1
|
Brito Nunes C, Borges MC, Freathy RM, Lawlor DA, Qvigstad E, Evans DM, Moen GH. Understanding the Genetic Landscape of Gestational Diabetes: Insights into the Causes and Consequences of Elevated Glucose Levels in Pregnancy. Metabolites 2024; 14:508. [PMID: 39330515 PMCID: PMC11434570 DOI: 10.3390/metabo14090508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Background/Objectives: During pregnancy, physiological changes in maternal circulating glucose levels and its metabolism are essential to meet maternal and fetal energy demands. Major changes in glucose metabolism occur throughout pregnancy and consist of higher insulin resistance and a compensatory increase in insulin secretion to maintain glucose homeostasis. For some women, this change is insufficient to maintain normoglycemia, leading to gestational diabetes mellitus (GDM), a condition characterized by maternal glucose intolerance and hyperglycaemia first diagnosed during the second or third trimester of pregnancy. GDM is diagnosed in approximately 14.0% of pregnancies globally, and it is often associated with short- and long-term adverse health outcomes in both mothers and offspring. Although recent studies have highlighted the role of genetic determinants in the development of GDM, research in this area is still lacking, hindering the development of prevention and treatment strategies. Methods: In this paper, we review recent advances in the understanding of genetic determinants of GDM and glycaemic traits during pregnancy. Results/Conclusions: Our review highlights the need for further collaborative efforts as well as larger and more diverse genotyped pregnancy cohorts to deepen our understanding of the genetic aetiology of GDM, address research gaps, and further improve diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Caroline Brito Nunes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Rachel M. Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 4PY, UK;
| | - Deborah A. Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Elisabeth Qvigstad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - David M. Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Frazer Institute, University of Queensland, Brisbane 4102, Australia
| | - Gunn-Helen Moen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Frazer Institute, University of Queensland, Brisbane 4102, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
2
|
Guo H, Peng H, Wang S, Hou T, Li Y, Zhang H, Jiang J, Ma B, Wang M, Wu Y, Qin X, Tang X, Chen D, Li J, Hu Y, Wu T. Healthy Lifestyles Modify the Association of Melatonin Receptor 1B Gene and Ischemic Stroke: A Family-Based Cohort Study in Northern China. J Pineal Res 2024; 76:e13000. [PMID: 39101387 DOI: 10.1111/jpi.13000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/15/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
Limited research has reported the association between MTNR1B gene polymorphisms and ischemic stroke (IS), and there is insufficient evidence on whether adopting a healthy lifestyle can mitigate genetic risks in this context. This study aimed to investigate the associations between MTNR1B gene variants (rs10830963 and rs1387153) and IS, examining the potential effect of gene-lifestyle interactions on IS risk. Conducted in northern China, this family-based cohort study involved 5116 initially IS-free subjects. Genotype data for rs10830963 and rs1387153 in MTNR1B were collected. Eight modifiable lifestyle factors, including body mass index (BMI), smoking, alcohol consumption, dietary habits, physical activity, sedentary time, sleep duration, and chronotype, were considered in calculating healthy lifestyle scores. Multilevel Cox models were used to examine the associations between MTNR1B variants and IS. Participants carrying the rs10830963-G and rs1387153-T alleles exhibited an elevated IS risk. Each additional rs10830963-G allele and rs1387153-T allele increased the IS risk by 36% (HR = 1.36, 95% CI, 1.12-1.65) and 32% (HR = 1.32, 95% CI, 1.09-1.60), respectively. Participants were stratified into low, medium, and high healthy lifestyle score groups (1537, 2188, and 1391 participants, respectively). Genetic-lifestyle interactions were observed for rs10830963 and rs1387153 (p for interaction < 0.001). Notably, as the healthy lifestyle score increased, the effect of MTNR1B gene variants on IS risk diminished (p for trend < 0.001). This study underscores the association between the MTNR1B gene and IS, emphasizing that adherence to a healthy lifestyle can mitigate the genetic predisposition to IS.
