151
|
The endogenous circadian clock programs animals to eat at certain times of the 24-hour day: What if we ignore the clock? Physiol Behav 2018; 193:211-217. [PMID: 29673860 DOI: 10.1016/j.physbeh.2018.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 02/06/2023]
Abstract
The discovery of the molecular mechanisms underlying the circadian clock, which functions in virtually every cell throughout the body to coordinate biological processes to anticipate and better adapt to daily rhythmic changes in the environment, is one of the major biomedical breakthroughs in the 20th century. Twenty years after this breakthrough, the biomedical community is now at a new frontier to incorporate the circadian clock mechanisms into many areas of biomedical research, as studies continue to reveal an important role of the circadian clock in a wide range of biological functions and diseases. A forefront of this exciting area is the research of interactions between the clock and energy metabolism. In this review, we summarize animal and human studies linking disruptions of the circadian clock, either environmental or genetic, to metabolic dysfunctions associated with obesity, diabetes, and other metabolic disorders. We also discuss how these advances in circadian biology may pave the way to revolutionize clinical practice in the era of precision medicine.
Collapse
|
152
|
Spracklen CN, Shi J, Vadlamudi S, Wu Y, Zou M, Raulerson CK, Davis JP, Zeynalzadeh M, Jackson K, Yuan W, Wang H, Shou W, Wang Y, Luo J, Lange LA, Lange EM, Popkin BM, Gordon-Larsen P, Du S, Huang W, Mohlke KL. Identification and functional analysis of glycemic trait loci in the China Health and Nutrition Survey. PLoS Genet 2018; 14:e1007275. [PMID: 29621232 PMCID: PMC5886383 DOI: 10.1371/journal.pgen.1007275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/23/2018] [Indexed: 12/17/2022] Open
Abstract
To identify genetic contributions to type 2 diabetes (T2D) and related glycemic traits (fasting glucose, fasting insulin, and HbA1c), we conducted genome-wide association analyses (GWAS) in up to 7,178 Chinese subjects from nine provinces in the China Health and Nutrition Survey (CHNS). We examined patterns of population structure within CHNS and found that allele frequencies differed across provinces, consistent with genetic drift and population substructure. We further validated 32 previously described T2D- and glycemic trait-loci, including G6PC2 and SIX3-SIX2 associated with fasting glucose. At G6PC2, we replicated a known fasting glucose-associated variant (rs34177044) and identified a second signal (rs2232326), a low-frequency (4%), probably damaging missense variant (S324P). A variant within the lead fasting glucose-associated signal at SIX3-SIX2 co-localized with pancreatic islet expression quantitative trait loci (eQTL) for SIX3, SIX2, and three noncoding transcripts. To identify variants functionally responsible for the fasting glucose association at SIX3-SIX2, we tested five candidate variants for allelic differences in regulatory function. The rs12712928-C allele, associated with higher fasting glucose and lower transcript expression level, showed lower transcriptional activity in reporter assays and increased binding to GABP compared to the rs12712928-G, suggesting that rs12712928-C contributes to elevated fasting glucose levels by disrupting an islet enhancer, resulting in reduced gene expression. Taken together, these analyses identified multiple loci associated with glycemic traits across China, and suggest a regulatory mechanism at the SIX3-SIX2 fasting glucose GWAS locus.
Collapse
Affiliation(s)
- Cassandra N. Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jinxiu Shi
- Department of Genetics, Shanghai-MOST Key Laboratory of Heath and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute, Shanghai, China
| | - Swarooparani Vadlamudi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ying Wu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Meng Zou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Chelsea K. Raulerson
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - James P. Davis
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Monica Zeynalzadeh
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kayla Jackson
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wentao Yuan
- Department of Genetics, Shanghai-MOST Key Laboratory of Heath and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute, Shanghai, China
| | - Haifeng Wang
- Department of Genetics, Shanghai-MOST Key Laboratory of Heath and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute, Shanghai, China
| | - Weihua Shou
- Department of Genetics, Shanghai-MOST Key Laboratory of Heath and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute, Shanghai, China
| | - Ying Wang
- Department of Genetics, Shanghai-MOST Key Laboratory of Heath and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute, Shanghai, China
| | - Jingchun Luo
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Leslie A. Lange
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Ethan M. Lange
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Barry M. Popkin
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Penny Gordon-Larsen
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Shufa Du
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Heath and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute, Shanghai, China
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
153
|
Onk D, Onk OA, Erol HS, Özkaraca M, Çomaklı S, Ayazoğlu TA, Kuyrukluyıldız U, Ünver S. Effect of melatonin on antioxidant capacity, ınflammation and apoptotic cell death in lung tissue of diabetic rats. Acta Cir Bras 2018; 33:375-385. [DOI: 10.1590/s0102-865020180040000009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/13/2018] [Indexed: 01/05/2023] Open
|
154
|
Humphries PS, Bersot R, Kincaid J, Mabery E, McCluskie K, Park T, Renner T, Riegler E, Steinfeld T, Turtle ED, Wei ZL, Willis E. Carbazole-containing amides and ureas: Discovery of cryptochrome modulators as antihyperglycemic agents. Bioorg Med Chem Lett 2018; 28:293-297. [DOI: 10.1016/j.bmcl.2017.12.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
|
155
|
A common variant within the HNF1B gene is associated with overall survival of multiple myeloma patients: results from the IMMEnSE consortium and meta-analysis. Oncotarget 2018; 7:59029-59048. [PMID: 27437873 PMCID: PMC5312293 DOI: 10.18632/oncotarget.10665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/19/2016] [Indexed: 01/01/2023] Open
Abstract
Diabetogenic single nucleotide polymorphisms (SNPs) have recently been associated with multiple myeloma (MM) risk but their impact on overall survival (OS) of MM patients has not been analysed yet. In order to investigate the impact of 58 GWAS-identified variants for type 2 diabetes (T2D) on OS of patients with MM, we analysed genotyping data of 936 MM patients collected by the International Multiple Myeloma rESEarch (IMMENSE) consortium and an independent set of 700 MM patients recruited by the University Clinic of Heidelberg. A meta-analysis of the cox regression results of the two sets showed that rs7501939 located in the HNF1B gene negatively impacted OS (HRRec= 1.44, 95% CI = 1.18-1.76, P = 0.0001). The meta-analysis also showed a noteworthy gender-specific association of the SLC30A8rs13266634 SNP with OS. The presence of each additional copy of the minor allele at rs13266634 was associated with poor OS in men whereas no association was seen in women (HRMen-Add = 1.32, 95% CI 1.13-1.54, P = 0.0003). In conclusion, these data suggest that the HNF1Brs7501939 SNP confers poor OS in patients with MM and that a SNP in SLC30A8 affect OS in men.
Collapse
|
156
|
Abstract
We examined whether established metabolic risk genetic variants in the population confer a risk for increased waist circumference in patients with schizophrenia spectrum disorders and also an association with schizophrenia spectrum disorders irrespective of waist circumference.
Collapse
|
157
|
Abstract
This chapter reviews both statistical and physiologic issues related to the pathophysiologic effects of genetic variation in the context of type 2 diabetes. The goal is to review current methodologies used to analyze disease-related quantitative traits for those who do not have extensive quantitative and physiologic background, as an attempt to bridge that gap. We leverage mathematical modeling to illustrate the strengths and weaknesses of different approaches and attempt to reinforce with real data analysis. Topics reviewed include phenotype selection, phenotype specificity, multiple variant analysis via the genetic risk score, and consideration of multiple disease-related phenotypes. Type 2 diabetes is used as the example, not only because of the extensive existing knowledge at the genetic, physiologic, clinical, and epidemiologic levels, but also because type 2 diabetes has been at the forefront of complex disease genetics, with many examples to draw from.
Collapse
Affiliation(s)
- Richard M Watanabe
- Departments of Preventive Medicine and Physiology & Biophysics, Keck School of Medicine of USC, 2250 Alcazar Street, CSC-204, Los Angeles, CA, 90089-9073, USA.
| |
Collapse
|
158
|
Central Circadian Clock Regulates Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:79-103. [PMID: 30390286 DOI: 10.1007/978-981-13-1286-1_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Our body not only responds to environmental changes but also anticipates them. The light and dark cycle with the period of about 24 h is a recurring environmental change that determines the diurnal variation in food availability and safety from predators in nature. As a result, the circadian clock is evolved in most animals to align locomotor behaviors and energy metabolism with the light cue. The central circadian clock in mammals is located at the suprachiasmatic nucleus (SCN) of the hypothalamus in the brain. We here review the molecular and anatomic architecture of the central circadian clock in mammals, describe the experimental and observational evidence that suggests a critical role of the central circadian clock in shaping systemic energy metabolism, and discuss the involvement of endocrine factors, neuropeptides, and the autonomic nervous system in the metabolic functions of the central circadian clock.
Collapse
|
159
|
Spinedi E, Cardinali DP. The Polycystic Ovary Syndrome and the Metabolic Syndrome: A Possible Chronobiotic-Cytoprotective Adjuvant Therapy. Int J Endocrinol 2018; 2018:1349868. [PMID: 30147722 PMCID: PMC6083563 DOI: 10.1155/2018/1349868] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome is a highly frequent reproductive-endocrine disorder affecting up to 8-10% of women worldwide at reproductive age. Although its etiology is not fully understood, evidence suggests that insulin resistance, with or without compensatory hyperinsulinemia, and hyperandrogenism are very common features of the polycystic ovary syndrome phenotype. Dysfunctional white adipose tissue has been identified as a major contributing factor for insulin resistance in polycystic ovary syndrome. Environmental (e.g., chronodisruption) and genetic/epigenetic factors may also play relevant roles in syndrome development. Overweight and/or obesity are very common in women with polycystic ovary syndrome, thus suggesting that some polycystic ovary syndrome and metabolic syndrome female phenotypes share common characteristics. Sleep disturbances have been reported to double in women with PCOS and obstructive sleep apnea is a common feature in polycystic ovary syndrome patients. Maturation of the luteinizing hormone-releasing hormone secretion pattern in girls in puberty is closely related to changes in the sleep-wake cycle and could have relevance in the pathogenesis of polycystic ovary syndrome. This review article focuses on two main issues in the polycystic ovary syndrome-metabolic syndrome phenotype development: (a) the impact of androgen excess on white adipose tissue function and (b) the possible efficacy of adjuvant melatonin therapy to improve the chronobiologic profile in polycystic ovary syndrome-metabolic syndrome individuals. Genetic variants in melatonin receptor have been linked to increased risk of developing polycystic ovary syndrome, to impairments in insulin secretion, and to increased fasting glucose levels. Melatonin therapy may protect against several metabolic syndrome comorbidities in polycystic ovary syndrome and could be applied from the initial phases of patients' treatment.
