S
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Jane Yang
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Panagiotis G. Georgiou
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Katherine Snell
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Théo Pesenti
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeffrey Collins
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
| | - Mikayla Tamboline
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
| | - Shili Xu
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
- Jonsson
Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United
States
| | - R. Michael van Dam
- Department
of Molecular and Medical Pharmacology and Crump Institute for Molecular
Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United States
- Jonsson
Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1735, United
States
| | - Kathryn M. M. Messina
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| | - Andrea L. Hevener
- Department
of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Dr, Los Angeles, California 90095, United States
- VA
Greater
Los Angeles Healthcare System GRECC, Los Angeles, California 90073, United States
| | - Heather D. Maynard
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1569, United States
| |
Collapse
|
3
|
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
|
4
|
Thorson JF, Prezotto LD. Malnutrition alters protein expression of KNDy neuropeptides in the arcuate nucleus of mature ewes. Front Physiol 2024; 15:1372944. [PMID: 38911326 PMCID: PMC11190783 DOI: 10.3389/fphys.2024.1372944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/15/2024] [Indexed: 06/25/2024] Open
Abstract
The neuropeptides kisspeptin, neurokinin B, and dynorphin A are imperative for the pulsatile secretion of gonadotropin-releasing hormone and luteinizing hormone to ultimately regulate reproductive cyclicity. A population of neurons co-expressing these neuropeptides, KNDy neurons, within the arcuate nucleus of the hypothalamus (ARC) are positioned to integrate energy status from afferent neuronal and glial cells. We hypothesized that KNDy-expressing neurons in the ARC of mature ewes are influenced by energy balance. To test this hypothesis, ovary-intact, mature ewes were fed to lose, maintain, or gain body weight and hypothalamic tissue harvested during the luteal phase of the estrous cycle. Fluorescent, multiplex immunohistochemistry with direct antibody conjugation was employed to identify and quantify neurons expressing a single neuropeptide, as well as for the first time report co-expression of kisspeptin, neurokinin B, and dynorphin A protein in the ARC. Previous reports using this population of ewes demonstrated that concentrations of insulin and leptin differed between ewes fed to achieve different body weights and that ewes fed to gain body weight had increased concentrations of progesterone. Moreover, within this population of ewes tanycyte density and cellular penetration into the ARC was increased in ewes fed to gain body weight. Within the current report we have revealed that the number of neurons in the ARC expressing kisspeptin, neurokinin B, and dynorphin A protein was increased in ewes fed to gain body weight. Moreover, the number of KNDy neurons in the ARC expressing all three neuropeptides within a single neuron was decreased in ewes fed to lose body weight and increased in ewes fed to gain body weight when compared to ewes fed to maintain body weight. The cumulative findings of this experimental model suggest that expression of kisspeptin, neurokinin B, and dynorphin A protein in the ARC during the luteal phase of the estrous cycle are influenced by energy balance-induced alterations in circulating concentrations of progesterone that drive changes in morphology and density of tanycytes to ultimately regulate central perception of global energy status. Moreover, these results demonstrate that changes in KNDy neurons within the ARC occur as an adaptation to energy balance, potentially regulated divergently by metabolic milieu via proopiomelanocortin afferents.
Collapse
Affiliation(s)
- Jennifer F. Thorson
- Nutrition, Growth and Physiology Research Unit, U.S. Meat Animal Research Center, Agricultural Research Service, United States Department of Agriculture, Clay Center, United States
| | - Ligia D. Prezotto
- Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, United States
| |
Collapse
|
5
|
Chen K, Wang Y, Li D, Wu R, Wang J, Wei W, Zhu W, Xie W, Feng D, He Y. Biological clock regulation by the PER gene family: a new perspective on tumor development. Front Cell Dev Biol 2024; 12:1332506. [PMID: 38813085 PMCID: PMC11133573 DOI: 10.3389/fcell.2024.1332506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
The Period (PER) gene family is one of the core components of the circadian clock, with substantial correlations between the PER genes and cancers identified in extensive researches. Abnormal mutations in PER genes can influence cell function, metabolic activity, immunity, and therapy responses, thereby promoting the initiation and development of cancers. This ultimately results in unequal cancers progression and prognosis in patients. This leads to variable cancer progression and prognosis among patients. In-depth studies on the interactions between the PER genes and cancers can reveal novel strategies for cancer detection and treatment. In this review, we aim to provide a comprehensive overview of the latest research on the role of the PER gene family in cancer.
Collapse
Affiliation(s)
- Kai Chen
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yaohui Wang
- Department of Urology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhu
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
| | - Wenhua Xie
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Yi He
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
| |
Collapse
|
6
|
Speksnijder EM, Bisschop PH, Siegelaar SE, Stenvers DJ, Kalsbeek A. Circadian desynchrony and glucose metabolism. J Pineal Res 2024; 76:e12956. [PMID: 38695262 DOI: 10.1111/jpi.12956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Esther M Speksnijder
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
8
|
Tiwari A, Rathor P, Trivedi PK, Ch R. Multi-Omics Reveal Interplay between Circadian Dysfunction and Type2 Diabetes. BIOLOGY 2023; 12:301. [PMID: 36829576 PMCID: PMC9953493 DOI: 10.3390/biology12020301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Type 2 diabetes is one of the leading threats to human health in the 21st century. It is a metabolic disorder characterized by a dysregulated glucose metabolism resulting from impaired insulin secretion or insulin resistance. More recently, accumulated epidemiological and animal model studies have confirmed that circadian dysfunction caused by shift work, late meal timing, and sleep loss leads to type 2 diabetes. Circadian rhythms, 24-h endogenous biological oscillations, are a fundamental feature of nearly all organisms and control many physiological and cellular functions. In mammals, light synchronizes brain clocks and feeding is a main stimulus that synchronizes the peripheral clocks in metabolic tissues, such as liver, pancreas, muscles, and adipose tissues. Circadian arrhythmia causes the loss of synchrony of the clocks of these metabolic tissues and leads to an impaired pancreas β-cell metabolism coupled with altered insulin secretion. In addition to these, gut microbes and circadian rhythms are intertwined via metabolic regulation. Omics approaches play a significant role in unraveling how a disrupted circadian metabolism causes type 2 diabetes. In the present review, we emphasize the discoveries of several genes, proteins, and metabolites that contribute to the emergence of type 2 diabetes mellitus (T2D). The implications of these discoveries for comprehending the circadian clock network in T2D may lead to new therapeutic solutions.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Priya Rathor
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Prabodh Kumar Trivedi
- Department of Biotechnology, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
| | - Ratnasekhar Ch
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
- School of Biological Sciences, Queen’s University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
9
|
Yang C, Sheng T, Hou W, Zhang J, Cheng L, Wang H, Liu W, Wang S, Yu X, Zhang Y, Yu J, Gu Z. Glucose-responsive microneedle patch for closed-loop dual-hormone delivery in mice and pigs. SCIENCE ADVANCES 2022; 8:eadd3197. [PMID: 36449622 PMCID: PMC9710866 DOI: 10.1126/sciadv.add3197] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/15/2022] [Indexed: 06/17/2023]
Abstract
Insulin and glucagon secreted from the pancreas with dynamic balance play a vital role in regulating blood glucose levels. Although distinct glucose-responsive insulin delivery systems have been developed, the lack of a self-regulated glucagon release module limits their clinical applications due to the potential risk of hypoglycemia. Here, we describe a transdermal polymeric microneedle patch for glucose-responsive closed-loop insulin and glucagon delivery to achieve glycemic regulation with minimized risk of hypoglycemia. The glucose-responsive phenylboronic acid units can bind to glucose to reversibly shift the net charge (from positive to negative) of the entire polymeric matrix within microneedles. Therefore, the release ratio of the negatively charged insulin and the positively charged glucagon analog from the patch can be dynamically tuned upon the fluctuation of blood glucose levels to realize glycemic homeostasis. In both chemically induced type 1 diabetic mouse and minipig models, this glucose-responsive dual-hormone microneedle patch demonstrated tight long-term regulation in blood glucose levels (>24 hours in minipigs).
Collapse
Affiliation(s)
- Changwei Yang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Sheng
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenhui Hou
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- College of Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Juan Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Cheng
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hao Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shiqi Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinmin Yu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuqi Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jicheng Yu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
10
|
Chan K, Wong FS, Pearson JA. Circadian rhythms and pancreas physiology: A review. Front Endocrinol (Lausanne) 2022; 13:920261. [PMID: 36034454 PMCID: PMC9399605 DOI: 10.3389/fendo.2022.920261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022] Open
Abstract
Type 2 diabetes mellitus, obesity and metabolic syndrome are becoming more prevalent worldwide and will present an increasingly challenging burden on healthcare systems. These interlinked metabolic abnormalities predispose affected individuals to a plethora of complications and comorbidities. Furthermore, diabetes is estimated by the World Health Organization to have caused 1.5 million deaths in 2019, with this figure projected to rise in coming years. This highlights the need for further research into the management of metabolic diseases and their complications. Studies on circadian rhythms, referring to physiological and behavioral changes which repeat approximately every 24 hours, may provide important insight into managing metabolic disease. Epidemiological studies show that populations who are at risk of circadian disruption such as night shift workers and regular long-haul flyers are also at an elevated risk of metabolic abnormalities such as insulin resistance and obesity. Aberrant expression of circadian genes appears to contribute to the dysregulation of metabolic functions such as insulin secretion, glucose homeostasis and energy expenditure. The potential clinical implications of these findings have been highlighted in animal studies and pilot studies in humans giving rise to the development of circadian interventions strategies including chronotherapy (time-specific therapy), time-restricted feeding, and circadian molecule stabilizers/analogues. Research into these areas will provide insights into the future of circadian medicine in metabolic diseases. In this review, we discuss the physiology of metabolism and the role of circadian timing in regulating these metabolic functions. Also, we review the clinical aspects of circadian physiology and the impact that ongoing and future research may have on the management of metabolic disease.