Collapse
Affiliation(s)
- Eduardo Spinedi
- Centre for Experimental and Applied Endocrinology (CENEXA, UNLP-CONICET-FCM), CEAS-CICPBA, La Plata Medical School, La Plata, Argentina
| | - Daniel P. Cardinali
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
160
|
Riddy DM, Delerive P, Summers RJ, Sexton PM, Langmead CJ. G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacol Rev 2018; 70:39-67. [PMID: 29233848 DOI: 10.1124/pr.117.014373] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/13/2017] [Indexed: 03/21/2025] Open
Abstract
G protein-coupled receptors (GPCRs) continue to be important discovery targets for the treatment of type 2 diabetes mellitus (T2DM). Many GPCRs are directly involved in the development of insulin resistance and β-cell dysfunction, and in the etiology of inflammation that can lead to obesity-induced T2DM. This review summarizes the current literature describing a number of well-validated GPCR targets, but also outlines several new and promising targets for drug discovery. We highlight the importance of understanding the role of these receptors in the disease pathology, and their basic pharmacology, which will pave the way to the development of novel pharmacological probes that will enable these targets to fulfill their promise for the treatment of these metabolic disorders.
Collapse
Affiliation(s)
- Darren M Riddy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Philippe Delerive
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| |
Collapse
|
161
|
Mercader JM, Florez JC. The Genetic Basis of Type 2 Diabetes in Hispanics and Latin Americans: Challenges and Opportunities. Front Public Health 2017; 5:329. [PMID: 29376044 PMCID: PMC5763127 DOI: 10.3389/fpubh.2017.00329] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022] Open
Abstract
Type 2 diabetes (T2D) affects 415 million people worldwide, and has a much higher prevalence in Hispanics (16.9%), compared to non-Hispanic whites (10.2%). Genome-wide association studies and whole-genome and whole-exome sequencing studies have discovered more than 100 genetic regions associated with modified risk for T2D. However, the identified genetic factors explain a very small fraction of the estimated heritability. Until recently, little attention has been put in studying other non European populations that suffer from a higher burden of T2D, such as Hispanics/Latinos. In the past few years, genetic studies in Hispanic populations have started to provide new insights into the genetic architecture of T2D in this ancestry group. Of note, several genetic variants that are absent or very rare in non-Hispanic populations but more common in Hispanics have shown from moderate to strong association with T2D and have provided new insights into the biology of T2D, which may be ultimately useful for developing novel therapeutic strategies applicable to all populations. Studying diverse populations can also improve the ability to find the causal variants in known T2D loci by a multi-ancestry fine-mapping approach, which leverages the different patterns of linkage disequilibrium between the causal and the ascertained genetic variants. In this mini-review, we summarize the main genetic findings discovered in Hispanics and discuss the limitations and challenges of performing genetic studies in these populations. Finally, we present possible next steps to make studies in Latino populations more valuable in providing a deeper understanding of T2D and anticipate their future application to the development of predictive and preventive medicine and personalized therapies.
Collapse
Affiliation(s)
- Josep M Mercader
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, United States.,Diabetes Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, United States.,Diabetes Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
162
|
Exposure to Night-Time Traffic Noise, Melatonin-Regulating Gene Variants and Change in Glycemia in Adults. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14121492. [PMID: 29194408 PMCID: PMC5750910 DOI: 10.3390/ijerph14121492] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 11/16/2022]
Abstract
Traffic noise has been linked to diabetes, with limited understanding of its mechanisms. We hypothesize that night-time road traffic noise (RTN) may impair glucose homeostasis through circadian rhythm disturbances. We prospectively investigated the relationship between residential night-time RTN and subsequent eight-year change in glycosylated hemoglobin (ΔHbA1c) in 3350 participants of the Swiss Cohort Study on Air Pollution and Lung and Heart Diseases in Adults (SAPALDIA), adjusting for diabetes risk factors and air pollution levels. Annual average RTN (Lnight) was assigned to participants in 2001 using validated Swiss noise models. HbA1c was measured in 2002 and 2011 using liquid chromatography. We applied mixed linear models to explore RTN–ΔHbA1c association and its modification by a genetic risk score of six common circadian-related MTNR1B variants (MGRS). A 10 dB difference in RTN was associated with a 0.02% (0.003–0.04%) increase in mean ΔHbA1c in 2142 non-movers. RTN–ΔHbA1c association was modified by MGRS among diabetic participants (Pinteraction = 0.001). A similar trend in non-diabetic participants was non-significant. Among the single variants, we observed strongest interactions with rs10830963, an acknowledged diabetes risk variant also implicated in melatonin profile dysregulation. Night-time RTN may impair glycemic control, especially in diabetic individuals, through circadian rhythm disturbances. Experimental sleep studies are needed to test whether noise control may help individuals to attain optimal glycemic levels.
Collapse
|
163
|
Szabo M, Máté B, Csép K, Benedek T. Genetic Approaches to the Study of Gene Variants and Their Impact on the Pathophysiology of Type 2 Diabetes. Biochem Genet 2017; 56:22-55. [DOI: 10.1007/s10528-017-9827-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022]
|
164
|
Cardinali DP, Vigo DE. Melatonin, mitochondria, and the metabolic syndrome. Cell Mol Life Sci 2017; 74:3941-3954. [PMID: 28819865 PMCID: PMC11107716 DOI: 10.1007/s00018-017-2611-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/03/2017] [Indexed: 12/12/2022]
Abstract
A number of risk factors for cardiovascular disease including hyperinsulinemia, glucose intolerance, dyslipidemia, obesity, and elevated blood pressure are collectively known as metabolic syndrome (MS). Since mitochondrial activity is modulated by the availability of energy in cells, the disruption of key regulators of metabolism in MS not only affects the activity of mitochondria but also their dynamics and turnover. Therefore, a link of MS with mitochondrial dysfunction has been suspected since long. As a chronobiotic/cytoprotective agent, melatonin has a special place in prevention and treatment of MS. Melatonin levels are reduced in diseases associated with insulin resistance like MS. Melatonin improves sleep efficiency and has antioxidant and anti-inflammatory properties, partly for its role as a metabolic regulator and mitochondrial protector. We discuss in the present review the several cytoprotective melatonin actions that attenuate inflammatory responses in MS. The clinical data that support the potential therapeutical value of melatonin in human MS are reviewed.
Collapse
Affiliation(s)
- Daniel P Cardinali
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Av. Alicia Moreau de Justo 1500, 4o piso, 1107, Buenos Aires, Argentina.
| | - Daniel E Vigo
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Av. Alicia Moreau de Justo 1500, 4o piso, 1107, Buenos Aires, Argentina
| |
Collapse
|
165
|
Bonnefond A, Froguel P. Disentangling the Role of Melatonin and its Receptor MTNR1B in Type 2 Diabetes: Still a Long Way to Go? Curr Diab Rep 2017; 17:122. [PMID: 29063374 DOI: 10.1007/s11892-017-0957-1] [Citation(s) in RCA: 13] [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] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Type 2 diabetes (T2D) is a complex genetic metabolic disorder. T2D heritability has been estimated around 40-70%. In the last decade, exponential progress has been made in identifying T2D genetic determinants, through genome-wide association studies (GWAS). Among single-nucleotide polymorphisms mostly associated with T2D risk, rs10830963 is located in the MTNR1B gene, encoding one of the two receptors of melatonin, a neurohormone involved in circadian rhythms. Subsequent studies aiming to disentangle the role of MTNR1B in T2D pathophysiology led to controversies. In this review, we will tackle them and will try to give some directions to get a better view of MTNR1B contribution to T2D pathophysiology. RECENT FINDINGS Recent studies either based on genetic/genomic analyses, clinical/epidemiology data, functional analyses at rs10830963 locus, insulin secretion assays in response to melatonin (involving or not MTNR1B) or animal model analyses have led to strong controversies at each level of interpretation. We discuss possible caveats in these studies and present ways to go beyond these issues, towards a better understanding of T2D molecular mechanisms, keeping in mind that melatonin is a versatile hormone and regulates many functions via its primary role in the body clock.
Collapse
Affiliation(s)
- Amélie Bonnefond
- CNRS UMR 8199. European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Pôle Recherche-1er - 1er étage Aile Ouest, 1 place de Verdun, 59045, Lille Cedex, France.
| | - Philippe Froguel
- CNRS UMR 8199. European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Pôle Recherche-1er - 1er étage Aile Ouest, 1 place de Verdun, 59045, Lille Cedex, France
- Genomics of Common Disease, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
166
|
Wheeler E, Marenne G, Barroso I. Genetic aetiology of glycaemic traits: approaches and insights. Hum Mol Genet 2017; 26:R172-R184. [PMID: 28977447 PMCID: PMC5886471 DOI: 10.1093/hmg/ddx293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 07/18/2017] [Accepted: 07/21/2017] [Indexed: 12/17/2022] Open
Abstract
Glycaemic traits such as fasting and post-challenge glucose and insulin measures, as well as glycated haemoglobin (HbA1c), are used to diagnose and monitor diabetes. These traits are risk factors for cardiovascular disease even below the diabetic threshold, and their study can additionally yield insights into the pathophysiology of type 2 diabetes. To date, a diverse set of genetic approaches have led to the discovery of over 97 loci influencing glycaemic traits. In this review, we will focus on recent advances in the genetic aetiology of glycaemic traits, and the resulting biological insights. We will provide a brief overview of results ranging from common, to low- and rare-frequency variant-trait association studies, studies leveraging the diversity across populations, and studies harnessing the power of genetic and genomic approaches to gain insights into the biological underpinnings of these traits.
Collapse
Affiliation(s)
- Eleanor Wheeler
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Gaëlle Marenne
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Inês Barroso
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
167
|
Su SC, Ho YC, Liu YF, Reiter RJ, Chou CH, Yeh CM, Lee HL, Chung WH, Hsieh MJ, Yang SF. Association of melatonin membrane receptor 1A/1B gene polymorphisms with the occurrence and metastasis of hepatocellular carcinoma. Oncotarget 2017; 8:85655-85669. [PMID: 29156748 PMCID: PMC5689638 DOI: 10.18632/oncotarget.21107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 09/04/2017] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent primary neoplasm of the liver, whose heterogeneous global incidence suggests the likely impact of genetic variations among individuals on the susceptibility to this disease. Increasing evidence indicates that melatonin exhibits oncostatic properties in many cancer types at least in part mediated by its membrane-bound receptors, melatonin receptor 1A (encoded by MTNR1A) and 1B (MTNR1B). In this study, the effect of melatonin receptor gene polymorphisms on the risk and progression of hepatic tumors was evaluated between 335 HCC patients and 1196 cancer-free subjects. We detected a significant association of MTNR1A single nucleotide polymorphism (SNP), rs6553010, with the elevated risk of HCC (AOR, 1.587; 95% CI, 1.053–2.389; p = 0.027) after being adjusted for two potential confounders, age and alcohol use. In addition, patients who carry at least one polymorphic allele (heterozygote or homozygote) of MTNR1A rs2119882 or rs2375801 were more prone to develop distant metastasis (OR, 5.202; 95% CI, 1.163–23.270; p = 0.031, and OR, 7.782; 95% CI, 1.015–59.663; p = 0.048, for rs2119882 and rs2375801, respectively). Further analyses revealed that rs2119882 is located on the consensus binding site of GATA2 transcription factor within the promoter region of MTNR1A gene, and that a correlation between the levels of GATA2 and melatonin receptor 1A was observed in the TCGA (The Cancer Genome Atlas) dataset. Moreover, individuals bearing a specific haplotype of four MTNR1B SNPs were more prone to develop HCC. In conclusion, our data suggest an association of melatonin receptor gene polymorphisms with the risk of HCC and hepatic cancer metastasis.