Collapse
Affiliation(s)
- Karl Chan
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - F. Susan Wong
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James Alexander Pearson
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
11
|
Uryash A, Mijares A, Lopez CE, Adams JA, Lopez JR. Chronic Elevation of Skeletal Muscle [Ca 2+] i Impairs Glucose Uptake. An in Vivo and in Vitro Study. Front Physiol 2022; 13:872624. [PMID: 35547584 PMCID: PMC9083325 DOI: 10.3389/fphys.2022.872624] [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: 02/09/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle is the primary site of insulin-mediated glucose uptake through the body and, therefore, an essential contributor to glucose homeostasis maintenance. We have recently provided evidence that chronic elevated intracellular Ca2+ concentration at rest [(Ca2+)i] compromises glucose homeostasis in malignant hyperthermia muscle cells. To further investigate how chronic elevated muscle [Ca2+]i modifies insulin-mediated glucose homeostasis, we measured [Ca2+]i and glucose uptake in vivo and in vitro in intact polarized muscle cells from glucose-intolerant RYR1-p.R163C and db/db mice. Glucose-intolerant RYR1-p.R163C and db/db mice have significantly elevated muscle [Ca2+]i and reduced muscle glucose uptake compared to WT muscle cells. Dantrolene treatment (1.5 mg/kg IP injection for 2 weeks) caused a significant reduction in fasting blood glucose levels and muscle [Ca2+]i and increased muscle glucose uptake compared to untreated RYR1-p.R163C and db/db mice. Furthermore, RYR1-p.R163C and db/db mice had abnormal basal insulin levels and response to glucose-stimulated insulin secretion. In vitro experiments conducted on single muscle fibers, dantrolene improved insulin-mediated glucose uptake in RYR1-p.R163C and db/db muscle fibers without affecting WT muscle fibers. In muscle cells with chronic elevated [Ca2+]i, GLUT4 expression was significantly lower, and the subcellular fraction (plasma membrane/cytoplasmic) was abnormal compared to WT. The results of this study suggest that i) Chronic elevated muscle [Ca2+]i decreases insulin-stimulated glucose uptake and consequently causes hyperglycemia; ii) Reduced muscle [Ca2+]i by dantrolene improves muscle glucose uptake and subsequent hyperglycemia; iii) The mechanism by which chronic high levels of [Ca2+]i interfere with insulin action appears to involve the expression of GLUT4 and its subcellular fractionation.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Alfredo Mijares
- Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - Carlos E Lopez
- Department of Physiotherapy, Wellmax Medical Center, Miami, FL, United States
| | - Jose A Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, United States
| |
Collapse
|
12
|
Fitzpatrick R, Davison G, Wilson JJ, McMahon G, McClean C. Exercise, type 1 diabetes mellitus and blood glucose: The implications of exercise timing. Front Endocrinol (Lausanne) 2022; 13:1021800. [PMID: 36246914 PMCID: PMC9555792 DOI: 10.3389/fendo.2022.1021800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
The scientific literature shows that exercise has many benefits for individuals with type 1 diabetes. Yet, several barriers to exercise in this population exist, such as post-exercise hypoglycaemia or hyperglycaemia. Several studies suggest that the timing of exercise may be an important factor in preventing exercise-induced hypoglycaemia or hyperglycaemia. However, there is a paucity of evidence solely focused on summarising findings regarding exercise timing and the impact it has on glucose metabolism in type 1 diabetes. This report suggests that resistance or high-intensity interval exercise/training (often known as HIIT) may be best commenced at the time of day when an individual is most likely to experience a hypoglycaemic event (i.e., afternoon/evening) due to the superior blood glucose stability resistance and HIIT exercise provides. Continuous aerobic-based exercise is advised to be performed in the morning due to circadian elevations in blood glucose at this time, thereby providing added protection against a hypoglycaemic episode. Ultimately, the evidence concerning exercise timing and glycaemic control remains at an embryonic stage. Carefully designed investigations of this nexus are required, which could be harnessed to determine the most effective, and possibly safest, time to exercise for those with type 1 diabetes.
Collapse
|
13
|
Worth C, Harper S, Salomon-Estebanez M, O'Shea E, Nutter PW, Dunne MJ, Banerjee I. Clustering of Hypoglycemia Events in Patients With Hyperinsulinism: Extension of the Digital Phenotype Through Retrospective Data Analysis. J Med Internet Res 2021; 23:e26957. [PMID: 34435596 PMCID: PMC8590184 DOI: 10.2196/26957] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Background Hyperinsulinism (HI) due to excess and dysregulated insulin secretion is the most common cause of severe and recurrent hypoglycemia in childhood. High cerebral glucose use in the early hours results in a high risk of hypoglycemia in people with diabetes and carries a significant risk of brain injury. Prevention of hypoglycemia is the cornerstone of the management of HI, but the risk of hypoglycemia at night or the timing of hypoglycemia in children with HI has not been studied; thus, the digital phenotype remains incomplete and management suboptimal. Objective This study aims to quantify the timing of hypoglycemia in patients with HI to describe glycemic variability and to extend the digital phenotype. This will facilitate future work using computational modeling to enable behavior change and reduce exposure of patients with HI to injurious hypoglycemic events. Methods Patients underwent continuous glucose monitoring (CGM) with a Dexcom G4 or G6 CGM device as part of their clinical assessment for either HI (N=23) or idiopathic ketotic hypoglycemia (IKH; N=24). The CGM data were analyzed for temporal trends. Hypoglycemia was defined as glucose levels <3.5 mmol/L. Results A total of 449 hypoglycemic events totaling 15,610 minutes were captured over 237 days from 47 patients (29 males; mean age 70 months, SD 53). The mean length of hypoglycemic events was 35 minutes. There was a clear tendency for hypoglycemia in the early hours (3-7 AM), particularly for patients with HI older than 10 months who experienced hypoglycemia 7.6% (1480/19,370 minutes) of time in this period compared with 2.6% (2405/92,840 minutes) of time outside this period (P<.001). This tendency was less pronounced in patients with HI who were younger than 10 months, patients with a negative genetic test result, and patients with IKH. Despite real-time CGM, there were 42 hypoglycemic events from 13 separate patients with HI lasting >30 minutes. Conclusions This is the first study to have taken the first step in extending the digital phenotype of HI by describing the glycemic trends and identifying the timing of hypoglycemia measured by CGM. We have identified the early hours as a time of high hypoglycemia risk for patients with HI and demonstrated that simple provision of CGM data to patients is not sufficient to eliminate hypoglycemia. Future work in HI should concentrate on the early hours as a period of high risk for hypoglycemia and must target personalized hypoglycemia predictions. Focus must move to the human-computer interaction as an aspect of the digital phenotype that is susceptible to change rather than simple mathematical modeling to produce small improvements in hypoglycemia prediction accuracy.
Collapse
Affiliation(s)
- Chris Worth
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, United Kingdom.,Department of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Simon Harper
- Department of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Maria Salomon-Estebanez
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Elaine O'Shea
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Paul W Nutter
- Department of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Mark J Dunne
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Indraneel Banerjee
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Ding G, Li X, Hou X, Zhou W, Gong Y, Liu F, He Y, Song J, Wang J, Basil P, Li W, Qian S, Saha P, Wang J, Cui C, Yang T, Zou K, Han Y, Amos CI, Xu Y, Chen L, Sun Z. REV-ERB in GABAergic neurons controls diurnal hepatic insulin sensitivity. Nature 2021; 592:763-767. [PMID: 33762728 PMCID: PMC8085086 DOI: 10.1038/s41586-021-03358-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Systemic insulin sensitivity shows a diurnal rhythm with a peak upon waking1,2. The molecular mechanism that underlies this temporal pattern is unclear. Here we show that the nuclear receptors REV-ERB-α and REV-ERB-β (referred to here as 'REV-ERB') in the GABAergic (γ-aminobutyric acid-producing) neurons in the suprachiasmatic nucleus (SCN) (SCNGABA neurons) control the diurnal rhythm of insulin-mediated suppression of hepatic glucose production in mice, without affecting diurnal eating or locomotor behaviours during regular light-dark cycles. REV-ERB regulates the rhythmic expression of genes that are involved in neurotransmission in the SCN, and modulates the oscillatory firing activity of SCNGABA neurons. Chemogenetic stimulation of SCNGABA neurons at waking leads to glucose intolerance, whereas restoration of the temporal pattern of either SCNGABA neuron firing or REV-ERB expression rescues the time-dependent glucose metabolic phenotype caused by REV-ERB depletion. In individuals with diabetes, an increased level of blood glucose after waking is a defining feature of the 'extended dawn phenomenon'3,4. Patients with type 2 diabetes with the extended dawn phenomenon exhibit a differential temporal pattern of expression of REV-ERB genes compared to patients with type 2 diabetes who do not have the extended dawn phenomenon. These findings provide mechanistic insights into how the central circadian clock regulates the diurnal rhythm of hepatic insulin sensitivity, with implications for our understanding of the extended dawn phenomenon in type 2 diabetes.
Collapse
Affiliation(s)
- Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Xin Li
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjun Zhou
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Yingyun Gong
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fuqiang Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Laboratory of Brain Glycemia and Metabolism Control, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Jia Song
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Paul Basil
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Wenbo Li
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Sichong Qian
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Pradip Saha
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jinbang Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Cui
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Yang
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kexin Zou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Younghun Han
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I Amos
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China.
| | - Zheng Sun
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
15
|
Prevention of Diabetes and Cardiovascular Disease in Obesity. Int J Mol Sci 2020; 21:ijms21218178. [PMID: 33142938 PMCID: PMC7663329 DOI: 10.3390/ijms21218178] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is one of the major risk factors for the development of both impaired glucose tolerance (IGT, or prediabetes) and type 2 diabetes (T2D), and its prevalence worldwide drives toward an increased rate of cardiovascular morbidity and mortality. Given the estimations of the World Health Organization (WHO) and the recommendation of the Diabetes Prevention Program (DPP), where IGT and diabetes are considered as risk factors for the development of cardiovascular complications and obesity, the development of diabetes should be treated because of its potential reversibility. In this view, several interventions such as diet, lifestyle changes, and pharmacological treatment are effective, including bariatric metabolic surgery (BMS), which is the most incisive way to efficiently lower body weight. In this review, we sought to summarize some of the major aspects linked to diabetes prevention in overweight/obesity, focusing on the use of surgery; we also attempted to elucidate molecular pathways involved in a variety of obesity-induced processes able to favor the progression of chronic diseases, such as diabetes and its complications.
Collapse
|
16
|
Masís-Vargas A, Hicks D, Kalsbeek A, Mendoza J. Blue light at night acutely impairs glucose tolerance and increases sugar intake in the diurnal rodent Arvicanthis ansorgei in a sex-dependent manner. Physiol Rep 2020; 7:e14257. [PMID: 31646762 PMCID: PMC6811685 DOI: 10.14814/phy2.14257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
In our modern society, the exposure to light at night (LAN) has increased considerably, which may impact human health negatively. Especially exposure to light at night containing short wavelength emissions (~450–500 nm) can disrupt the normal function of the biological clock, altering sleep‐wake cycles and inducing metabolic changes. Recently, we reported that light at night acutely impairs glucose tolerance in nocturnal rats. However, light at night in nocturnal rodents coincides with their activity period, in contrast to artificial light at night exposure in humans. The aim of this study was to evaluate the acute effects of blue (λ = 490 ± 20 nm) artificial light at night (bALAN) on glucose metabolism and food intake in both male and female diurnal Sudanian grass rats (Arvicanthis ansorgei) fed either regular chow or a free choice high‐fat high sucrose diet (HFHS). In both chow and HFHS fed male Arvicanthis, 1‐hour of bALAN exposure induced a higher glucose response in the oral glucose tolerance test (OGTT) accompanied by a significant decrease in plasma insulin. Furthermore, in HFHS fed animals, bALAN induced an increase in sucrose intake during the dark phase in males but not in females. Additionally, 1‐h of bALAN increased the nonfasted glucose levels together with plasma corticosterone in female grass rats. These results provide new and further evidence for the deleterious effects of exposure to short wavelength emission‐containing artificial light at night on glucose metabolism in a diurnal rodent in a sex‐dependent manner.
Collapse
Affiliation(s)
- Anayanci Masís-Vargas
- Institute of Cellular and Integrative Neurosciences (INCI), UPR-3212 CNRS, University of Strasbourg, Strasbourg, France.,Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - David Hicks
- Institute of Cellular and Integrative Neurosciences (INCI), UPR-3212 CNRS, University of Strasbourg, Strasbourg, France
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences (INCI), UPR-3212 CNRS, University of Strasbourg, Strasbourg, France
| |
Collapse
|
17
|
Effect of Type 2 Diabetes Mellitus on the Hypoxia-Inducible Factor 1-Alpha Expression. Is There a Relationship with the Clock Genes? J Clin Med 2020; 9:jcm9082632. [PMID: 32823749 PMCID: PMC7465909 DOI: 10.3390/jcm9082632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 01/21/2023] Open
Abstract
Limited reports exist on the relationships between regulation of oxygen homeostasis and circadian clock genes in type 2 diabetes. We examined whether the expression of Hypoxia-Inducible Factor-1α (HIF-1α) and HIF-2α relates to changes in the expression of clock genes (Period homolog proteins (PER)1, PER2, PER3, Retinoid-related orphan receptor alpha (RORA), Aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL), Circadian locomotor output cycles kaput (CLOCK), and Cryptochrome proteins (CRY) 1 and CRY2) in patients with type 2 diabetes. A total of 129 subjects were evaluated in this cross-sectional study (48% with diabetes). The gene expression was measured by polymerase chain reaction. The lactate and pyruvate levels were used as surrogate of the hypoxia induced anaerobic glycolysis activity. Patients with diabetes showed an increased plasma concentration of both lactate (2102.1 ± 688.2 vs. 1730.4 ± 694.4 uM/L, p = 0.013) and pyruvate (61.9 ± 25.6 vs. 50.3 ± 23.1 uM/L, p = 0.026) in comparison to controls. However, this finding was accompanied by a blunted HIF-1α expression (1.1 (0.2 to 5.0) vs. 1.7 (0.4 to 9.2) arbitrary units (AU), p ≤ 0.001). Patients with diabetes also showed a significant reduction of all assessed clock genes’ expression. Univariate analysis showed that HIF-1α and almost all clock genes were significantly and negatively correlated with HbA1c concentration. In addition, positive correlations between HIF-1α and the clock genes were observed. The stepwise multivariate regression analysis showed that HbA1c and clock genes independently predicted the expression of HIF-1α. Type 2 diabetes modifies the expression of HIF-1α and clock genes, which correlates with the degree of metabolic control.
Collapse
|
18
|
Hogenboom R, Kalsbeek MJ, Korpel NL, de Goede P, Koenen M, Buijs RM, Romijn JA, Swaab DF, Kalsbeek A, Yi CX. Loss of arginine vasopressin- and vasoactive intestinal polypeptide-containing neurons and glial cells in the suprachiasmatic nucleus of individuals with type 2 diabetes. Diabetologia 2019; 62:2088-2093. [PMID: 31327049 PMCID: PMC6805798 DOI: 10.1007/s00125-019-4953-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS The central pacemaker of the mammalian biological timing system is located within the suprachiasmatic nucleus (SCN) in the anterior hypothalamus. Together with the peripheral clocks, this central brain clock ensures a timely, up-to-date and proper behaviour for an individual throughout the day-night cycle. A mismatch between the central and peripheral clocks results in a disturbance of daily rhythms in physiology and behaviour. It is known that the number of rhythmically expressed genes is reduced in peripheral tissue of individuals with type 2 diabetes mellitus. However, it is not known whether the central SCN clock is also affected in the pathogenesis of type 2 diabetes. In the current study, we compared the profiles of the SCN neurons and glial cells between type 2 diabetic and control individuals. METHODS We collected post-mortem hypothalamic tissues from 28 type 2 diabetic individuals and 12 non-diabetic control individuals. We performed immunohistochemical analysis for three SCN neuropeptides, arginine vasopressin (AVP), vasoactive intestinal polypeptide (VIP) and neurotensin (NT), and for two proteins expressed in glial cells, ionised calcium-binding adapter molecule 1 (IBA1, a marker of microglia) and glial fibrillary acidic protein (GFAP, a marker of astroglial cells). RESULTS The numbers of AVP immunoreactive (AVP-ir) and VIP-ir neurons and GFAP-ir astroglial cells in the SCN of type 2 diabetic individuals were significantly decreased compared with the numbers in the SCN of the control individuals. In addition, the relative intensity of AVP immunoreactivity was reduced in the individuals with type 2 diabetes. The number of NT-ir neurons and IBA1-ir microglial cells in the SCN was similar in the two groups. CONCLUSIONS/INTERPRETATION Our data show that type 2 diabetes differentially affects the numbers of AVP- and VIP-expressing neurons and GFAP-ir astroglial cells in the SCN, each of which could affect the daily rhythmicity of the SCN biological clock machinery. Therefore, for effectively treating type 2 diabetes, lifestyle changes and/or medication to normalise central biological clock functioning might be helpful.
Collapse
Affiliation(s)
- Rick Hogenboom
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Martin J Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Nikita L Korpel
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Paul de Goede
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Marit Koenen
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Ruud M Buijs
- Department of Cell Biology and Physiology, Institute for Biomedical Research, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Johannes A Romijn
- Department of Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Dick F Swaab
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Location AMC, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands.
- Laboratory of Endocrinology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands.
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
19
|
Pastore N, Vainshtein A, Herz NJ, Huynh T, Brunetti L, Klisch TJ, Mutarelli M, Annunziata P, Kinouchi K, Brunetti-Pierri N, Sassone-Corsi P, Ballabio A. Nutrient-sensitive transcription factors TFEB and TFE3 couple autophagy and metabolism to the peripheral clock. EMBO J 2019; 38:embj.2018101347. [PMID: 31126958 DOI: 10.15252/embj.2018101347] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/03/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
Autophagy and energy metabolism are known to follow a circadian pattern. However, it is unclear whether autophagy and the circadian clock are coordinated by common control mechanisms. Here, we show that the oscillation of autophagy genes is dependent on the nutrient-sensitive activation of TFEB and TFE3, key regulators of autophagy, lysosomal biogenesis, and cell homeostasis. TFEB and TFE3 display a circadian activation over the 24-h cycle and are responsible for the rhythmic induction of genes involved in autophagy during the light phase. Genetic ablation of TFEB and TFE3 in mice results in deregulated autophagy over the diurnal cycle and altered gene expression causing abnormal circadian wheel-running behavior. In addition, TFEB and TFE3 directly regulate the expression of Rev-erbα (Nr1d1), a transcriptional repressor component of the core clock machinery also involved in the regulation of whole-body metabolism and autophagy. Comparative analysis of the cistromes of TFEB/TFE3 and REV-ERBα showed an extensive overlap of their binding sites, particularly in genes involved in autophagy and metabolic functions. These data reveal a direct link between nutrient and clock-dependent regulation of gene expression shedding a new light on the crosstalk between autophagy, metabolism, and circadian cycles.
Collapse
Affiliation(s)
- Nunzia Pastore
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Anna Vainshtein
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Niculin J Herz
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Tuong Huynh
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lorenzo Brunetti
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Tiemo J Klisch
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Kenichiro Kinouchi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, U1233 INSERM, School of Medicine, University of California Irvine (UCI), Irvine, CA, USA
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Medical and Translational Sciences, Medical Genetics, Federico II University, Naples, Italy
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, U1233 INSERM, School of Medicine, University of California Irvine (UCI), Irvine, CA, USA
| | - Andrea Ballabio
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Medical and Translational Sciences, Medical Genetics, Federico II University, Naples, Italy
| |
Collapse
|
20
|
Zhang S, Bories G, Lantz C, Emmons R, Becker A, Liu E, Abecassis MM, Yvan-Charvet L, Thorp EB. Immunometabolism of Phagocytes and Relationships to Cardiac Repair. Front Cardiovasc Med 2019; 6:42. [PMID: 31032261 PMCID: PMC6470271 DOI: 10.3389/fcvm.2019.00042] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide. Myocardial ischemia is a major contributor to cardiovascular morbidity and mortality. In the case of acute myocardial infarction, subsequent cardiac repair relies upon the acute, and coordinated response to injury by innate myeloid phagocytes. This includes neutrophils, monocytes, macrophage subsets, and immature dendritic cells. Phagocytes function to remove necrotic cardiomyocytes, apoptotic inflammatory cells, and to remodel extracellular matrix. These innate immune cells also secrete cytokines and growth factors that promote tissue replacement through fibrosis and angiogenesis. Within the injured myocardium, macrophages polarize from pro-inflammatory to inflammation-resolving phenotypes. At the core of this functional plasticity is cellular metabolism, which has gained an appreciation for its integration with phagocyte function and remodeling of the transcriptional and epigenetic landscape. Immunometabolic rewiring is particularly relevant after ischemia and clinical reperfusion given the rapidly changing oxygen and metabolic milieu. Hypoxia reduces mitochondrial oxidative phosphorylation and leads to increased reliance on glycolysis, which can support biosynthesis of pro-inflammatory cytokines. Reoxygenation is permissive for shifts back to mitochondrial metabolism and fatty acid oxidation and this is ultimately linked to pro-reparative macrophage polarization. Improved understanding of mechanisms that regulate metabolic adaptations holds the potential to identify new metabolite targets and strategies to reduce cardiac damage through nutrient signaling.