Collapse
Affiliation(s)
- Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Linkou and Keelung, Taiwan
| | - Yung-Chuan Ho
- School of Medical Applied Chemistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, College of Medicine Sciences and Technology, Chung Shan Medical University, Taichung, Taiwan
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Chia-Hsuan Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Ming Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Deptartment of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wen-Hung Chung
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Linkou and Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Ju Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
168
|
de Luis DA, Izaola O, Primo D, Aller R. Association of the rs10830963 polymorphism in melatonin receptor type 1B (MTNR1B) with metabolic response after weight loss secondary to a hypocaloric diet based in Mediterranean style. Clin Nutr 2017; 37:1563-1568. [PMID: 28869073 DOI: 10.1016/j.clnu.2017.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/02/2017] [Accepted: 08/14/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Some genetic variants within MTNR1B were related with fasting glucose levels or the increased prevalence of diabetes mellitus and obesity. The aims of the present investigation were to determine the influence of rs10830963 MTNR1B variant in relation to body weight loss, insulin resistance and adipokine levels in response to a hypocaloric diet with Mediterranean pattern. METHODS A Caucasian population of 80 obese patients was studied before and after 12 weeks on a hypocaloric diet. Body weight, fat mass, waist circumference, blood pressure, fasting blood glucose, C-reactive protein (CRP), insulin concentration, insulin resistance (HOMA-IR), lipoprotein levels and adipocytokines levels (leptin, adiponectin and resistin) were measured. Genotype of MTNR1B gene single nucleotide polymorphism (rs10830963) was evaluated. RESULTS In total, 44 patients (55%) had the genotype CC, 27 patients CG (33.8%) and 9 patients GG (11.2%). With the dietary intervention body mass index, weight, fat mass, systolic blood pressure, leptin levels and waist circumference decreased in both groups. There were no significant differences between gender groups on the reported effects in each genotype group. However, the improvement of anthropometric parameters was higher in subjects with CC genotype than (GC + GG) genotype. After dietary intervention and in males with CC genotype, insulin levels (-5.3 ± 4.8 UI/L vs 1.2 ± 4.1 UI/L; p < 0.05) and HOMA-IR (-1.4 ± 2.1 units vs 0.4 ± 2.0 units; p < 0.05) decreased. In the group of females with CC genotype, insulin levels (-3.5 ± 2.1 UI/L vs. -1.4 ± 2.2 UI/L: p < 0.05) and HOMA-IR (-1.4 ± 1.2 units vs. -0.1 ± 1.3 units: p < 0.05) decreased, too. However, these parameters remained unchanged in (GC + GG) group. Fasting glucose levels were higher in patients in (GC + GG). CONCLUSIONS This study showed the association of rs10830963 MTNR1B single nucleotide polymorphism with body weight loss and changes in fasting insulin levels and HOMA-IR in obese subjects.
Collapse
Affiliation(s)
- Daniel Antonio de Luis
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain.
| | - Olatz Izaola
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| | - David Primo
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| | - R Aller
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| |
Collapse
|
169
|
Yeh CM, Su SC, Lin CW, Yang WE, Chien MH, Reiter RJ, Yang SF. Melatonin as a potential inhibitory agent in head and neck cancer. Oncotarget 2017; 8:90545-90556. [PMID: 29163852 PMCID: PMC5685773 DOI: 10.18632/oncotarget.20079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/26/2017] [Indexed: 12/29/2022] Open
Abstract
Melatonin is a molecule secreted by the pineal gland; it is an important regulator of sleep and circadian rhythms. Through multiple interrelated mechanisms, melatonin exhibits various inhibitory properties at different stages of tumor progression. Many studies have explored the oncostatic effects of melatonin on hormone-dependent tumors. In this review, we highlight recent advances in understanding the effects of melatonin on the development of head and neck cancers, including molecular mechanisms identified through experimental and clinical observations. Because melatonin exerts a wide range of effects, melatonin may influence many mechanisms that influence the development of cancer. These include cell proliferation, apoptosis, angiogenesis, extracellular matrix remodeling through matrix metalloproteinases, and genetic polymorphism. Thus, the evidence discussed in this article will serve as a basis for basic and clinical research to promote the use of melatonin for understanding and controlling the development of head and neck cancers.
Collapse
Affiliation(s)
- Chia-Ming Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
170
|
Chung RH, Chiu YF, Hung YJ, Lee WJ, Wu KD, Chen HL, Lin MW, Chen YDI, Quertermous T, Hsiung CA. Genome-wide copy number variation analysis identified deletions in SFMBT1 associated with fasting plasma glucose in a Han Chinese population. BMC Genomics 2017; 18:591. [PMID: 28789618 PMCID: PMC5549306 DOI: 10.1186/s12864-017-3975-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 07/31/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Fasting glucose and fasting insulin are glycemic traits closely related to diabetes, and understanding the role of genetic factors in these traits can help reveal the etiology of type 2 diabetes. Although single nucleotide polymorphisms (SNPs) in several candidate genes have been found to be associated with fasting glucose and fasting insulin, copy number variations (CNVs), which have been reported to be associated with several complex traits, have not been reported for association with these two traits. We aimed to identify CNVs associated with fasting glucose and fasting insulin. RESULTS We conducted a genome-wide CNV association analysis for fasting plasma glucose (FPG) and fasting plasma insulin (FPI) using a family-based genome-wide association study sample from a Han Chinese population in Taiwan. A family-based CNV association test was developed in this study to identify common CNVs (i.e., CNVs with frequencies ≥ 5%), and a generalized estimating equation approach was used to test the associations between the traits and counts of global rare CNVs (i.e., CNVs with frequencies <5%). We found a significant genome-wide association for common deletions with a frequency of 5.2% in the Scm-like with four mbt domains 1 (SFMBT1) gene with FPG (association p-value = 2×10-4 and an adjusted p-value = 0.0478 for multiple testing). No significant association was observed between global rare CNVs and FPG or FPI. The deletions in 20 individuals with DNA samples available were successfully validated using PCR-based amplification. The association of the deletions in SFMBT1 with FPG was further evaluated using an independent population-based replication sample obtained from the Taiwan Biobank. An association p-value of 0.065, which was close to the significance level of 0.05, for FPG was obtained by testing 9 individuals with CNVs in the SFMBT1 gene region and 11,692 individuals with normal copies in the replication cohort. CONCLUSIONS Previous studies have found that SNPs in SFMBT1 are associated with blood pressure and serum urate concentration, suggesting that SFMBT1 may have functional implications in some metabolic-related traits.
Collapse
Affiliation(s)
- Ren-Hua Chung
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, No 35, Keyan Road, Zhunan, Miaoli, 350, Taiwan
| | - Yen-Feng Chiu
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, No 35, Keyan Road, Zhunan, Miaoli, 350, Taiwan
| | - Yi-Jen Hung
- Division of Endocrinology and Metabolism, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Social Work, Tunghai University, Taichung, Taiwan
| | - Kwan-Dun Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hui-Ling Chen
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, No 35, Keyan Road, Zhunan, Miaoli, 350, Taiwan
| | - Ming-Wei Lin
- Institute of Public Health, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Yii-Der I Chen
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine and Stanford Cardiovascular Institute, Falk Cardiovascular Research Center, Stanford University, Stanford, California, USA
| | - Chao A Hsiung
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, No 35, Keyan Road, Zhunan, Miaoli, 350, Taiwan.
| |
Collapse
|
171
|
Billings LK, Jablonski KA, Warner AS, Cheng YC, McAteer JB, Tipton L, Shuldiner AR, Ehrmann DA, Manning AK, Dabelea D, Franks PW, Kahn SE, Pollin TI, Knowler WC, Altshuler D, Florez JC, for the Diabetes Prevention Program Research Group . Variation in Maturity-Onset Diabetes of the Young Genes Influence Response to Interventions for Diabetes Prevention. J Clin Endocrinol Metab 2017; 102:2678-2689. [PMID: 28453780 PMCID: PMC5546852 DOI: 10.1210/jc.2016-3429] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/21/2017] [Indexed: 11/19/2022]
Abstract
Context Variation in genes that cause maturity-onset diabetes of the young (MODY) has been associated with diabetes incidence and glycemic traits. Objectives This study aimed to determine whether genetic variation in MODY genes leads to differential responses to insulin-sensitizing interventions. Design and Setting This was a secondary analysis of a multicenter, randomized clinical trial, the Diabetes Prevention Program (DPP), involving 27 US academic institutions. We genotyped 22 missense and 221 common variants in the MODY-causing genes in the participants in the DPP. Participants and Interventions The study included 2806 genotyped DPP participants randomized to receive intensive lifestyle intervention (n = 935), metformin (n = 927), or placebo (n = 944). Main Outcome Measures Association of MODY genetic variants with diabetes incidence at a median of 3 years and measures of 1-year β-cell function, insulinogenic index, and oral disposition index. Analyses were stratified by treatment group for significant single-nucleotide polymorphism × treatment interaction (Pint < 0.05). Sequence kernel association tests examined the association between an aggregate of rare missense variants and insulinogenic traits. Results After 1 year, the minor allele of rs3212185 (HNF4A) was associated with improved β-cell function in the metformin and lifestyle groups but not the placebo group; the minor allele of rs6719578 (NEUROD1) was associated with an increase in insulin secretion in the metformin group but not in the placebo and lifestyle groups. Conclusions These results provide evidence that genetic variation among MODY genes may influence response to insulin-sensitizing interventions.