Collapse
Affiliation(s)
- Shuang Zhang
- Departments of Pathology and Pediatrics, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gael Bories
- UMR INSERM U1065/UNS, C3M, Bâtiment Universitaire ARCHIMED, Nice, France
| | - Connor Lantz
- Departments of Pathology and Pediatrics, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Russel Emmons
- Departments of Pathology and Pediatrics, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Amanda Becker
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL, United States
| | - Esther Liu
- Departments of Pathology and Pediatrics, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Michael M. Abecassis
- Comprehensive Transplant Center, Northwestern Feinberg School of Medicine, Chicago, IL, United States
| | | | - Edward B. Thorp
- Departments of Pathology and Pediatrics, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
21
|
Abstract
The epidemic of Type 2 diabetes mellitus necessitates development of novel therapeutic and preventative strategies to attenuate expansion of this debilitating disease. Evidence links the circadian system to various aspects of diabetes pathophysiology and treatment. The aim of this review will be to outline the rationale for therapeutic targeting of the circadian system in the treatment and prevention of Type 2 diabetes mellitus and consequent metabolic comorbidities.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic School of Medicine, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
22
|
Zhou L, Xiao X, Zhang Q, Zheng J, Li M, Yu M, Wang X, Deng M, Zhai X, Li R, Liu J. Dietary Genistein Could Modulate Hypothalamic Circadian Entrainment, Reduce Body Weight, and Improve Glucose and Lipid Metabolism in Female Mice. Int J Endocrinol 2019; 2019:2163838. [PMID: 31139215 PMCID: PMC6500629 DOI: 10.1155/2019/2163838] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 12/13/2022] Open
Abstract
Genistein has beneficial effects on metabolic disorders. However, the specific mechanism is not clearly understood. In light of the significant role of the hypothalamus in energy and metabolic homeostasis, this study was designed to explore whether dietary genistein intake could mitigate the harmful effects of a high-fat diet on glucose and lipid metabolism and whether any alterations caused by dietary genistein were associated with hypothalamic gene expression profiles. C57BL/6 female mice were fed a high-fat diet without genistein (HF), a high-fat diet with genistein (HFG), or a normal control diet (CON) for 8 weeks. Body weight and energy intake were assessed. At the end of the study, glucose tolerance and serum levels of insulin and lipids were analyzed. Hypothalamic tissue was collected for whole transcriptome sequencing and reverse transcription quantitative PCR (RT-qPCR) validation. Energy intake and body weight were significantly reduced in the mice of the HFG group compared with those of the HF group. Mice fed the HFG diet had improved glucose tolerance and decreased serum triacylglycerol, free fatty acids, and low-density lipoprotein cholesterol compared with those fed the HF diet. The HFG diet also modulated gene expression in the hypothalamus; the most abundant genes were enriched in the circadian entrainment pathway. Dietary genistein intake could reduce body weight, improve glucose and lipid metabolism, and regulate hypothalamic circadian entrainment. The ability of genistein intake to influence regulation of the hypothalamic circadian rhythm is important since this could provide a novel target for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Liyuan Zhou
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia Zheng
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Li
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Miao Yu
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Wang
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingqun Deng
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Zhai
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rongrong Li
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jieying Liu
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Du S, Shi MJ, Sun ZZ, Li W. Clinical diagnosis for dusk phenomenon of diabetes. Medicine (Baltimore) 2018; 97:e11873. [PMID: 30142783 PMCID: PMC6112941 DOI: 10.1097/md.0000000000011873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/25/2018] [Indexed: 11/25/2022] Open
Abstract
The diabetes dusk phenomenon (spontaneous and transient pre-dinner hyperglycemia) anecdotally exists but has not been investigated.A total of 80 diabetic patients that received continuous subcutaneous insulin infusions were retrospectively studied. They were grouped into a routine group (R) (consecutive δDG [dusk blood glucose difference] <0 mmol/L) and a classic dusk phenomenon group (CDP, consecutive δDG≥0 mmol/L). δDG represents differences in blood glucose measurements between pre-dinner and post-lunch (δDG: dusk blood glucose difference). Other patients were placed in a suspicious group (S). The suspicious group was further divided into 3 groups based on the frequency at which the δDG occurred: suspicious 1 group (S1), δDG≥0 mmol/L occurred once only; suspicious 3 group (S3), δDG < 0 mmol/L occurred once only, and the remaining patients were grouped in the suspicious 2 group (S2).We identified the CDP and S3 groups as the "clinical dusk phenomenon" group (CLDP). We confirmed that the S1 and R groups to be in the "clinical routine" group. The S2 group was significantly different from the CDP group. In addition, the S2 group had significant differences in δDG measurements and post-lunch blood glucose values compared with the R group, but no differences in other parameters were seen. Multiple comparisons with the other suspicious groups also showed no statistical difference in many parameters. Thus, we placed these patients into the "suspicious clinical dusk phenomenon" group (SDP). The δDG cut-off for the CLDP group was 1.0167 mmol/L. The pre-dinner-pre-lunch blood glucose cut-off for this group was 2.72 mmol/L. The δDG cut-off for the SDP group was -0.95 mmol/L. The pre-dinner-pre-lunch blood glucose cut-off for this group was 0.87 mmol/L. The cut-off points for the post-dinner-post-lunch blood glucose measurements in the CLDP and SDP groups were both 1.2667 mmol/L.A consecutive δDG≥0 or a once only δDG < 0 could be diagnosed as falling into the CLDP group. The CLDP could be excluded when a consecutive δDG < 0 or a once only δDG≥0 was found. Patients falling into other categories were placed into the SDP group.
Collapse
Affiliation(s)
- Sina Du
- Department of Endocrinology, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi
| | - Min-jia Shi
- Department of Endocrinology, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi
| | - Zhan-zhan Sun
- Department of Endocrinology, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi
| | - Wei Li
- Department of Endocrinology, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
24
|
Dyar KA, Hubert MJ, Mir AA, Ciciliot S, Lutter D, Greulich F, Quagliarini F, Kleinert M, Fischer K, Eichmann TO, Wright LE, Peña Paz MI, Casarin A, Pertegato V, Romanello V, Albiero M, Mazzucco S, Rizzuto R, Salviati L, Biolo G, Blaauw B, Schiaffino S, Uhlenhaut NH. Transcriptional programming of lipid and amino acid metabolism by the skeletal muscle circadian clock. PLoS Biol 2018; 16:e2005886. [PMID: 30096135 PMCID: PMC6105032 DOI: 10.1371/journal.pbio.2005886] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/22/2018] [Accepted: 07/27/2018] [Indexed: 12/30/2022] Open
Abstract
Circadian clocks are fundamental physiological regulators of energy homeostasis, but direct transcriptional targets of the muscle clock machinery are unknown. To understand how the muscle clock directs rhythmic metabolism, we determined genome-wide binding of the master clock regulators brain and muscle ARNT-like protein 1 (BMAL1) and REV-ERBα in murine muscles. Integrating occupancy with 24-hr gene expression and metabolomics after muscle-specific loss of BMAL1 and REV-ERBα, here we unravel novel molecular mechanisms connecting muscle clock function to daily cycles of lipid and protein metabolism. Validating BMAL1 and REV-ERBα targets using luciferase assays and in vivo rescue, we demonstrate how a major role of the muscle clock is to promote diurnal cycles of neutral lipid storage while coordinately inhibiting lipid and protein catabolism prior to awakening. This occurs by BMAL1-dependent activation of Dgat2 and REV-ERBα-dependent repression of major targets involved in lipid metabolism and protein turnover (MuRF-1, Atrogin-1). Accordingly, muscle-specific loss of BMAL1 is associated with metabolic inefficiency, impaired muscle triglyceride biosynthesis, and accumulation of bioactive lipids and amino acids. Taken together, our data provide a comprehensive overview of how genomic binding of BMAL1 and REV-ERBα is related to temporal changes in gene expression and metabolite fluctuations.
Collapse
Affiliation(s)
- Kenneth Allen Dyar
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Michaël Jean Hubert
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | - Ashfaq Ali Mir
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | | | - Dominik Lutter
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | - Franziska Greulich
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | - Fabiana Quagliarini
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | - Maximilian Kleinert
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | - Katrin Fischer
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
| | | | | | | | - Alberto Casarin
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Vanessa Pertegato
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | | | - Mattia Albiero
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sara Mazzucco
- Clinica Medica, Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Leonardo Salviati
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy
| | - Gianni Biolo
- Clinica Medica, Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - N. Henriette Uhlenhaut
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for Diabetes and Obesity (IDO), Munich, Germany
- Gene Center, Ludwig-Maximilians-Universitaet (LMU), Munich, Germany
| |
Collapse
|
25
|
Tsuneki H, Wada T, Sasaoka T. Chronopathophysiological implications of orexin in sleep disturbances and lifestyle-related disorders. Pharmacol Ther 2018; 186:25-44. [PMID: 29289556 DOI: 10.1016/j.pharmthera.2017.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sleep, a mysterious behavior, has recently been recognized as a crucial factor for health and longevity. The daily sleep/wake cycle provides the basis of biorhythms controlling whole-body homeostasis and homeodynamics; therefore, disruption of sleep causes several physical and psychological disorders, including cardiovascular disease, obesity, diabetes, cancer, anxiety, depression, and cognitive dysfunction. However, the mechanism linking sleep disturbances and sleep-related disorders remains unknown. Orexin (also known as hypocretin) is a neuropeptide produced in the hypothalamus. Central levels of orexin oscillate with the daily rhythm and peak at the awake phase. Orexin plays a major role in stabilizing the wakefulness state. Orexin deficiency causes sleep/wake-state instability, resulting in narcolepsy. Hyper-activation of the orexin system also causes sleep disturbances, such as insomnia, and hence, suvorexant, an orexin receptor antagonist, has been clinically used to treat insomnia. Importantly, central actions of orexin regulate motivated behaviors, stress response, and energy/glucose metabolism by coordinating the central-autonomic nervous systems and endocrine systems. These multiple actions of orexin maintain survival. However, it remains unknown whether chronopharmacological interventions targeting the orexin system ameliorate sleep-related disorders as well as sleep in humans. To understand the significance of adequate orexin action for prevention of these disorders, this review summarizes the physiological functions of daily orexin action and pathological implications of its mistimed or reduced action in sleep disturbances and sleep-related disorders (lifestyle-related physical and neurological disorders in particular). Timed administration of drugs targeting the orexin system may prevent lifestyle-related diseases by improving the quality of life in patients with sleep disturbances.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
26
|
ter Horst KW, Lammers NM, Trinko R, Opland DM, Figee M, Ackermans MT, Booij J, van den Munckhof P, Schuurman PR, Fliers E, Denys D, DiLeone RJ, la Fleur SE, Serlie MJ. Striatal dopamine regulates systemic glucose metabolism in humans and mice. Sci Transl Med 2018; 10:10/442/eaar3752. [DOI: 10.1126/scitranslmed.aar3752] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
|
27
|
Effect of developmental NMDAR antagonism with CGP 39551 on aspartame-induced hypothalamic and adrenal gene expression. PLoS One 2018; 13:e0194416. [PMID: 29561882 PMCID: PMC5862471 DOI: 10.1371/journal.pone.0194416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/04/2018] [Indexed: 01/16/2023] Open
Abstract
Rationale Aspartame (L-aspartyl phenylalanine methyl ester) is a non-nutritive sweetener (NNS) approved for use in more than 6000 dietary products and pharmaceuticals consumed by the general public including adults and children, pregnant and nursing mothers. However a recent prospective study reported a doubling of the risk of being overweight amongst 1-year old children whose mothers consumed NNS-sweetened beverages daily during pregnancy. We have previously shown that chronic aspartame (ASP) exposure commencing in utero may detrimentally affect adulthood adiposity status, glucose metabolism and aspects of behavior and spatial cognition, and that this can be modulated by developmental N-methyl-D-aspartate receptor (NMDAR) blockade with the competitive antagonist CGP 39551 (CGP). Since glucose homeostasis and certain aspects of behavior and locomotion are regulated in part by the NMDAR-rich hypothalamus, which is part of the hypothalamic-pituitary-adrenal- (HPA) axis, we have elected to examine changes in hypothalamic and adrenal gene expression in response to ASP exposure in the presence or absence of developmental NMDAR antagonism with CGP, using Affymetrix microarray analysis. Results Using 2-factor ANOVA we identified 189 ASP-responsive differentially expressed genes (DEGs) in the adult male hypothalamus and 2188 in the adrenals, and a further 23 hypothalamic and 232 adrenal genes significantly regulated by developmental treatment with CGP alone. ASP exposure robustly elevated the expression of a network of genes involved in hypothalamic neurosteroidogenesis, together with cell stress and inflammatory genes, consistent with previous reports of aspartame-induced CNS stress and oxidative damage. These genes were not differentially expressed in ASP mice with CGP antagonism. In the adrenal glands of ASP-exposed mice, GABA and Glutamate receptor subunit genes were amongst those most highly upregulated. Developmental NMDAR antagonism alone had less effect on adulthood gene expression and affected mainly hypothalamic neurogenesis and adrenal steroid metabolism. Combined ASP + CGP treatment mainly upregulated genes involved in adrenal drug and cholesterol metabolism. Conclusion ASP exposure increased the expression of functional networks of genes involved in hypothalamic neurosteroidogenesis and adrenal catecholamine synthesis, patterns of expression which were not present in ASP-exposed mice with developmental NMDAR antagonism.