Collapse
Affiliation(s)
- Liana K. Billings
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois 60201
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
| | | | - A. Sofia Warner
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Yu-Chien Cheng
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois 60201
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Jarred B. McAteer
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Laura Tipton
- Biostatistics Center, George Washington University, Rockville, Maryland 20852
| | - Alan R. Shuldiner
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - David A. Ehrmann
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Alisa K. Manning
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, Colorado 80045
| | - Paul W. Franks
- Department of Clinical Sciences, Genetic, and Molecular Epidemiology Unit, Lund University Diabetes Center, Skåne University Hospital Malmö, SE-205 02 Malmö, Sweden
| | - Steven E. Kahn
- Division of Metabolism, Endocrinology, and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98195
| | - Toni I. Pollin
- Departments of Medicine (Division of Endocrinology, Diabetes, and Nutrition) and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - William C. Knowler
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85014
| | - David Altshuler
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
- Vertex Pharmaceuticals, Boston, Massachusetts 02210
| | - Jose C. Florez
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - for the Diabetes Prevention Program Research Group
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois 60201
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
- Biostatistics Center, George Washington University, Rockville, Maryland 20852
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, Colorado 80045
- Department of Clinical Sciences, Genetic, and Molecular Epidemiology Unit, Lund University Diabetes Center, Skåne University Hospital Malmö, SE-205 02 Malmö, Sweden
- Division of Metabolism, Endocrinology, and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98195
- Departments of Medicine (Division of Endocrinology, Diabetes, and Nutrition) and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85014
- Vertex Pharmaceuticals, Boston, Massachusetts 02210
| |
Collapse
|
172
|
Song J, Whitcomb DJ, Kim BC. The role of melatonin in the onset and progression of type 3 diabetes. Mol Brain 2017; 10:35. [PMID: 28764741 PMCID: PMC5539639 DOI: 10.1186/s13041-017-0315-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is defined by the excessive accumulation of toxic peptides, such as beta amyloid (Aβ) plaques and intracellular neurofibrillary tangles (NFT). The risk factors associated with AD include genetic mutations, aging, insulin resistance, and oxidative stress. To date, several studies that have demonstrated an association between AD and diabetes have revealed that the common risk factors include insulin resistance, sleep disturbances, blood brain barrier (BBB) disruption, and altered glucose homeostasis. Many researchers have discovered that there are mechanisms common to both diabetes and AD. AD that results from insulin resistance in the brain is termed “type 3 diabetes”. Melatonin synthesized by the pineal gland is known to contribute to circadian rhythms, insulin resistance, protection of the BBB, and cell survival mechanisms. Here, we review the relationship between melatonin and type 3 diabetes, and suggest that melatonin might regulate the risk factors for type 3 diabetes. We suggest that melatonin is crucial for attenuating the onset of type 3 diabetes by intervening in Aβ accumulation, insulin resistance, glucose metabolism, and BBB permeability.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, 61469, South Korea
| | - Daniel J Whitcomb
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Healthy Sciences, University of Bristol, Whitson street, Bristol, BS1 3NY, UK
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| |
Collapse
|
173
|
Mazzoccoli G, Dagostino MP, Paroni G, Seripa D, Ciccone F, Addante F, Favuzzi G, Grandone E, Avola R, Mazza T, Fusilli C, Greco A, De Cosmo S. Analysis of MTNR1B gene polymorphisms in relationship with IRS2 gene variants, epicardial fat thickness, glucose homeostasis and cognitive performance in the elderly. Chronobiol Int 2017; 34:1083-1093. [PMID: 28708046 DOI: 10.1080/07420528.2017.1340894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ABSTARCT Genome-wide association studies pinpointed common variants in or near the MTNR1B gene encoding MT2 melatonin receptor to be strongly associated with fasting glucose levels. IRS2 gene polymorphisms impact insulin resistance and epicardial fat (EF) thickness, which in turn is correlated with visceral adiposity, cognitive ability and risk for metabolic plus cardiovascular disease. We aimed to discover the interactions between MTNR1B and IRS2 gene polymorphisms, insulin sensitivity, EF thickness and cognitive performance in the elderly. In 60 subjects aged 60 years and older, we evaluated five single nucleotide polymorphisms (SNPs) within the MTNR1B locus (rs10830962, rs4753426, rs12804291, rs10830963, rs3781638), the Gly1057Asp variant of IRS2 gene (rs1805097), biochemical parameters, cognitive performance by the Mini Mental State Examination (MMSE) and EF thickness by transthoracic echocardiography. We found that MTNR1B and IRS2 gene variants impacted EF thickness, lipid profile and glucose homeostasis. IRS2 but not MTNR1B variants impacted MMSE scores. In conclusion, MTNR1B SNPs interact with IRS2 gene variant, correlate with the amount of epicardial adipose tissue and impact glucose homeostasis and lipid profile influencing cardiometabolic risk.
Collapse
Affiliation(s)
- Gianluigi Mazzoccoli
- a Department of Medical Sciences , Division of Internal Medicine and Chronobiology Unit
| | - Mariangela Pia Dagostino
- b Department of Medical Sciences , Geriatrics Unit and Gerontology-Geriatrics Research Laboratory, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Giulia Paroni
- b Department of Medical Sciences , Geriatrics Unit and Gerontology-Geriatrics Research Laboratory, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Davide Seripa
- b Department of Medical Sciences , Geriatrics Unit and Gerontology-Geriatrics Research Laboratory, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Filomena Ciccone
- b Department of Medical Sciences , Geriatrics Unit and Gerontology-Geriatrics Research Laboratory, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Filomena Addante
- b Department of Medical Sciences , Geriatrics Unit and Gerontology-Geriatrics Research Laboratory, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Giovanni Favuzzi
- c Research Laboratory of Thrombosis and Hemostasis, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", Opera di Padre Pio da Pietrelcina , San Giovanni Rotondo , Italy
| | - Elvira Grandone
- c Research Laboratory of Thrombosis and Hemostasis, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", Opera di Padre Pio da Pietrelcina , San Giovanni Rotondo , Italy
| | - Roberto Avola
- d Department of Biomedical and Biotechnological Sciences , University of Catania , Catania , Italy
| | - Tommaso Mazza
- e Bioinformatics Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", Opera di Padre Pio da Pietrelcina , San Giovanni Rotondo , Italy
| | - Caterina Fusilli
- e Bioinformatics Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", Opera di Padre Pio da Pietrelcina , San Giovanni Rotondo , Italy
| | - Antonio Greco
- b Department of Medical Sciences , Geriatrics Unit and Gerontology-Geriatrics Research Laboratory, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza" , San Giovanni Rotondo , Italy
| | - Salvatore De Cosmo
- a Department of Medical Sciences , Division of Internal Medicine and Chronobiology Unit
| |
Collapse
|
174
|
Meta-analyses of the association of G6PC2 allele variants with elevated fasting glucose and type 2 diabetes. PLoS One 2017; 12:e0181232. [PMID: 28704540 PMCID: PMC5509327 DOI: 10.1371/journal.pone.0181232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 06/28/2017] [Indexed: 12/19/2022] Open
Abstract
Objective To collectively evaluate the association of glucose-6-phosphatase catalytic unit 2 (G6PC2) allele variants with elevated fasting glucose (FG) and type 2 diabetes (T2D). Design Meta-analysis Data sources PubMed, Web of Knowledge and Embase databases. Study selection Full text articles of studies that identified an association of G6PC2 with T2D and elevated FG. Patient involvement There was no T2D patient involvement in the analyses on the association of FG with G6PC2, there were T2D patients and non-diabetes patient involvement in the analyses on the association of T2D with G6PC2. Statistical analysis Random-effects meta-analyses were used to calculate the pool effect sizes. I2 metric and H2 tests were used to calculate the heterogeneity. Begg's funnel plot and Egger’s linear regression test were done to assess publication bias. Results Of the 423 studies identified, 21 were eligible and included. Data on three loci (rs560887, rs16856187 and rs573225) were available. The G allele at rs560887 in three ethnicities, the C allele at rs16856187 and the A allele at rs573225 all had a positive association with elevated FG. Per increment of G allele at rs560887 and A allele at rs573225 resulted in a FG 0.070 mmol/l and 0.075 mmol/l higher (ß (95% CI) = 0.070 (0.060, 0.079), p = 4.635e-50 and 0.075 (0.065, 0.085), p = 5.856e-48, respectively). With regard to the relationship of rs16856187 and FG, an increase of 0.152 (95% CI: 0.034–0.270; p = 0.011) and 0.317 (95% CI: 0.193–0.442, p = 6.046e-07) was found in the standardized mean difference (SMD) of FG for the AC and CC genotypes, respectively, when compared with the AA reference genotype. However, the G-allele of rs560887 in Caucasians under the additive model and the C-allele of rs16856187 under the allele and dominant models were associated with a decreased risk of T2D (OR (95% CI) = 0.964 (0.947, 0.981), p = 0.570e-4; OR (95% CI) = 0.892 (0.832, 0.956), p = 0.001; and OR (95% CI) = 0.923(0.892, 0.955), p = 5.301e-6, respectively). Conclusions Our meta-analyses demonstrate that all three allele variants of G6PC2 (rs560887, rs16856187 and rs573225) are associated with elevated FG, with two variants (rs560887 in the Caucasians subgroup and rs16856187 under the allele and dominant model) being associated with T2D as well. Further studies utilizing larger sample sizes and different ethnic populations are needed to extend and confirm these findings.
Collapse
|
175
|
Nduhirabandi F, Huisamen B, Strijdom H, Lochner A. Role of melatonin in glucose uptake by cardiomyocytes from insulin-resistant Wistar rats. Cardiovasc J Afr 2017; 28:362-369. [PMID: 28556852 PMCID: PMC5885054 DOI: 10.5830/cvja-2017-018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 04/04/2017] [Indexed: 12/31/2022] Open
Abstract
Aim Melatonin supplementation reduces insulin resistance and protects the heart in obese rats. However, its role in myocardial glucose uptake remains unknown. This study investigated the effect of short-term melatonin treatment on glucose uptake by cardiomyocytes isolated from obese and insulin-resistant rats. Methods Cardiomyocytes were isolated from obese rats fed a high-calorie diet for 16 to 23 weeks, their age-matched controls, as well as young control rats aged four to eight weeks. After incubation with melatonin with or without insulin, glucose uptake was initiated by the addition of 2-deoxy-D-[3H] glucose and measured after 30 minutes. Additional control and obese rats received melatonin in the drinking water (4 mg/kg/day) for the last six weeks of feeding (20 weeks) and glucose uptake was determined in isolated cardiomyocytes after incubation with insulin. Intraperitoneal glucose tolerance and biometric parameters were also measured. Results Obese rats (fed for more than 20 weeks) developed glucose intolerance. Cardiomyocytes isolated from these obese rats had a reduced response to insulin-stimulated glucose uptake (ISGU) (p < 0.05). Melatonin administration in vitro had no effect on glucose uptake per se. However, it increased ISGU by cardiomyocytes from the young rats (p < 0.05), while having no effect on ISGU by cardiomyocytes from the older control and obese groups. Melatonin in vivo had no significant effect on glucose tolerance, but it increased basal (p < 0.05) and ISGU by cardiomyocytes from the obese rats (50.1 ± 1.7 vs 32.1 ± 5.1 pmol/mg protein/30 min, p < 0.01). Conclusion These data suggest that short-term melatonin treatment in vivo but not in vitro improved glucose uptake and insulin responsiveness of cardiomyocytes in obesity and insulin-resistance states.
Collapse
Affiliation(s)
- Frederic Nduhirabandi
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa.
| | - Barbara Huisamen
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa; Biotechnology, Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Hans Strijdom
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Amanda Lochner
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
176
|
Mulder H. Melatonin signalling and type 2 diabetes risk: too little, too much or just right? Diabetologia 2017; 60:826-829. [PMID: 28303303 DOI: 10.1007/s00125-017-4249-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/06/2017] [Indexed: 01/29/2023]
Abstract
Of the associations of genetic variants with type 2 diabetes, the one of an SNP in an intron of the gene encoding the melatonin receptor 1B (MTNR1B) has been remarkably robust. Work from our group and others has provided support for a model where carriers of this risk G allele exhibit increased MTNR1B expression in islets of Langerhans. Most published studies to date favour that melatonin's action on the beta cell is inhibition of insulin secretion. Hence, our model proposes that this inhibitory effect of melatonin is exaggerated in carriers of the MTNR1B risk G allele. This would explain why this genetic association causes reduced insulin secretion and greater risk of future type 2 diabetes, as has been observed in numerous studies. Concurrently, another body of work has shown that rare MTNR1B alleles, which could perturb receptor function, also associate with type 2 diabetes. In this commentary, it is suggested that such apparently conflicting observations can be reconciled by the fact that non-coding (intronic; frequent) and coding (exonic; rare) alleles of MTNR1B give rise to different phenotypes. Thus, altered gene transcription may explain why SNPs, which do not alter coding sequences, exhibit cell-specific effects. In contrast, SNPs that change protein sequences are more likely to exert generalised effects since an altered protein will appear in all cells expressing the gene.