Collapse
|
28
|
Oishi K, Ohyama S, Higo-Yamamoto S. Chronic sleep disorder induced by psychophysiological stress induces glucose intolerance without adipose inflammation in mice. Biochem Biophys Res Commun 2018; 495:2616-2621. [PMID: 29288667 DOI: 10.1016/j.bbrc.2017.12.158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 12/26/2017] [Indexed: 01/11/2023]
Abstract
Sleep disturbances are associated with various metabolic diseases such as hypertension and diabetes. We had previously established a mouse model of a psychophysiological stress-induced chronic sleep disorder (CSD) characterized by disrupted circadian rhythms of wheel-running activity, core body temperature, and sleep-wake cycles. To evaluate the underlying mechanisms of metabolic disorders induced by CSD, we created mice with CSD for six weeks and fed them with a high-fat diet. Glucose intolerance with hyperglycemia resulted, although plasma insulin levels and body weight increases were identical between control and CSD mice. Gluconeogenesis and glycolysis were enhanced and suppressed, respectively, in the livers of CSD mice, because the mRNA expression of Pck1 was significantly increased, whereas that of Gck and Pklr were significantly decreased in the CSD mice. Adipose inflammation induced by the high-fat diet seemed suppressed by the CSD, because the mRNA expression levels of Adgre1, Ccl2, and Tnf were significantly downregulated in the adipose tissues of CSD mice. These findings suggest that CSD impair glucose tolerance by inducing gluconeogenesis and suppressing glycolysis. Hyperphasia with hypoleptinemia, hypercorticosteronemia, and increased plasma free fatty acids might be involved in the impaired glucose metabolism under a CSD. Further studies are needed to elucidate the endocrine and molecular mechanisms underlying the associations between sleep disorders and impaired glucose homeostasis that consequently causes diabetes.
Collapse
Affiliation(s)
- Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan.
| | - Sumika Ohyama
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Sayaka Higo-Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| |
Collapse
|
29
|
Alfaro FJ, Gavrieli A, Saade-Lemus P, Lioutas VA, Upadhyay J, Novak V. White matter microstructure and cognitive decline in metabolic syndrome: a review of diffusion tensor imaging. Metabolism 2018; 78:52-68. [PMID: 28920863 PMCID: PMC5732847 DOI: 10.1016/j.metabol.2017.08.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022]
Abstract
Metabolic syndrome is a cluster of cardiovascular risk factors defined by the presence of abdominal obesity, glucose intolerance, hypertension and/or dyslipidemia. It is a major public health epidemic worldwide, and a known risk factor for the development of cognitive dysfunction and dementia. Several studies have demonstrated a positive association between the presence of metabolic syndrome and worse cognitive outcomes, however, evidence of brain structure pathology is limited. Diffusion tensor imaging has offered new opportunities to detect microstructural white matter changes in metabolic syndrome, and a possibility to detect associations between functional and structural abnormalities. This review analyzes the impact of metabolic syndrome on white matter microstructural integrity, brain structure abnormalities and their relationship to cognitive function. Each of the metabolic syndrome components exerts a specific signature of white matter microstructural abnormalities. Metabolic syndrome and its components exert both additive/synergistic, as well as, independent effects on brain microstructure thus accelerating brain aging and cognitive decline.
Collapse
Affiliation(s)
- Freddy J Alfaro
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Road, Palmer 127, Boston, MA 02215, USA.
| | - Anna Gavrieli
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Road, Palmer 127, Boston, MA 02215, USA.
| | - Patricia Saade-Lemus
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Road, Palmer 127, Boston, MA 02215, USA.
| | - Vasileios-Arsenios Lioutas
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Road, Palmer 127, Boston, MA 02215, USA.
| | - Jagriti Upadhyay
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215,USA.
| | - Vera Novak
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 185 Pilgrim Road, Palmer 127, Boston, MA 02215, USA.
| |
Collapse
|
30
|
Zarifkar M, Noshad S, Shahriari M, Afarideh M, Khajeh E, Karimi Z, Ghajar A, Esteghamati A. Inverse Association of Peripheral Orexin-A with Insulin Resistance in Type 2 Diabetes Mellitus: A Randomized Clinical Trial. Rev Diabet Stud 2017; 14:301-310. [PMID: 29145540 DOI: 10.1900/rds.2017.14.301] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AIMS To investigate the association between serum orexin concentrations and insulin resistance/sensitivity in a sample of patients with type 2 diabetes mellitus, and to study the effects of anti-hyperglycemic treatment on orexin concentrations over three months. METHODS This study was designed as a randomized, open-label, clinical trial. Before allocation, sixty medication-naïve, newly-diagnosed, type 2 diabetes patients underwent a 75 g oral glucose tolerance test (OGTT). Afterwards, using a randomized trial design (IRCT201102275917N1) patients were allocated to either the metformin (1000 mg daily) or pioglitazone (30 mg daily) arm, and were reexamined after three months. Serum insulin, plasma glucose, and orexin concentrations were measured at baseline, during OGTT, and after three months. RESULTS Orexin concentrations significantly decreased after OGTT (0 vs. 120 min: 0.63 ± 0.07 vs. 0.31 ± 0.03 ng/ml, p < 0.001). Insulin resistance determined by homeostasis model assessment of insulin resistance (HOMA-IR) was significantly and negatively correlated with orexin (r = -0.301, p = 0.024). Furthermore, orexin concentrations were significantly and positively correlated with the insulin sensitivity index derived from OGTT (r = 0.326, p = 0.014). Three-month treatment with metformin and pioglitazone significantly improved insulin sensitivity and increased orexin concentrations by 26% (p = 0.025) and 14% (p = 0.076), respectively. Between-group analysis showed that changes in orexin concentrations with metformin and pioglitazone were not significantly different (p = 0.742). CONCLUSIONS There was a negative association between peripheral orexin concentrations and insulin resistance in type 2 diabetes patients. Three-month anti-hyperglycemic treatment with proportionate doses of metformin or pioglitazone increased orexin concentrations via amelioration of insulin resistance and improvement of glycemic control.
Collapse
Affiliation(s)
- Mitra Zarifkar
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Noshad
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Shahriari
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Afarideh
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elias Khajeh
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Karimi
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Ghajar
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Esteghamati
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
The sweet tooth of the circadian clock. Biochem Soc Trans 2017; 45:871-884. [PMID: 28673939 DOI: 10.1042/bst20160183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/04/2017] [Accepted: 05/16/2017] [Indexed: 12/19/2022]
Abstract
The endogenous circadian clock is a key regulator of daily metabolic processes. On the other hand, circadian clocks in a broad range of tissues can be tuned by extrinsic and intrinsic metabolic cues. The bidirectional interaction between circadian clocks and metabolism involves both transcriptional and post-translational mechanisms. Nuclear receptors exemplify the transcriptional programs that couple molecular clocks to metabolism. The post-translational modifications of the core clock machinery are known to play a key role in metabolic entrainment of circadian clocks. O-linked N-acetylglucosamine modification (O-GlcNAcylation) of intracellular proteins is a key mediator of metabolic response to nutrient availability. This review highlights our current understanding of the role of protein O-GlcNAcylation in mediating metabolic input and output of the circadian clock.
Collapse
|
32
|
Taurine Treatment Modulates Circadian Rhythms in Mice Fed A High Fat Diet. Sci Rep 2016; 6:36801. [PMID: 27857215 PMCID: PMC5114685 DOI: 10.1038/srep36801] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 10/18/2016] [Indexed: 11/08/2022] Open
Abstract
Close ties have been made among certain nutrients, obesity, type 2 diabetes and circadian clocks. Among nutrients, taurine has been documented as being effective against obesity and type 2 diabetes. However, the impact of taurine on circadian clocks has not been elucidated. We investigated whether taurine can modulate or correct disturbances in daily rhythms caused by a high-fat diet in mice. Male C57BL/6 mice were divided in four groups: control (C), control + taurine (C+T), high-fat diet (HFD) and HFD + taurine (HFD+T). They were administered 2% taurine in their drinking water for 10 weeks. Mice were euthanized at 6:00, 12:00, 18:00, and 24:00. HFD mice increased body weight, visceral fat and food intake, as well as higher levels of glucose, insulin and leptin, throughout the 24 h. Taurine prevented increments in food intake, body weight and visceral fat, improved glucose tolerance and insulin sensitivity and reduced disturbances in the 24 h patterns of plasma insulin and leptin. HFD downregulated the expression of clock genes Rev-erbα, Bmal1, and Per1 in pancreatic islets. Taurine normalized the gene and protein expression of PER1 in beta-cells, which suggests that it could be beneficial for the correction of daily rhythms and the amelioration of obesity and diabetes.