Collapse
Affiliation(s)
- Hindrik Mulder
- Unit of Molecular Metabolism, Lund University Diabetes Centre, Jan Waldenströms gata 35, SE-205 02, Malmö, Sweden.
| |
Collapse
|
177
|
Jiang P, Turek FW. Timing of meals: when is as critical as what and how much. Am J Physiol Endocrinol Metab 2017; 312:E369-E380. [PMID: 28143856 PMCID: PMC6105931 DOI: 10.1152/ajpendo.00295.2016] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 02/08/2023]
Abstract
Over the past decade, a large body of literature has demonstrated that disruptions of the endogenous circadian clock, whether environmental or genetic, lead to metabolic dysfunctions that are associated with obesity, diabetes, and other metabolic disorders. The phrase, "It is not only what you eat and how much you eat, but also when you eat" sends a simple message about circadian timing and body weight regulation. Communicating this message to clinicians and patients, while also elucidating the neuroendocrine, molecular, and genetic mechanisms underlying this phrase is essential to embrace the growing knowledge of the circadian impact on metabolism as a part of healthy life style as well as to incorporate it into clinical practice for improvement of overall human health. In this review, we discuss findings from animal models, as well as epidemiological and clinical studies in humans, which collectively promote the awareness of the role of circadian clock in metabolic functions and dysfunctions.
Collapse
Affiliation(s)
- Peng Jiang
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - Fred W Turek
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, Illinois
| |
Collapse
|
178
|
Lopez-Minguez J, Saxena R, Bandín C, Scheer FA, Garaulet M. Late dinner impairs glucose tolerance in MTNR1B risk allele carriers: A randomized, cross-over study. Clin Nutr 2017; 37:1133-1140. [PMID: 28455106 DOI: 10.1016/j.clnu.2017.04.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Late-night dinner eating is associated with increased risk for type-2 diabetes. The underlying mechanism is unclear. One explanatory hypothesis is that the concurrence of elevated circulating melatonin and high glucose concentrations (characterizing late eating) leads to impaired glucose tolerance. However, to date no study has tested the influence of physiological melatonin concentrations on glucose-tolerance. The discovery of melatonin receptor MTNR1B as a diabetes risk gene provides evidence for a role of physiological levels of melatonin in glucose control. The aim of our study was to test the hypothesis that elevated endogenous melatonin concentrations worsen glucose control when eating late. Registered under ClinicalTrials.gov Identifier no. NCT03003936. METHODS We performed a randomized, cross-over trial to compare glucose tolerance in the presence (late dinner) or absence (early dinner) of elevated physiological melatonin concentrations and we compared the results between homozygous carriers and non-carriers of the MTNR1B risk allele. RESULTS The concurrence of meal timing with elevated endogenous melatonin concentrations resulted in impaired glucose tolerance. This effect was stronger in MTNR1B risk-carriers than in non-carriers. Furthermore, eating late significantly impaired glucose tolerance only in risk-carriers and not in the non-risk carriers. CONCLUSIONS The interaction of dinner timing with MTNR1B supports a causal role of endogenous melatonin in the impairment of glucose tolerance. These results suggest that moving the dinner to an earlier time may result in better glucose tolerance specially in MTNR1B carriers. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT03003936.
Collapse
Affiliation(s)
- Jesus Lopez-Minguez
- Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain
| | - Richa Saxena
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, USA; Broad Institute, Cambridge, MA, USA
| | - Cristina Bandín
- Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain
| | - Frank A Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| | - Marta Garaulet
- Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain.
| |
Collapse
|
179
|
Beaumont RN, Horikoshi M, McCarthy MI, Freathy RM. How Can Genetic Studies Help Us to Understand Links Between Birth Weight and Type 2 Diabetes? Curr Diab Rep 2017; 17:22. [PMID: 28293907 PMCID: PMC5350261 DOI: 10.1007/s11892-017-0852-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW In observational epidemiology, both low and high birth weights are associated with later type 2 diabetes. The mechanisms underlying the associations are poorly understood. We review evidence for the roles of genetic and non-genetic factors linking both sides of the birth weight distribution to risk of type 2 diabetes, focusing on contributions made by the most recent genome-wide association studies (GWAS) of birth weight. RECENT FINDINGS There are now nine genetic loci robustly implicated in both fetal growth and type 2 diabetes. At many of these, the same alleles are associated both with a higher risk of type 2 diabetes and a lower birth weight. This supports the Fetal Insulin Hypothesis and reflects a general pattern for type 2 diabetes susceptibility alleles: genome-wide, there is an inverse genetic correlation with birth weight, and initial estimates suggest genetic factors explain a large part of the covariance between the two traits. However, the associations at individual loci show heterogeneity; some fetal risk alleles are associated with higher birth weight. For most of these, the association reflects their correlation with the maternal risk allele which raises maternal glucose, thus increasing fetal insulin-mediated growth. GWAS have improved our understanding of the mechanisms underlying associations between type 2 diabetes and birth weight but questions remain about the relative importance of genetic versus non-genetic factors and of maternal versus fetal genotypes. To answer these questions, future work will require well-powered analyses of parents and offspring.
Collapse
Affiliation(s)
- Robin N. Beaumont
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, RILD Building, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW UK
| | - Momoko Horikoshi
- grid.4991.5Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Dr., Oxford, OX3 7BN UK
- grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Mark I. McCarthy
- grid.4991.5Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Dr., Oxford, OX3 7BN UK
- grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- grid.415719.fOxford National Institute for Health Research (NIHR) Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7IL UK
| | - Rachel M. Freathy
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, RILD Building, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW UK
- grid.5337.2Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| |
Collapse
|
180
|
Boortz KA, Syring KE, Pound LD, Mo H, Bastarache L, Oeser JK, McGuinness OP, Denny JC, O’Brien RM. Effects of G6pc2 deletion on body weight and cholesterol in mice. J Mol Endocrinol 2017; 58:127-139. [PMID: 28122818 PMCID: PMC5380368 DOI: 10.1530/jme-16-0202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 11/08/2022]
Abstract
Genome-wide association study (GWAS) data have linked the G6PC2 gene to variations in fasting blood glucose (FBG). G6PC2 encodes an islet-specific glucose-6-phosphatase catalytic subunit that forms a substrate cycle with the beta cell glucose sensor glucokinase. This cycle modulates the glucose sensitivity of insulin secretion and hence FBG. GWAS data have not linked G6PC2 to variations in body weight but we previously reported that female C57BL/6J G6pc2-knockout (KO) mice were lighter than wild-type littermates on both a chow and high-fat diet. The purpose of this study was to compare the effects of G6pc2 deletion on FBG and body weight in both chow-fed and high-fat-fed mice on two other genetic backgrounds. FBG was reduced in G6pc2 KO mice largely independent of gender, genetic background or diet. In contrast, the effect of G6pc2 deletion on body weight was markedly influenced by these variables. Deletion of G6pc2 conferred a marked protection against diet-induced obesity in male mixed genetic background mice, whereas in 129SvEv mice deletion of G6pc2 had no effect on body weight. G6pc2 deletion also reduced plasma cholesterol levels in a manner dependent on gender, genetic background and diet. An association between G6PC2 and plasma cholesterol was also observed in humans through electronic health record-derived phenotype analyses. These observations suggest that the action of G6PC2 on FBG is largely independent of the influences of environment, modifier genes or epigenetic events, whereas the action of G6PC2 on body weight and cholesterol are influenced by unknown variables.
Collapse
Affiliation(s)
- Kayla A. Boortz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kristen E. Syring
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Lynley D. Pound
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Huan Mo
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - James K. Oeser
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Joshua C. Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Richard M. O’Brien
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
181
|
Troponin T levels associated with genetic variants in NOTCH2 and MTNR1B in women with psychosis. Psychiatry Res 2017; 250:217-220. [PMID: 28167435 DOI: 10.1016/j.psychres.2017.01.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/27/2016] [Accepted: 01/13/2017] [Indexed: 01/13/2023]
Abstract
Psychosis patients have increased prevalence of metabolic disorders, which increase the risk for cardiovascular disease. Elevated troponin T level is an early biomarker of cardiovascular damage. We tested for association between troponin T levels and genetic risk variants of elevated blood glucose level in psychosis. Glucose and troponin T levels correlated positively. MTNR1B rs10830963 and NOTCH2 rs10923931 associated with troponin T levels in women, adjusted for glucose levels. These findings may indicate metabolic genetic influences on troponin T levels among women with psychosis.
Collapse
|
182
|
Yap TWC, Leow AHR, Azmi AN, Callahan DL, Perez-Perez GI, Loke MF, Goh KL, Vadivelu J. Global Fecal and Plasma Metabolic Dynamics Related to Helicobacter pylori Eradication. Front Microbiol 2017; 8:536. [PMID: 28424674 PMCID: PMC5371670 DOI: 10.3389/fmicb.2017.00536] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/14/2017] [Indexed: 12/20/2022] Open
Abstract
Background:Helicobacter pylori colonizes the gastric mucosa of more than half of the world's population. There is increasing evidence H. pylori protects against the development of obesity and childhood asthma/allergies in which the development of these diseases coincide with transient dysbiosis. However, the mechanism underlying the association of H. pylori eradication with human metabolic and immunological disorders is not well-established. In this study, we aimed to investigate the local and systemic effects of H. pylori eradication through untargeted fecal lipidomics and plasma metabolomics approaches by liquid chromatography mass spectrometry (LC-MS). Results: Our study revealed that eradication of H. pylori eradication (i.e., loss of H. pylori and/or H. pylori eradication therapy) changed many global metabolite/lipid features, with the majority being down-regulated. Our findings primarily show that H. pylori eradication affects the host energy and lipid metabolism which may eventually lead to the development of metabolic disorders. Conclusion: These predictive metabolic signatures of metabolic and immunological disorders following H. pylori eradication can provide insights into dynamic local and systemic metabolism related to H. pylori eradication in modulating human health.
Collapse
Affiliation(s)
- Theresa Wan-Chen Yap
- Department of Medical Microbiology, Faculty of Medicine, University of MalayaKuala Lumpur, Malaysia
| | - Alex Hwong-Ruey Leow
- Department of Medicine, Faculty of Medicine, University of MalayaKuala Lumpur, Malaysia
| | - Ahmad Najib Azmi
- Department of Medicine, Faculty of Medicine, University of MalayaKuala Lumpur, Malaysia.,Faculty of Medicine and Health Sciences, Universiti Sains Islam MalaysiaKuala Lumpur, Malaysia
| | - Damien L Callahan
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Deakin UniversityGeelong, VIC, Australia
| | - Guillermo I Perez-Perez
- Department of Medicine, New York University School of MedicineNew York, NY, USA.,Department of Microbiology, New York University School of MedicineNew York, NY, USA
| | - Mun-Fai Loke
- Department of Medical Microbiology, Faculty of Medicine, University of MalayaKuala Lumpur, Malaysia.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Khean-Lee Goh
- Department of Medicine, Faculty of Medicine, University of MalayaKuala Lumpur, Malaysia
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of MalayaKuala Lumpur, Malaysia
| |
Collapse
|
183
|
Adhikary N, Shrestha SL, Sun JZ. Metabolic disturbances: role of the circadian timing system and sleep. Diabetol Int 2017; 8:14-22. [PMID: 30603302 PMCID: PMC6224937 DOI: 10.1007/s13340-016-0279-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/28/2016] [Indexed: 12/21/2022]
Abstract
The incidence of metabolic disorders such as obesity and diabetes is on the rise, and food quality is not alone to blame. Sleep disturbances, altered feeding time and circadian disruption are linked to metabolic disturbances in many clinical research studies and cross-sectional analyses. This review tried to summarize the role of the circadian timing system and sleep on energy and metabolic homeostasis. We also tried to explain the molecular and endocrine mechanisms behind circadian misalignment and sleep disorders that lead to metabolic disorders.