Collapse
|
33
|
Suárez-Trujillo A, Casey TM. Serotoninergic and Circadian Systems: Driving Mammary Gland Development and Function. Front Physiol 2016; 7:301. [PMID: 27471474 PMCID: PMC4945644 DOI: 10.3389/fphys.2016.00301] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/29/2016] [Indexed: 12/23/2022] Open
Abstract
Since lactation is one of the most metabolically demanding states in adult female mammals, beautifully complex regulatory mechanisms are in place to time lactation to begin after birth and cease when the neonate is weaned. Lactation is regulated by numerous different homeorhetic factors, all of them tightly coordinated with the demands of milk production. Emerging evidence support that among these factors are the serotonergic and circadian clock systems. Here we review the serotoninergic and circadian clock systems and their roles in the regulation of mammary gland development and lactation physiology. We conclude by presenting our hypothesis that these two systems interact to accommodate the metabolic demands of lactation and thus adaptive changes in these systems occur to maintain mammary and systemic homeostasis through the reproductive cycles of female mammals.
Collapse
Affiliation(s)
- Aridany Suárez-Trujillo
- Animal Production and Biotechnology Group, Institute of Animal Health and Food Safety, Universidad de Las Palmas de Gran CanariaArucas, Spain
| | - Theresa M. Casey
- Department of Animal Sciences, Purdue UniversityWest Lafayette, IN, USA
| |
Collapse
|
34
|
Jha PK, Foppen E, Kalsbeek A, Challet E. Sleep restriction acutely impairs glucose tolerance in rats. Physiol Rep 2016; 4:e12839. [PMID: 27354542 PMCID: PMC4923238 DOI: 10.14814/phy2.12839] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/23/2016] [Accepted: 05/31/2016] [Indexed: 11/24/2022] Open
Abstract
Chronic sleep curtailment in humans has been related to impairment of glucose metabolism. To better understand the underlying mechanisms, the purpose of the present study was to investigate the effect of acute sleep deprivation on glucose tolerance in rats. A group of rats was challenged by 4-h sleep deprivation in the early rest period, leading to prolonged (16 h) wakefulness. Another group of rats was allowed to sleep during the first 4 h of the light period and sleep deprived in the next 4 h. During treatment, food was withdrawn to avoid a postmeal rise in plasma glucose. An intravenous glucose tolerance test (IVGTT) was performed immediately after the sleep deprivation period. Sleep deprivation at both times of the day similarly impaired glucose tolerance and reduced the early-phase insulin responses to a glucose challenge. Basal concentrations of plasma glucose, insulin, and corticosterone remained unchanged after sleep deprivation. Throughout IVGTTs, plasma corticosterone concentrations were not different between the control and sleep-deprived group. Together, these results demonstrate that independent of time of day and sleep pressure, short sleep deprivation during the resting phase favors glucose intolerance in rats by attenuating the first-phase insulin response to a glucose load. In conclusion, this study highlights the acute adverse effects of only a short sleep restriction on glucose homeostasis.
Collapse
Affiliation(s)
- Pawan K Jha
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands Regulation of Circadian Clocks team, Institute of Cellular and Integrative Neurosciences UPR3212 Centre National de la Recherche Scientifique (CNRS) University of Strasbourg, Strasbourg, France International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| | - Ewout Foppen
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| | - Etienne Challet
- Regulation of Circadian Clocks team, Institute of Cellular and Integrative Neurosciences UPR3212 Centre National de la Recherche Scientifique (CNRS) University of Strasbourg, Strasbourg, France International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| |
Collapse
|
35
|
Pitchaimani V, Arumugam S, Thandavarayan RA, Karuppagounder V, Afrin MR, Sreedhar R, Harima M, Suzuki H, Miyashita S, Suzuki K, Nakamura M, Ueno K, Watanabe K. Hypothalamic glucagon signaling in fasting hypoglycemia. Life Sci 2016; 153:118-23. [PMID: 27084528 DOI: 10.1016/j.lfs.2016.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/25/2016] [Accepted: 04/05/2016] [Indexed: 11/18/2022]
Abstract
AIMS Sustained glucagon infusion increases hepatic glucose production, but this effect is transient due to hypothalamic glucagon signaling. In hypoglycemia, glucagon acts as a major defense to sustain the blood glucose level and this raises the question regarding glucagon signaling associated glucose production in prolonged fasting hypoglycemia. In this study, we investigated the proteins associated with hypothalamic glucagon signaling and liver gluconeogenesis during fasting hypoglycemia. MAIN METHODS 8-9week old, male C57BL6/J mice were fasted for 4, 8, 12, 18, 24, 30, 36 or 42h. In the hypothalamus, we investigated glucagon signaling by analyzing the glucagon receptor and its downstream protein, peroxisome proliferator-activated receptor-gamma coactivator 1 (PGC-1) expression. In the liver, we investigated gluconeogenesis by analyzing p-protein kinase A (PKA)(Ser/Thr) substrate and phosphoenolpyruvate carboxykinase - cytosolic (PEPCK-C) expression using the western blotting technique. KEY FINDINGS The elevated or trended higher hypothalamic glucagon receptor and PGC-1 expressions at 18 and 42h were correlated with the attenuated liver p-PKA(Ser/Thr) substrate expression. The attenuated hypothalamic glucagon receptor and PGC-1 expressions at 12, 24, 30 and 36h were correlated with the elevated or trended higher liver p-PKA(Ser/Thr) substrate expression. SIGNIFICANCE The hypothalamic glucagon signaling during fasting hypoglycemia might have been modulated by circadian rhythm and this possibly attenuates the liver p-PKA(Ser/Thr) substrate to modify the gluconeogenesis pathway. This mechanism will help to understand the hyperglucagonemia associated complications in diabetes.
Collapse
Affiliation(s)
- Vigneshwaran Pitchaimani
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Somasundaram Arumugam
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | | | - Vengadeshprabhu Karuppagounder
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Mst Rejina Afrin
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Remya Sreedhar
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Meilei Harima
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Hiroshi Suzuki
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Shizuka Miyashita
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Kenji Suzuki
- Department of Gastroenterology, Niigata University of Graduate School of Medicine and Dental Sciences, Niigata City 951-8510, Japan
| | - Masahiko Nakamura
- Department of Cardiology, Yamanashi Prefectural Central Hospital, Kofu, Yamanashi 400-8506, Japan
| | - Kazuyuki Ueno
- Department of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Kenichi Watanabe
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan.
| |
Collapse
|
36
|
Foppen E, Tan AAT, Ackermans MT, Fliers E, Kalsbeek A. Suprachiasmatic Nucleus Neuropeptides and Their Control of Endogenous Glucose Production. J Neuroendocrinol 2016; 28. [PMID: 26791158 DOI: 10.1111/jne.12365] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/11/2016] [Accepted: 01/14/2016] [Indexed: 12/21/2022]
Abstract
Defective control of endogenous glucose production is an important factor responsible for hyperglycaemia in the diabetic individual. During the past decade, progressively more evidence has appeared indicating a strong and potentially causal relationship between disturbances of the circadian system and defects of metabolic regulation, including glucose metabolism. The detrimental effects of disturbed circadian rhythms may have their origin in disturbances of the molecular clock mechanisms in peripheral organs, such as the pancreas and liver, or in the central brain clock in the hypothalamic suprachiasmatic nuclei (SCN). To assess the role of SCN output per se on glucose metabolism, we investigated (i) the effect of several SCN neurotransmitters on endogenous glucose production and (ii) the effect of SCN neuronal activity on hepatic and systemic insulin sensitivity. We show that silencing of SCN neuronal activity results in decreased hepatic insulin sensitivity and increased peripheral insulin sensitivity. Furthermore, both oxytocin neurones in the paraventricular nucleus of the hypothalamus (PVN) and orexin neurones in the lateral hypothalamus may be important targets for the SCN control of glucose metabolism. These data further highlight the role of the central clock in the pathophysiology of insulin resistance.
Collapse
Affiliation(s)
- E Foppen
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - A A T Tan
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - M T Ackermans
- Department of Clinical Chemistry, Laboratory of Endocrinology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - E Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - A Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| |
Collapse
|
37
|
Owino S, Contreras-Alcantara S, Baba K, Tosini G. Melatonin Signaling Controls the Daily Rhythm in Blood Glucose Levels Independent of Peripheral Clocks. PLoS One 2016; 11:e0148214. [PMID: 26824606 PMCID: PMC4732609 DOI: 10.1371/journal.pone.0148214] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/14/2016] [Indexed: 01/01/2023] Open
Abstract
Melatonin is rhythmically secreted by both the pineal gland and retina in a circadian fashion, with its peak synthesis occurring during the night. Once synthesized, melatonin exerts its effects by binding to two specific G-protein coupled receptors-melatonin receptor type 1(MT1) and melatonin receptor type 2(MT2). Recent studies suggest the involvement of MT1 and MT2 in the regulation of glucose homeostasis; however the ability of melatonin signaling to impart timing cues on glucose metabolism remains poorly understood. Here we report that the removal of MT1 or MT2 in mice abolishes the daily rhythm in blood glucose levels. Interestingly, removal of melatonin receptors produced small effects on the rhythmic expression patterns of clock genes within skeletal muscle, liver, and adipose tissue. Taken together, our data suggest that the loss of the daily rhythm in blood glucose observed in MT1(-/-) and MT2(-/-) mice does not occur as a consequence of 'disrupted' clocks within insulin sensitive tissues. Finally our results highlight a diurnal contribution of melatonin receptor signaling in the daily regulation of blood glucose levels.
Collapse
MESH Headings
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/metabolism
- CLOCK Proteins/genetics
- CLOCK Proteins/metabolism
- Circadian Rhythm/genetics
- Gene Expression Regulation
- Homeostasis
- Liver/metabolism
- Male
- Melatonin/metabolism
- Mice
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Pineal Gland/metabolism
- Receptor, Melatonin, MT1/deficiency
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT2/deficiency
- Receptor, Melatonin, MT2/genetics
- Retina/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Sharon Owino
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Susana Contreras-Alcantara
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Kenkichi Baba
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (KB); (GT)
| | - Gianluca Tosini
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (KB); (GT)
| |
Collapse
|
38
|
Tsuneki H, Nagata T, Fujita M, Kon K, Wu N, Takatsuki M, Yamaguchi K, Wada T, Nishijo H, Yanagisawa M, Sakurai T, Sasaoka T. Nighttime Administration of Nicotine Improves Hepatic Glucose Metabolism via the Hypothalamic Orexin System in Mice. Endocrinology 2016; 157:195-206. [PMID: 26492471 DOI: 10.1210/en.2015-1488] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nicotine is known to affect the metabolism of glucose; however, the underlying mechanism remains unclear. Therefore, we here investigated whether nicotine promoted the central regulation of glucose metabolism, which is closely linked to the circadian system. The oral intake of nicotine in drinking water, which mainly occurred during the nighttime active period, enhanced daily hypothalamic prepro-orexin gene expression and reduced hyperglycemia in type 2 diabetic db/db mice without affecting body weight, body fat content, and serum levels of insulin. Nicotine administered at the active period appears to be responsible for the effect on blood glucose, because nighttime but not daytime injections of nicotine lowered blood glucose levels in db/db mice. The chronic oral treatment with nicotine suppressed the mRNA levels of glucose-6-phosphatase, the rate-limiting enzyme of gluconeogenesis, in the liver of db/db and wild-type control mice. In the pyruvate tolerance test to evaluate hepatic gluconeogenic activity, the oral nicotine treatment moderately suppressed glucose elevations in normal mice and mice lacking dopamine receptors, whereas this effect was abolished in orexin-deficient mice and hepatic parasympathectomized mice. Under high-fat diet conditions, the oral intake of nicotine lowered blood glucose levels at the daytime resting period in wild-type, but not orexin-deficient, mice. These results indicated that the chronic daily administration of nicotine suppressed hepatic gluconeogenesis via the hypothalamic orexin-parasympathetic nervous system. Thus, the results of the present study may provide an insight into novel chronotherapy for type 2 diabetes that targets the central cholinergic and orexinergic systems.
Collapse
MESH Headings
- Animals
- Crosses, Genetic
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat/adverse effects
- Drug Chronotherapy
- Gene Expression Regulation/drug effects
- Gluconeogenesis/drug effects
- Hyperglycemia/prevention & control
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/therapeutic use
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Insulin Resistance
- Liver/drug effects
- Liver/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Nicotine/administration & dosage
- Nicotine/therapeutic use
- Nicotinic Agonists/administration & dosage
- Nicotinic Agonists/therapeutic use
- Obesity/complications
- Obesity/etiology
- Orexins/agonists
- Orexins/genetics
- Orexins/metabolism
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Takashi Nagata
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Mikio Fujita
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Kanta Kon
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Naizhen Wu
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Mayumi Takatsuki
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Kaoru Yamaguchi
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Hisao Nishijo
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Masashi Yanagisawa
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Takeshi Sakurai
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology (H.T., T.N., M.F., K.K., N.W., M.T., K.Y., T.W., T.Sas.) and System Emotional Science (H.N.), University of Toyama, Toyama 930-0194, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Y.), University of Tsukuba, Tsukuba 305-8575, Japan; Department of Molecular Genetics (M.Y.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Molecular Neuroscience and Integrative Physiology (T.Sak.), Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
39
|
Kumar Jha P, Challet E, Kalsbeek A. Circadian rhythms in glucose and lipid metabolism in nocturnal and diurnal mammals. Mol Cell Endocrinol 2015; 418 Pt 1:74-88. [PMID: 25662277 DOI: 10.1016/j.mce.2015.01.024] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/12/2015] [Accepted: 01/19/2015] [Indexed: 12/22/2022]
Abstract
Most aspects of energy metabolism display clear variations during day and night. This daily rhythmicity of metabolic functions, including hormone release, is governed by a circadian system that consists of the master clock in the suprachiasmatic nuclei of the hypothalamus (SCN) and many secondary clocks in the brain and peripheral organs. The SCN control peripheral timing via the autonomic and neuroendocrine system, as well as via behavioral outputs. The sleep-wake cycle, the feeding/fasting rhythm and most hormonal rhythms, including that of leptin, ghrelin and glucocorticoids, usually show an opposite phase (relative to the light-dark cycle) in diurnal and nocturnal species. By contrast, the SCN clock is most active at the same astronomical times in these two categories of mammals. Moreover, in both species, pineal melatonin is secreted only at night. In this review we describe the current knowledge on the regulation of glucose and lipid metabolism by central and peripheral clock mechanisms. Most experimental knowledge comes from studies in nocturnal laboratory rodents. Nevertheless, we will also mention some relevant findings in diurnal mammals, including humans. It will become clear that as a consequence of the tight connections between the circadian clock system and energy metabolism, circadian clock impairments (e.g., mutations or knock-out of clock genes) and circadian clock misalignments (such as during shift work and chronic jet-lag) have an adverse effect on energy metabolism, that may trigger or enhancing obese and diabetic symptoms.
Collapse
Affiliation(s)
- Pawan Kumar Jha
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France; International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| | - Etienne Challet
- Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France; International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands; Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Mayeuf-Louchart A, Staels B, Duez H. Skeletal muscle functions around the clock. Diabetes Obes Metab 2015; 17 Suppl 1:39-46. [PMID: 26332967 DOI: 10.1111/dom.12517] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/03/2015] [Indexed: 12/16/2022]
Abstract
In mammals, the central clock localized in the central nervous system imposes a circadian rhythmicity to all organs. This is achieved thanks to a well-conserved molecular clockwork, involving interactions between several transcription factors, whose pace is conveyed to peripheral tissues through neuronal and humoral signals. The molecular clock plays a key role in the control of numerous physiological processes and takes part in the regulation of metabolism and energy balance. Skeletal muscle is one of the peripheral organs whose function is under the control of the molecular clock. However, although skeletal muscle metabolism and performances display circadian rhythmicity, the role of the molecular clock in the skeletal muscle has remained unappreciated for years. Peripheral organs such as skeletal muscle, and the liver, among others, can be desynchronized from the central clock by external stimuli, such as feeding or exercise, which impose a new rhythm at the organism level. In this review, we discuss our current understanding of the clock in skeletal muscle circadian physiology, focusing on the control of myogenesis and skeletal muscle metabolism.
Collapse
Affiliation(s)
- A Mayeuf-Louchart
- University of Lille, U1011, EGID, F-59000, Lille, France
- INSERM, U1011, F-59000 Lille, France
- CHU Lille, F-59000, Lille, France
- Institut Pasteur de Lille, U1011, F-59000 Lille, France
| | - B Staels
- University of Lille, U1011, EGID, F-59000, Lille, France
- INSERM, U1011, F-59000 Lille, France
- CHU Lille, F-59000, Lille, France
- Institut Pasteur de Lille, U1011, F-59000 Lille, France
| | - H Duez
- University of Lille, U1011, EGID, F-59000, Lille, France
- INSERM, U1011, F-59000 Lille, France
- CHU Lille, F-59000, Lille, France
- Institut Pasteur de Lille, U1011, F-59000 Lille, France
| |
Collapse
|
41
|
Tsuneki H, Tokai E, Nakamura Y, Takahashi K, Fujita M, Asaoka T, Kon K, Anzawa Y, Wada T, Takasaki I, Kimura K, Inoue H, Yanagisawa M, Sakurai T, Sasaoka T. Hypothalamic orexin prevents hepatic insulin resistance via daily bidirectional regulation of autonomic nervous system in mice. Diabetes 2015; 64:459-70. [PMID: 25249578 DOI: 10.2337/db14-0695] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Circadian rhythm is crucial for preventing hepatic insulin resistance, although the mechanism remains uncovered. Here we report that the wake-active hypothalamic orexin system plays a key role in this regulation. Wild-type mice showed that a daily rhythm in blood glucose levels peaked at the awake period; however, the glucose rhythm disappeared in orexin knockout mice despite normal feeding rhythm. Central administration of orexin A during nighttime awake period acutely elevated blood glucose levels but subsequently lowered daytime glucose levels in normal and diabetic db/db mice. The glucose-elevating and -lowering effects of orexin A were suppressed by adrenergic antagonists and hepatic parasympathectomy, respectively. Moreover, the expression levels of hepatic gluconeogenic genes, including Pepck, were increased and decreased by orexin A at nanomolar and femtomolar doses, respectively. These results indicate that orexin can bidirectionally regulate hepatic gluconeogenesis via control of autonomic balance, leading to generation of the daily blood glucose oscillation. Furthermore, during aging, orexin deficiency enhanced endoplasmic reticulum (ER) stress in the liver and caused impairment of hepatic insulin signaling and abnormal gluconeogenic activity in pyruvate tolerance test. Collectively, the daily glucose rhythm under control of orexin appears to be important for maintaining ER homeostasis, thereby preventing insulin resistance in the liver.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Emi Tokai
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Yuya Nakamura
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Keisuke Takahashi
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Mikio Fujita
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Takehiro Asaoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Kanta Kon
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Yuuki Anzawa
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Ichiro Takasaki
- Division of Molecular Genetics Research, University of Toyama, Toyama, Japan
| | - Kumi Kimura
- Frontier Science Organization, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroshi Inoue
- Frontier Science Organization, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan Howard Hughes Medical Institute, Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Takeshi Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| |
Collapse
|
42
|
Bisschop PH, Fliers E, Kalsbeek A. Autonomic Regulation of Hepatic Glucose Production. Compr Physiol 2014; 5:147-65. [DOI: 10.1002/cphy.c140009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
Vieira E, Burris TP, Quesada I. Clock genes, pancreatic function, and diabetes. Trends Mol Med 2014; 20:685-93. [PMID: 25457619 DOI: 10.1016/j.molmed.2014.10.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/10/2014] [Accepted: 10/14/2014] [Indexed: 11/28/2022]
Abstract
Circadian physiology is responsible for the temporal regulation of metabolism to optimize energy homeostasis throughout the day. Disturbances in the light/dark cycle, sleep/wake schedule, or feeding/activity behavior can affect the circadian function of the clocks located in the brain and peripheral tissues. These alterations have been associated with impaired glucose tolerance and type 2 diabetes. Animal models with molecular manipulation of clock genes and genetic studies in humans also support these links. It has been demonstrated that the endocrine pancreas has an intrinsic self-sustained clock, and recent studies have revealed an important role of clock genes in pancreatic β cells, glucose homeostasis, and diabetes.
Collapse
Affiliation(s)
- Elaine Vieira
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08033 Barcelona, Spain.
| | - Thomas P Burris
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Ivan Quesada
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08033 Barcelona, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández, 03202 Elche, Spain.
| |
Collapse
|
44
|
Mendoza J, Challet E. Circadian insights into dopamine mechanisms. Neuroscience 2014; 282:230-42. [PMID: 25281877 DOI: 10.1016/j.neuroscience.2014.07.081] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 07/22/2014] [Accepted: 07/24/2014] [Indexed: 01/11/2023]
Abstract
Almost every physiological or behavioral process in mammals follows rhythmic patterns, which depend mainly on a master circadian clock located in the hypothalamic suprachiasmatic nucleus (SCN). The dopaminergic (DAergic) system in the brain is principally implicated in motor functions, motivation and drug intake. Interestingly, DA-related parameters and behaviors linked to the motivational and arousal states, show daily rhythms that could be regulated by the SCN or by extra-SCN circadian oscillator(s) modulating DAergic systems. Here we examine what is currently understood about the anatomical and functional central multi-oscillatory circadian system, highlighting how the main SCN clock communicates timing information with other brain clocks to regulate the DAergic system and conversely, how DAergic cues may have feedback effects on the SCN. These studies give new insights into the role of the brain circadian system in DA-related neurologic pathologies, such as Parkinson's disease, attention deficit/hyperactive disorder and drug addiction.