Collapse
Affiliation(s)
- Navin Adhikary
- Department of Endocrinology, Zhongnan Hospital, Wuhan University, Wuhan, 430071 China
| | - Santosh Lal Shrestha
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, 430060 China
| | - Jia Zhong Sun
- Department of Endocrinology, Zhongnan Hospital, Wuhan University, Wuhan, 430071 China
| |
Collapse
|
184
|
Al-Daghri NM, Pontremoli C, Cagliani R, Forni D, Alokail MS, Al-Attas OS, Sabico S, Riva S, Clerici M, Sironi M. Susceptibility to type 2 diabetes may be modulated by haplotypes in G6PC2, a target of positive selection. BMC Evol Biol 2017; 17:43. [PMID: 28173748 PMCID: PMC5297017 DOI: 10.1186/s12862-017-0897-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/26/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The endoplasmic reticulum enzyme glucose-6-phosphatase catalyzes the common terminal reaction in the gluconeogenic/glycogenolytic pathways and plays a central role in glucose homeostasis. In most mammals, different G6PC subunits are encoded by three paralogous genes (G6PC, G6PC2, and G6PC3). Mutations in G6PC and G6PC3 are responsible for human mendelian diseases, whereas variants in G6PC2 are associated with fasting glucose (FG) levels. RESULTS We analyzed the evolutionary history of G6Pase genes. Results indicated that the three paralogs originated during early vertebrate evolution and that negative selection was the major force shaping diversity at these genes in mammals. Nonetheless, site-wise estimation of evolutionary rates at corresponding sites revealed weak correlations, suggesting that mammalian G6Pases have evolved different structural features over time. We also detected pervasive positive selection at mammalian G6PC2. Most selected residues localize in the C-terminal protein region, where several human variants associated with FG levels also map. This region was re-sequenced in ~560 subjects from Saudi Arabia, 185 of whom suffering from type 2 diabetes (T2D). The frequency of rare missense and nonsense variants was not significantly different in T2D and controls. Association analysis with two common missense variants (V219L and S342C) revealed a weak but significant association for both SNPs when analyses were conditioned on rs560887, previously identified in a GWAS for FG. Two haplotypes were significantly associated with T2D with an opposite effect direction. CONCLUSIONS We detected pervasive positive selection at mammalian G6PC2 genes and we suggest that distinct haplotypes at the G6PC2 locus modulate susceptibility to T2D.
Collapse
Affiliation(s)
- Nasser M Al-Daghri
- Biomarker research program, Biochemistry Department, College of Science, King Saud Universiy, Riyadh, 11451, Kingdom of Saudi Arabia.,Prince Mutaib Chair for Biomarkers of Osteoporosis Research, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | | | - Rachele Cagliani
- Scientific Institute IRCCS E.MEDEA, Bosisio Parini, 23842, Italy
| | - Diego Forni
- Scientific Institute IRCCS E.MEDEA, Bosisio Parini, 23842, Italy
| | - Majed S Alokail
- Biomarker research program, Biochemistry Department, College of Science, King Saud Universiy, Riyadh, 11451, Kingdom of Saudi Arabia.,Prince Mutaib Chair for Biomarkers of Osteoporosis Research, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Omar S Al-Attas
- Biomarker research program, Biochemistry Department, College of Science, King Saud Universiy, Riyadh, 11451, Kingdom of Saudi Arabia.,Prince Mutaib Chair for Biomarkers of Osteoporosis Research, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Shaun Sabico
- Biomarker research program, Biochemistry Department, College of Science, King Saud Universiy, Riyadh, 11451, Kingdom of Saudi Arabia.,Prince Mutaib Chair for Biomarkers of Osteoporosis Research, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Stefania Riva
- Scientific Institute IRCCS E.MEDEA, Bosisio Parini, 23842, Italy
| | - Mario Clerici
- Department of Physiopathology and Transplantation, University of Milan, via F.lli Cervi 93, Segrate, 20090, Milan, Italy. .,Don Gnocchi Foundation, ONLUS, Milan, 20148, Italy.
| | - Manuela Sironi
- Scientific Institute IRCCS E.MEDEA, Bosisio Parini, 23842, Italy
| |
Collapse
|
185
|
McHill AW, Wright KP. Role of sleep and circadian disruption on energy expenditure and in metabolic predisposition to human obesity and metabolic disease. Obes Rev 2017; 18 Suppl 1:15-24. [PMID: 28164449 DOI: 10.1111/obr.12503] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022]
Abstract
Weight gain, obesity and diabetes have reached alarming levels in the developed world. Traditional risk factors such as over-eating, poor nutritional choices and lack of exercise cannot fully account for the high prevalence of metabolic disease. This review paper examines the scientific evidence on two novel risk factors that contribute to dys-regulated metabolic physiology: sleep disruption and circadian misalignment. Specifically, fundamental relationships between energy metabolism and sleep and circadian rhythms and the impact of sleep and circadian disruption on metabolic physiology are examined. Millions of individuals worldwide do not obtain sufficient sleep for healthy metabolic function, and many participate in shift work and social activities at times when the internal physiological clock is promoting sleep. These behaviours predispose an individual for poor metabolic health by promoting excess caloric intake in response to reduced sleep, food intake at internal biological times when metabolic physiology is not prepared, decreased energy expenditure when wakefulness and sleep are initiated at incorrect internal biological times, and disrupted glucose metabolism during short sleep and circadian misalignment. In addition to the traditional risk factors of poor diet and exercise, disturbed sleep and circadian rhythms represent modifiable risk factors for prevention and treatment of metabolic disease and for promotion of healthy metabolism.
Collapse
Affiliation(s)
- A W McHill
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - K P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
186
|
Laakso M, Kuusisto J, Stančáková A, Kuulasmaa T, Pajukanta P, Lusis AJ, Collins FS, Mohlke KL, Boehnke M. The Metabolic Syndrome in Men study: a resource for studies of metabolic and cardiovascular diseases. J Lipid Res 2017; 58:481-493. [PMID: 28119442 DOI: 10.1194/jlr.o072629] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/15/2017] [Indexed: 12/30/2022] Open
Abstract
The Metabolic Syndrome in Men (METSIM) study is a population-based study including 10,197 Finnish men examined in 2005-2010. The aim of the study is to investigate nongenetic and genetic factors associated with the risk of T2D and CVD, and with cardiovascular risk factors. The protocol includes a detailed phenotyping of the participants, an oral glucose tolerance test, fasting laboratory measurements including proton NMR measurements, mass spectometry metabolomics, adipose tissue biopsies from 1,400 participants, and a stool sample. In our ongoing follow-up study, we have, to date, reexamined 6,496 participants. Extensive genotyping and exome sequencing have been performed for essentially all METSIM participants, and >2,000 METSIM participants have been whole-genome sequenced. We have identified several nongenetic markers associated with the development of diabetes and cardiovascular events, and participated in several genetic association studies to identify gene variants associated with diabetes, hyperglycemia, and cardiovascular risk factors. The generation of a phenotype and genotype resource in the METSIM study allows us to proceed toward a "systems genetics" approach, which includes statistical methods to quantitate and integrate intermediate phenotypes, such as transcript, protein, or metabolite levels, to provide a global view of the molecular architecture of complex traits.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland .,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Päivi Pajukanta
- Departments of Human Genetics David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Aldons J Lusis
- Departments of Human Genetics David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
187
|
Rosta K, Al-Aissa Z, Hadarits O, Harreiter J, Nádasdi Á, Kelemen F, Bancher-Todesca D, Komlósi Z, Németh L, Rigó J, Sziller I, Somogyi A, Kautzky-Willer A, Firneisz G. Association Study with 77 SNPs Confirms the Robust Role for the rs10830963/G of MTNR1B Variant and Identifies Two Novel Associations in Gestational Diabetes Mellitus Development. PLoS One 2017; 12:e0169781. [PMID: 28072873 PMCID: PMC5224877 DOI: 10.1371/journal.pone.0169781] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/21/2016] [Indexed: 12/31/2022] Open
Abstract
CONTEXT Genetic variation in human maternal DNA contributes to the susceptibility for development of gestational diabetes mellitus (GDM). OBJECTIVE We assessed 77 maternal single nucleotide gene polymorphisms (SNPs) for associations with GDM or plasma glucose levels at OGTT in pregnancy. METHODS 960 pregnant women (after dropouts 820: case/control: m99'WHO: 303/517, IADPSG: 287/533) were enrolled in two countries into this case-control study. After genomic DNA isolation the 820 samples were collected in a GDM biobank and assessed using KASP (LGC Genomics) genotyping assay. Logistic regression risk models were used to calculate ORs according to IADPSG/m'99WHO criteria based on standard OGTT values. RESULTS The most important risk alleles associated with GDM were rs10830963/G of MTNR1B (OR = 1.84/1.64 [IADPSG/m'99WHO], p = 0.0007/0.006), rs7754840/C (OR = 1.51/NS, p = 0.016) of CDKAL1 and rs1799884/T (OR = 1.4/1.56, p = 0.04/0.006) of GCK. The rs13266634/T (SLC30A8, OR = 0.74/0.71, p = 0.05/0.02) and rs7578326/G (LOC646736/IRS1, OR = 0.62/0.60, p = 0.001/0.006) variants were associated with lower risk to develop GDM. Carrying a minor allele of rs10830963 (MTNR1B); rs7903146 (TCF7L2); rs1799884 (GCK) SNPs were associated with increased plasma glucose levels at routine OGTT. CONCLUSIONS We confirmed the robust association of MTNR1B rs10830963/G variant with GDM binary and glycemic traits in this Caucasian case-control study. As novel associations we report the minor, G allele of the rs7578326 SNP in the LOC646736/IRS1 region as a significant and the rs13266634/T SNP (SLC30A8) as a suggestive protective variant against GDM development. Genetic susceptibility appears to be more preponderant in individuals who meet both the modified 99'WHO and the IADPSG GDM diagnostic criteria.