Collapse
Affiliation(s)
- J Mendoza
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, 5 rue Blaise Pascal, 67084 Strasbourg cedex, France.
| | - E Challet
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, 5 rue Blaise Pascal, 67084 Strasbourg cedex, France
| |
Collapse
|
45
|
Müller MH, Rödel F, Rüb U, Korf HW. Irradiation with X-rays phase-advances the molecular clockwork in liver, adrenal gland and pancreas. Chronobiol Int 2014; 32:27-36. [DOI: 10.3109/07420528.2014.949735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
46
|
Donjacour CEHM, Aziz NA, Overeem S, Kalsbeek A, Pijl H, Lammers GJ. Glucose and fat metabolism in narcolepsy and the effect of sodium oxybate: a hyperinsulinemic-euglycemic clamp study. Sleep 2014; 37:795-801. [PMID: 24899766 DOI: 10.5665/sleep.3592] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Narcolepsy is associated with obesity though it is uncertain whether this is caused by changes in glucose and fat metabolism. Therefore, we performed a detailed analysis of systemic energy homeostasis in narcolepsy patients, and additionally, investigated whether it was affected by three months of sodium oxybate (SXB) treatment. METHODS Nine hypocretin deficient patients with narcolepsy-cataplexy, and nine healthy sex, age, and BMI matched controls were enrolled. A hyperinsulinemic-euglycemic clamp combined with stable isotopes ([6,6-(2)H2]-glucose and [(2)H5]- glycerol) was performed at baseline. In seven patients a second study was performed after three months of SXB treatment. RESULTS Glucose disposal rate (GDR) per unit serum insulin was significantly higher in narcolepsy patients compared to matched controls (1.6 ± 0.2 vs. 1.1 ± 0.3 μmol/kgFFM/min/mU×L; P = 0.024), whereas β-cell function was similar (P = 0.50). Basal steady state glycerol appearance rate tended to be lower in narcolepsy patients (5.2 ± 0.4 vs. 7.5 ± 1.3 μmol/kgFM/min; P = 0.058), suggesting a lower rate of lipolysis. SXB treatment induced a trend in reduction of the GDR (1.4 ± 0.1 vs. 1.1 ± 0.2 μmol/kgFFM/min/mU×L; P = 0.063) and a reduction in endogenous glucose production (0.24 ± 0.03 vs. 0.16 ± 0.03 μmol/kgFFM/min/mU×L: P = 0.028) per unit serum insulin. After SXB treatment lipolysis increased (4.9 ± 0.4 vs. 6.5 ± 0.6 μmol/kgFM/min; P = 0.018), and body weight decreased in narcolepsy patients (99.2 ± 6.0 vs. 94.0 ± 5.4 kg; P = 0.044). CONCLUSION We show that narcolepsy patients are more insulin sensitive and may have a lower rate of lipolysis than matched controls. SXB stimulated lipolysis in narcolepsy patients, possibly accounting for the weight loss after treatment. While sodium oxybate tended to decrease systemic insulin sensitivity, it increased hepatic insulin sensitivity, suggesting tissue-specific effects.
Collapse
Affiliation(s)
| | - N Ahmad Aziz
- Department of Neurology Leiden University Medical Centre, Leiden, The Netherlands
| | - Sebastiaan Overeem
- Department of Neurology, Radboud University Medical Centre, Nijmegen, The Netherlands ; Sleep Medicine Centre "Kempenhaeghe," Heeze, The Netherlands
| | - Andries Kalsbeek
- Netherlands Institute for Neuroscience, Hypothalamic Integration Mechanisms, Amsterdam, The Netherlands ; Department of Endocrinology and Metabolism, Academic Medical Centre of the University of Amsterdam, Amsterdam, The Netherlands
| | - Hanno Pijl
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gert Jan Lammers
- Department of Neurology Leiden University Medical Centre, Leiden, The Netherlands ; Sleep Wake Center SEIN, Heemstede, The Netherlands
| |
Collapse
|
47
|
Kalsbeek A, la Fleur S, Fliers E. Circadian control of glucose metabolism. Mol Metab 2014; 3:372-83. [PMID: 24944897 PMCID: PMC4060304 DOI: 10.1016/j.molmet.2014.03.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 01/15/2023] Open
Abstract
The incidence of obesity and type 2 diabetes mellitus (T2DM) has risen to epidemic proportions. The pathophysiology of T2DM is complex and involves insulin resistance, pancreatic β-cell dysfunction and visceral adiposity. It has been known for decades that a disruption of biological rhythms (which happens the most profoundly with shift work) increases the risk of developing obesity and T2DM. Recent evidence from basal studies has further sparked interest in the involvement of daily rhythms (and their disruption) in the development of obesity and T2DM. Most living organisms have molecular clocks in almost every tissue, which govern rhythmicity in many domains of physiology, such as rest/activity rhythms, feeding/fasting rhythms, and hormonal secretion. Here we present the latest research describing the specific role played by the molecular clock mechanism in the control of glucose metabolism and speculate on how disruption of these tissue clocks may lead to the disturbances in glucose homeostasis.
Collapse
Affiliation(s)
- Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands ; Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Susanne la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| |
Collapse
|
48
|
Cui X, Abduljalil A, Manor BD, Peng CK, Novak V. Multi-scale glycemic variability: a link to gray matter atrophy and cognitive decline in type 2 diabetes. PLoS One 2014; 9:e86284. [PMID: 24475100 PMCID: PMC3901681 DOI: 10.1371/journal.pone.0086284] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 12/11/2013] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Type 2 diabetes mellitus (DM) accelerates brain aging and cognitive decline. Complex interactions between hyperglycemia, glycemic variability and brain aging remain unresolved. This study investigated the relationship between glycemic variability at multiple time scales, brain volumes and cognition in type 2 DM. RESEARCH DESIGN AND METHODS Forty-three older adults with and 26 without type 2 DM completed 72-hour continuous glucose monitoring, cognitive tests and anatomical MRI. We described a new analysis of continuous glucose monitoring, termed Multi-Scale glycemic variability (Multi-Scale GV), to examine glycemic variability at multiple time scales. Specifically, Ensemble Empirical Mode Decomposition was used to identify five unique ultradian glycemic variability cycles (GVC1-5) that modulate serum glucose with periods ranging from 0.5-12 hrs. RESULTS Type 2 DM subjects demonstrated greater variability in GVC3-5 (period 2.0-12 hrs) than controls (P<0.0001), during the day as well as during the night. Multi-Scale GV was related to conventional markers of glycemic variability (e.g. standard deviation and mean glycemic excursions), but demonstrated greater sensitivity and specificity to conventional markers, and was associated with worse long-term glycemic control (e.g. fasting glucose and HbA1c). Across all subjects, those with greater glycemic variability within higher frequency cycles (GVC1-3; 0.5-2.0 hrs) had less gray matter within the limbic system and temporo-parietal lobes (e.g. cingulum, insular, hippocampus), and exhibited worse cognitive performance. Specifically within those with type 2 DM, greater glycemic variability in GVC2-3 was associated with worse learning and memory scores. Greater variability in GVC5 was associated with longer DM duration and more depression. These relationships were independent of HbA1c and hypoglycemic episodes. CONCLUSIONS Type 2 DM is associated with dysregulation of glycemic variability over multiple scales of time. These time-scale-dependent glycemic fluctuations might contribute to brain atrophy and cognitive outcomes within this vulnerable population.
Collapse
Affiliation(s)
- Xingran Cui
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amir Abduljalil
- Wright Center of Innovation, Dept. of Radiology, The Ohio State University, Columbus Ohio, United States of America
| | - Brad D. Manor
- Institute for Aging Research, Hebrew SeniorLife, Roslindale, Massachusetts, United States of America
- Division of Gerontology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chung-Kang Peng
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Dynamical Biomarkers and Translational Medicine, National Central University, Chung-Li, Taiwan
| | - Vera Novak
- Division of Stroke, Dept. of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
49
|
Maury E, Hong HK, Bass J. Circadian disruption in the pathogenesis of metabolic syndrome. DIABETES & METABOLISM 2014; 40:338-46. [PMID: 24433933 DOI: 10.1016/j.diabet.2013.12.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 12/15/2013] [Accepted: 12/16/2013] [Indexed: 12/19/2022]
Abstract
Metabolic syndrome is a multifactorial process induced by a combination of genetic and environmental factors and recent evidence has highlighted that circadian disruption and sleep loss contribute to disease pathogenesis. Emerging work in experimental genetic models has provided insight into the mechanistic basis for clock disruption in disease. Indeed, disruption of the clock system perturbs both neuroendocrine pathways within the hypothalamus important in feeding and energetics, in addition to peripheral tissues involved in glucose and lipid metabolism. This review illustrates the impact of molecular clock disruptions at the level of both brain and behavior and peripheral tissues, with a focus on how such dysregulation in turn impacts lipid and glucose homeostasis, inflammation and cardiovascular function. New insight into circadian biology may ultimately lead to improved therapeutics for metabolic syndrome and cardiovascular disease in humans.
Collapse
Affiliation(s)
- E Maury
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Lurie 7-220, Chicago, Illinois 60611, USA.
| | - H K Hong
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Lurie 7-220, Chicago, Illinois 60611, USA
| | - J Bass
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Lurie 7-220, Chicago, Illinois 60611, USA.
| |
Collapse
|
50
|
Abstract
The diurnal variation of the geophysical position of the earth in relation to the sun has imposed considerable evolutionary pressure. The suprachiasmatic nucleus, which serves as the central biological clock, receives the input regarding light-dark through the optic nerves. This nucleus in turn conveys output in a diurnal fashion to other hypothalamic nuclei and to the autonomic nervous system. Sleep is the most extreme phenotypical adaptation to this diurnal light-dark cycle. In recent years, sleep duration has been reduced and sleep deprivation has become endemic in our modern 24/7 society, either by voluntary sleep restriction and/or through sleep disorders. Experimental studies in humans have documented that sleep deprivation induces insulin resistance in multiple metabolic pathways in both healthy subjects and patients with type 1 diabetes. Epidemiological studies have documented that sleep duration is an important risk factor for development of insulin resistance and type 2 diabetes. Several potential pathways have been suggested to contribute to insulin resistance after sleep restriction, including altered function of the autonomic nervous system, endocrine changes, and an altered inflammatory state. Nonetheless, the causal factors explaining the relation between altered sleep characteristics and insulin resistance in multiple organs need additional study, and most likely include central autonomic pathways.
Collapse
Affiliation(s)
- Esther Donga
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes A Romijn
- Department of Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|