Collapse
Affiliation(s)
- Klara Rosta
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
- 1 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Zahra Al-Aissa
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Orsolya Hadarits
- 1 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Jürgen Harreiter
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ákos Nádasdi
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Fanni Kelemen
- University of Szeged, Faculty of Medicine, Szeged, Hungary
| | | | - Zsolt Komlósi
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - László Németh
- Department of Probability Theory and Statistics, Eötvös Loránd University, Budapest, Hungary
| | - János Rigó
- 1 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - István Sziller
- Department of Obstetrics and Gynecology, Szent Imre Teaching Hospital, Budapest, Hungary
| | - Anikó Somogyi
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Alexandra Kautzky-Willer
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gábor Firneisz
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Academy of Sciences - Semmelweis University, Molecular Medicine Research Group, Budapest, Hungary
- * E-mail:
| |
Collapse
|
188
|
Type 2 Diabetes Susceptibility in the Greek-Cypriot Population: Replication of Associations with TCF7L2, FTO, HHEX, SLC30A8 and IGF2BP2 Polymorphisms. Genes (Basel) 2017; 8:genes8010016. [PMID: 28067832 PMCID: PMC5295011 DOI: 10.3390/genes8010016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/13/2016] [Accepted: 12/30/2016] [Indexed: 01/17/2023] Open
Abstract
Type 2 diabetes (T2D) has been the subject of numerous genetic studies in recent years which revealed associations of the disease with a large number of susceptibility loci. We hereby initiate the evaluation of T2D susceptibility loci in the Greek-Cypriot population by performing a replication case-control study. One thousand and eighteen individuals (528 T2D patients, 490 controls) were genotyped at 21 T2D susceptibility loci, using the allelic discrimination method. Statistically significant associations of T2D with five of the tested single nucleotide polymorphisms (SNPs) (TCF7L2 rs7901695, FTO rs8050136, HHEX rs5015480, SLC30A8 rs13266634 and IGF2BP2 rs4402960) were observed in this study population. Furthermore, 14 of the tested SNPs had odds ratios (ORs) in the same direction as the previously published studies, suggesting that these variants can potentially be used in the Greek-Cypriot population for predictive testing of T2D. In conclusion, our findings expand the genetic assessment of T2D susceptibility loci and reconfirm five of the worldwide established loci in a distinct, relatively small, newly investigated population.
Collapse
|
189
|
Gonzalez-Franquesa A, Patti ME. Insulin Resistance and Mitochondrial Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:465-520. [DOI: 10.1007/978-3-319-55330-6_25] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
190
|
Gesmundo I, Villanova T, Banfi D, Gamba G, Granata R. Role of Melatonin, Galanin, and RFamide Neuropeptides QRFP26 and QRFP43 in the Neuroendocrine Control of Pancreatic β-Cell Function. Front Endocrinol (Lausanne) 2017; 8:143. [PMID: 28729853 PMCID: PMC5499649 DOI: 10.3389/fendo.2017.00143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/06/2017] [Indexed: 12/12/2022] Open
Abstract
Glucose homeostasis is finely regulated by a number of hormones and peptides released mainly from the brain, gastrointestinal tract, and muscle, regulating pancreatic secretion through cellular receptors and their signal transduction cascades. The endocrine function of the pancreas is controlled by islets within the exocrine pancreatic tissue that release hormones like insulin, glucagon, somatostatin, pancreatic polypeptide, and ghrelin. Moreover, both exocrine and endocrine pancreatic functions are regulated by a variety of hormonal and neural mechanisms, such as ghrelin, glucagon-like peptide, glucose-dependent insulinotropic polypeptide, or the inhibitory peptide somatostatin. In this review, we describe the role of neurohormones that have been less characterized compared to others, on the regulation of insulin secretion. In particular, we will focus on melatonin, galanin, and RFamide neuropeptides QRFP26 and QRFP43, which display either insulinotropic or insulinostatic effects. In fact, in addition to other hormones, amino acids, cytokines, and a variety of proteins, brain-derived hormones are now considered as key regulators of glucose homeostasis, representing potential therapeutic targets for the treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Iacopo Gesmundo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Tania Villanova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dana Banfi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giacomo Gamba
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Riccarda Granata
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
- *Correspondence: Riccarda Granata,
| |
Collapse
|
191
|
Takahashi JS. Transcriptional architecture of the mammalian circadian clock. NATURE REVIEWS. GENETICS 2016. [PMID: 27990019 DOI: 10.1038/nrg.2016.150]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Circadian clocks are endogenous oscillators that control 24-hour physiological and behavioural processes in organisms. These cell-autonomous clocks are composed of a transcription-translation-based autoregulatory feedback loop. With the development of next-generation sequencing approaches, biochemical and genomic insights into circadian function have recently come into focus. Genome-wide analyses of the clock transcriptional feedback loop have revealed a global circadian regulation of processes such as transcription factor occupancy, RNA polymerase II recruitment and initiation, nascent transcription, and chromatin remodelling. The genomic targets of circadian clocks are pervasive and are intimately linked to the regulation of metabolism, cell growth and physiology.
Collapse
Affiliation(s)
- Joseph S Takahashi
- Howard Hughes Medical Institute, Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, NA4.118, Dallas, Texas 75390-9111, USA
| |
Collapse
|
192
|
Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet 2016; 18:164-179. [PMID: 27990019 DOI: 10.1038/nrg.2016.150] [Citation(s) in RCA: 1681] [Impact Index Per Article: 186.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Circadian clocks are endogenous oscillators that control 24-hour physiological and behavioural processes in organisms. These cell-autonomous clocks are composed of a transcription-translation-based autoregulatory feedback loop. With the development of next-generation sequencing approaches, biochemical and genomic insights into circadian function have recently come into focus. Genome-wide analyses of the clock transcriptional feedback loop have revealed a global circadian regulation of processes such as transcription factor occupancy, RNA polymerase II recruitment and initiation, nascent transcription, and chromatin remodelling. The genomic targets of circadian clocks are pervasive and are intimately linked to the regulation of metabolism, cell growth and physiology.
Collapse
Affiliation(s)
- Joseph S Takahashi
- Howard Hughes Medical Institute, Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, NA4.118, Dallas, Texas 75390-9111, USA
| |
Collapse
|
193
|
Chen BH, Hivert MF, Peters MJ, Pilling LC, Hogan JD, Pham LM, Harries LW, Fox CS, Bandinelli S, Dehghan A, Hernandez DG, Hofman A, Hong J, Joehanes R, Johnson AD, Munson PJ, Rybin DV, Singleton AB, Uitterlinden AG, Ying S, Melzer D, Levy D, van Meurs JBJ, Ferrucci L, Florez JC, Dupuis J, Meigs JB, Kolaczyk ED. Peripheral Blood Transcriptomic Signatures of Fasting Glucose and Insulin Concentrations. Diabetes 2016; 65:3794-3804. [PMID: 27625022 PMCID: PMC5127245 DOI: 10.2337/db16-0470] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/04/2016] [Indexed: 01/09/2023]
Abstract
Genome-wide association studies (GWAS) have successfully identified genetic loci associated with glycemic traits. However, characterizing the functional significance of these loci has proven challenging. We sought to gain insights into the regulation of fasting insulin and fasting glucose through the use of gene expression microarray data from peripheral blood samples of participants without diabetes in the Framingham Heart Study (FHS) (n = 5,056), the Rotterdam Study (RS) (n = 723), and the InCHIANTI Study (Invecchiare in Chianti) (n = 595). Using a false discovery rate q <0.05, we identified three transcripts associated with fasting glucose and 433 transcripts associated with fasting insulin levels after adjusting for age, sex, technical covariates, and complete blood cell counts. Among the findings, circulating IGF2BP2 transcript levels were positively associated with fasting insulin in both the FHS and RS. Using 1000 Genomes-imputed genotype data, we identified 47,587 cis-expression quantitative trait loci (eQTL) and 6,695 trans-eQTL associated with the 433 significant insulin-associated transcripts. Of note, we identified a trans-eQTL (rs592423), where the A allele was associated with higher IGF2BP2 levels and with fasting insulin in an independent genetic meta-analysis comprised of 50,823 individuals. We conclude that integration of genomic and transcriptomic data implicate circulating IGF2BP2 mRNA levels associated with glucose and insulin homeostasis.
Collapse
Affiliation(s)
- Brian H Chen
- Longitudinal Studies Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA
- Diabetes Research Center, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Marjolein J Peters
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging, Leiden and Rotterdam, the Netherlands
| | - Luke C Pilling
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, U.K
| | - John D Hogan
- Program in Bioinformatics, Boston University, Boston, MA
| | - Lisa M Pham
- Program in Bioinformatics, Boston University, Boston, MA
| | - Lorna W Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Caroline S Fox
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Stefania Bandinelli
- Geriatric Rehabilitation Unit, Azienda Sanitaria di Firenze, Florence, Italy
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dena G Hernandez
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Roby Joehanes
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
- Hebrew SeniorLife, Harvard Medical School, Boston, MA
| | - Andrew D Johnson
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Peter J Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD
| | - Denis V Rybin
- Data Coordinating Center, Boston University, Boston, MA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging, Leiden and Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Saixia Ying
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD
| | | | - David Melzer
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Daniel Levy
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging, Leiden and Rotterdam, the Netherlands
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Jose C Florez
- Diabetes Research Center, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Metabolism Program and Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Josée Dupuis
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - James B Meigs
- Metabolism Program and Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Eric D Kolaczyk
- Program in Bioinformatics, Boston University, Boston, MA
- Department of Mathematics and Statistics, Boston University, MA
| |
Collapse
|
194
|
Potter GDM, Skene DJ, Arendt J, Cade JE, Grant PJ, Hardie LJ. Circadian Rhythm and Sleep Disruption: Causes, Metabolic Consequences, and Countermeasures. Endocr Rev 2016; 37:584-608. [PMID: 27763782 PMCID: PMC5142605 DOI: 10.1210/er.2016-1083] [Citation(s) in RCA: 372] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circadian (∼24-hour) timing systems pervade all kingdoms of life and temporally optimize behavior and physiology in humans. Relatively recent changes to our environments, such as the introduction of artificial lighting, can disorganize the circadian system, from the level of the molecular clocks that regulate the timing of cellular activities to the level of synchronization between our daily cycles of behavior and the solar day. Sleep/wake cycles are intertwined with the circadian system, and global trends indicate that these, too, are increasingly subject to disruption. A large proportion of the world's population is at increased risk of environmentally driven circadian rhythm and sleep disruption, and a minority of individuals are also genetically predisposed to circadian misalignment and sleep disorders. The consequences of disruption to the circadian system and sleep are profound and include myriad metabolic ramifications, some of which may be compounded by adverse effects on dietary choices. If not addressed, the deleterious effects of such disruption will continue to cause widespread health problems; therefore, implementation of the numerous behavioral and pharmaceutical interventions that can help restore circadian system alignment and enhance sleep will be important.
Collapse
Affiliation(s)
- Gregory D M Potter
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Debra J Skene
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Josephine Arendt
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Janet E Cade
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Peter J Grant
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Laura J Hardie
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
195
|
Langlois C, Abadi A, Peralta-Romero J, Alyass A, Suarez F, Gomez-Zamudio J, Burguete-Garcia AI, Yazdi FT, Cruz M, Meyre D. Evaluating the transferability of 15 European-derived fasting plasma glucose SNPs in Mexican children and adolescents. Sci Rep 2016; 6:36202. [PMID: 27782183 PMCID: PMC5080582 DOI: 10.1038/srep36202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/12/2016] [Indexed: 12/15/2022] Open
Abstract
Genome wide association studies (GWAS) have identified single-nucleotide polymorphisms (SNPs) that are associated with fasting plasma glucose (FPG) in adult European populations. The contribution of these SNPs to FPG in non-Europeans and children is unclear. We studied the association of 15 GWAS SNPs and a genotype score (GS) with FPG and 7 metabolic traits in 1,421 Mexican children and adolescents from Mexico City. Genotyping of the 15 SNPs was performed using TaqMan Open Array. We used multivariate linear regression models adjusted for age, sex, body mass index standard deviation score, and recruitment center. We identified significant associations between 3 SNPs (G6PC2 (rs560887), GCKR (rs1260326), MTNR1B (rs10830963)), the GS and FPG level. The FPG risk alleles of 11 out of the 15 SNPs (73.3%) displayed significant or non-significant beta values for FPG directionally consistent with those reported in adult European GWAS. The risk allele frequencies for 11 of 15 (73.3%) SNPs differed significantly in Mexican children and adolescents compared to European adults from the 1000G Project, but no significant enrichment in FPG risk alleles was observed in the Mexican population. Our data support a partial transferability of European GWAS FPG association signals in children and adolescents from the admixed Mexican population.
Collapse
Affiliation(s)
- Christine Langlois
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Arkan Abadi
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Jesus Peralta-Romero
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Akram Alyass
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Fernando Suarez
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Jaime Gomez-Zamudio
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Ana I. Burguete-Garcia
- Centro de investigación sobre enfermedades infecciosas. Instituto Nacional de Salud Pública. Cuernavaca, Morelos, Mexico
| | - Fereshteh T. Yazdi
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Miguel Cruz
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
196
|
Wongchitrat P, Lansubsakul N, Kamsrijai U, Sae-Ung K, Mukda S, Govitrapong P. Melatonin attenuates the high-fat diet and streptozotocin-induced reduction in rat hippocampal neurogenesis. Neurochem Int 2016; 100:97-109. [PMID: 27620814 DOI: 10.1016/j.neuint.2016.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 08/10/2016] [Accepted: 09/06/2016] [Indexed: 12/15/2022]
Abstract
A deviant level of melatonin in blood circulation has been associated with the development of diabetes and with learning and memory deficiencies. Melatonin might have an important function in diabetes control; however, the mechanism of melatonin in diabetes remains unknown. The present study aimed to investigate the hyperglycemic condition induced by high-fat diet (HFD) feeding and streptozotocin (STZ) injection and to examine the effect of melatonin on adult hippocampal functions. HFD-fed and STZ-treated rats significantly increased blood glucose level. The present study showed that HFD-fed and STZ-treated rats significantly impaired memory in the Morris Water Maze task, reduced neurogenesis in the hippocampus shown by a reduction in nestin, doublecortin (DCX) and β-III tubulin immunoreactivities, reduced axon terminal markers, synaptophysin, reduced dendritic marker including postsynaptic density 95 (PSD-95) and the glutamate receptor subunit NR2A. Moreover, a significant downregulation of melatonin receptor, insulin receptor-β (IR-β) and both p-IR-β and phosphorylated extracellular signal-regulated kinase (p-ERK) occurred in HFD-fed and STZ-treated rats, while the level of glial fibrillary acidic protein (GFAP) increased. Treatment of melatonin, rats had shorter escape latencies and remained in the target quadrant longer compared to the HFD-fed and STZ-treated rats. Melatonin attenuated the reduction of neurogenesis, synaptogenesis and the induction of astrogliosis. Moreover, melatonin countered the reduction of melatonin receptor, insulin receptor and downstream signaling pathway for insulin. Our data suggested that the dysfunction of insulin signaling pathway occurred in the diabetes may provide a convergent mechanism of hippocampal impaired neurogenesis and synaptogenesis lead to impair memory while melatonin reverses these effects, suggesting that melatonin may reduce the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand
| | - Niyada Lansubsakul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand; Department of Anatomy, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Utcharaporn Kamsrijai
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand
| | - Kwankanit Sae-Ung
- Innovative Learning Center, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand; Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
197
|
Tsuneki H, Sasaoka T, Sakurai T. Sleep Control, GPCRs, and Glucose Metabolism. Trends Endocrinol Metab 2016; 27:633-642. [PMID: 27461005 DOI: 10.1016/j.tem.2016.06.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022]
Abstract
Modern lifestyles prolong daily activities into the nighttime, disrupting circadian rhythms, which may cause sleep disturbances. Sleep disturbances have been implicated in the dysregulation of blood glucose levels and reported to increase the risk of type 2 diabetes (T2D) and diabetic complications. Sleep disorders are treated using anti-insomnia drugs that target ionotropic and G protein-coupled receptors (GPCRs), including γ-aminobutyric acid (GABA) agonists, melatonin agonists, and orexin receptor antagonists. A deeper understanding of the effects of these medications on glucose metabolism and their underlying mechanisms of action is crucial for the treatment of diabetic patients with sleep disorders. In this review we focus on the beneficial impact of sleep on glucose metabolism and suggest a possible strategy for therapeutic intervention against sleep-related metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- 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.
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
198
|
Jockers R, Delagrange P, Dubocovich ML, Markus RP, Renault N, Tosini G, Cecon E, Zlotos DP. Update on melatonin receptors: IUPHAR Review 20. Br J Pharmacol 2016; 173:2702-25. [PMID: 27314810 DOI: 10.1111/bph.13536] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 02/06/2023] Open
Abstract
Melatonin receptors are seven transmembrane-spanning proteins belonging to the GPCR superfamily. In mammals, two melatonin receptor subtypes exist - MT1 and MT2 - encoded by the MTNR1A and MTNR1B genes respectively. The current review provides an update on melatonin receptors by the corresponding subcommittee of the International Union of Basic and Clinical Pharmacology. We will highlight recent developments of melatonin receptor ligands, including radioligands, and give an update on the latest phenotyping results of melatonin receptor knockout mice. The current status and perspectives of the structure of melatonin receptor will be summarized. The physiological importance of melatonin receptor dimers and biologically important and type 2 diabetes-associated genetic variants of melatonin receptors will be discussed. The role of melatonin receptors in physiology and disease will be further exemplified by their functions in the immune system and the CNS. Finally, antioxidant and free radical scavenger properties of melatonin and its relation to melatonin receptors will be critically addressed.
Collapse
Affiliation(s)
- Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,University Paris Descartes, Paris, France
| | | | - Margarita L Dubocovich
- Department Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Science, University at Buffalo (SUNY), Buffalo, USA
| | - Regina P Markus
- Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Gianluca Tosini
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Erika Cecon
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Darius P Zlotos
- Department of Pharmaceutical Chemistry, The German University in Cairo, New Cairo City, Cairo, Egypt
| |
Collapse
|
199
|
Kong X, Xing X, Hong J, Zhang X, Yang W. Association of a type 2 diabetes genetic risk score with insulin secretion modulated by insulin sensitivity among Chinese Hans. Clin Genet 2016; 91:832-842. [PMID: 27280334 DOI: 10.1111/cge.12817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 01/03/2023]
Abstract
Type 2 diabetes (T2D) is characterized by insulin resistance and impaired insulin secretion. The present study aimed to identify the influence of insulin sensitivity on the genetic risk of impaired insulin secretion among a Chinese Han population. For 3229 controls and 1994 treatment-naïve T2D, single nucleotide polymorphisms (SNPs) from 24 T2D-related genomic loci were genotyped and a genetic risk score (GRS) was constructed. Results showed that GRS was associated with insulin secretion and disposition indices in both controls and treatment-naïve T2Ds. Upon stratifying the participants into tertiles by the Matsuda index, we observed an inhibitory relationship between the GRS and insulin secretion in low insulin sensitive but not in high insulin sensitive controls and treatment-naïve T2Ds. Moreover, low insulin sensitive individuals exhibited more severe impairment in insulin secretion and beta cell response to insulin sensitivity with an increase in risk alleles. Our findings identified that the association of GRS with insulin secretion was strongly modulated by insulin sensitivity in both controls and T2Ds of Chinese Han. It indicates that insulin sensitization should be emphasized in prevention and treatment of T2D for beta cell protection.
Collapse
Affiliation(s)
- X Kong
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - X Xing
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - J Hong
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - X Zhang
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - W Yang
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
200
|
Palmer ND, Wagenknecht LE, Langefeld CD, Wang N, Buchanan TA, Xiang AH, Allayee H, Bergman RN, Raffel LJ, Chen YDI, Haritunians T, Fingerlin T, Goodarzi MO, Taylor KD, Rotter JI, Watanabe RM, Bowden DW. Improved Performance of Dynamic Measures of Insulin Response Over Surrogate Indices to Identify Genetic Contributors of Type 2 Diabetes: The GUARDIAN Consortium. Diabetes 2016; 65:2072-80. [PMID: 27207554 PMCID: PMC4915581 DOI: 10.2337/db15-1543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/09/2016] [Indexed: 01/24/2023]
Abstract
Type 2 diabetes (T2D) is a heterogeneous disorder with contributions from peripheral insulin resistance and β-cell dysfunction. For minimization of phenotypic heterogeneity, quantitative intermediate phenotypes characterizing basal glucose homeostasis (insulin resistance and HOMA of insulin resistance [HOMAIR] and of β-cell function [HOMAB]) have shown promise in relatively large samples. We investigated the utility of dynamic measures of glucose homeostasis (insulin sensitivity [SI] and acute insulin response [AIRg]) evaluating T2D-susceptibility variants (n = 57) in Hispanic Americans from the GUARDIAN Consortium (n = 2,560). Basal and dynamic measures were genetically correlated (HOMAB-AIRg: ρG = 0.28-0.73; HOMAIR-SI: ρG = -0.73 to -0.83) with increased heritability for the dynamic measure AIRg Significant association of variants with dynamic measures (P < 8.77 × 10(-4)) was observed. A pattern of superior performance of AIRg was observed for well-established loci including MTNR1B (P = 9.46 × 10(-12)), KCNQ1 (P = 1.35 × 10(-4)), and TCF7L2 (P = 5.10 × 10(-4)) with study-wise statistical significance. Notably, significant association of MTNR1B with AIRg (P < 1.38 × 10(-9)) was observed in a population one-fourteenth the size of the initial discovery cohort. These observations suggest that basal and dynamic measures provide different views and levels of sensitivity to discrete elements of glucose homeostasis. Although more costly to obtain, dynamic measures yield significant results that could be considered physiologically "closer" to causal pathways and provide insight into the discrete mechanisms of action.
Collapse
Affiliation(s)
- Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Carl D Langefeld
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Nan Wang
- Department of Preventative Medicine, Keck School of Medicine of USC, Los Angeles, CA Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA
| | - Thomas A Buchanan
- Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA Department of Medicine, Keck School of Medicine of USC, Los Angeles, CA
| | - Anny H Xiang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Hooman Allayee
- Department of Preventative Medicine, Keck School of Medicine of USC, Los Angeles, CA
| | - Richard N Bergman
- Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA
| | - Leslie J Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Tasha Fingerlin
- Department of Epidemiology, University of Colorado Denver, Aurora, CO Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, CO
| | - Mark O Goodarzi
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Kent D Taylor
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Richard M Watanabe
- Department of Preventative Medicine, Keck School of Medicine of USC, Los Angeles, CA Department of Physiology and Biophysics, Keck School of Medicine of USC, Los Angeles, CA
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|