Collapse
Affiliation(s)
- Huangda Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Hexiang Peng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Siyue Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tianjiao Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yixin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Hanyu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jin Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Bohao Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Mengying Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Yiqun Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xun Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dafang Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
3
|
Zhen J, Gu Y, Wang P, Wang W, Bian S, Huang S, Liang H, Huang M, Yu Y, Chen Q, Jiang G, Qiu X, Xiong L, Liu S. Genome-wide association and Mendelian randomisation analysis among 30,699 Chinese pregnant women identifies novel genetic and molecular risk factors for gestational diabetes and glycaemic traits. Diabetologia 2024; 67:703-713. [PMID: 38372780 PMCID: PMC10904416 DOI: 10.1007/s00125-023-06065-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/03/2023] [Indexed: 02/20/2024]
Abstract
AIMS/HYPOTHESIS Gestational diabetes mellitus (GDM) is the most common disorder in pregnancy; however, its underlying causes remain obscure. This study aimed to investigate the genetic and molecular risk factors contributing to GDM and glycaemic traits. METHODS We collected non-invasive prenatal test (NIPT) sequencing data along with four glycaemic and 55 biochemical measurements from 30,699 pregnant women during a 2 year period at Shenzhen Baoan Women's and Children's Hospital in China. Genome-wide association studies (GWAS) were conducted between genotypes derived from NIPTs and GDM diagnosis, baseline glycaemic levels and glycaemic levels after glucose challenges. In total, 3317 women were diagnosed with GDM, while 19,565 served as control participants. The results were replicated using two independent cohorts. Additionally, we performed one-sample Mendelian randomisation to explore potential causal associations between the 55 biochemical measurements and risk of GDM and glycaemic levels. RESULTS We identified four genetic loci significantly associated with GDM susceptibility. Among these, MTNR1B exhibited the highest significance (rs10830963-G, OR [95% CI] 1.57 [1.45, 1.70], p=4.42×10-29), although its effect on type 2 diabetes was modest. Furthermore, we found 31 genetic loci, including 14 novel loci, that were significantly associated with the four glycaemic traits. The replication rates of these associations with GDM, fasting plasma glucose levels and 0 h, 1 h and 2 h OGTT glucose levels were 4 out of 4, 6 out of 9, 10 out of 11, 5 out of 7 and 4 out of 4, respectively. Mendelian randomisation analysis suggested that a genetically regulated higher lymphocytes percentage and lower white blood cell count, neutrophil percentage and absolute neutrophil count were associated with elevated glucose levels and an increased risk of GDM. CONCLUSIONS/INTERPRETATION Our findings provide new insights into the genetic basis of GDM and glycaemic traits during pregnancy in an East Asian population and highlight the potential role of inflammatory pathways in the aetiology of GDM and variations in glycaemic levels. DATA AVAILABILITY Summary statistics for GDM; fasting plasma glucose; 0 h, 1 h and 2h OGTT; and the 55 biomarkers are available in the GWAS Atlas (study accession no.: GVP000001, https://ngdc.cncb.ac.cn/gwas/browse/GVP000001) .
Collapse
Affiliation(s)
- Jianxin Zhen
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuqin Gu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Piao Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Weihong Wang
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Shengzhe Bian
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shujia Huang
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hui Liang
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Birth Defects Research, Shenzhen, Guangdong, China
| | - Mingxi Huang
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yan Yu
- Department of Obstetrics, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Qing Chen
- Department of Pharmacy, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiu Qiu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Women's Health, Provincial Key Clinical Specialty of Woman and Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Likuan Xiong
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Birth Defects Research, Shenzhen, Guangdong, China.
| | - Siyang Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Guo C, He J, Deng X, Wang D, Yuan G. Potential therapeutic value of melatonin in diabetic nephropathy: improvement beyond anti-oxidative stress. Arch Physiol Biochem 2023; 129:1250-1261. [PMID: 34048666 DOI: 10.1080/13813455.2021.1933539] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022]
Abstract
Diabetic nephropathy (DN) is a common complication of diabetes, and it is also the main cause of chronic renal failure. Physiological/pathological changes mediated by high glucose are the main factors causing injury of DN, including the enhancement of polyol pathway, the accumulation of advanced glycation products (AGEs), and the activation of protein kinase C (PKC) and transforming growth factor-β (TGF-β) signals. In addition, the abnormal activation of renin-angiotensin system (RAS) and oxidative stress are also involved. Melatonin is a physiological hormone mainly secreted by the pineal gland which has been proved to be related to diabetes. Studies have shown that exogenous melatonin intervention can reduce blood glucose and alleviate high glucose mediated pathological damage. At the same time, melatonin also has a strong antioxidant effect, and can inhibit the activation of RAS. Therefore, it is of great significance to explore the therapeutic effect and value of melatonin on DN.
Collapse
Affiliation(s)
- Chang Guo
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xia Deng
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Dong Wang
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guoyue Yuan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
Ngwa NE, Matshazi DM, Davison GM, Kengne AP, Matsha TE. Association between the MTNR1B, HHEX, SLC30A8, and TCF7L2 single nucleotide polymorphisms and cardiometabolic risk profile in a mixed ancestry South African population. Sci Rep 2023; 13:17122. [PMID: 37816730 PMCID: PMC10564755 DOI: 10.1038/s41598-023-43560-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Single nucleotide polymorphisms of the TCF7L2, HHEX, SLC30A8, MTNR1B, SLC2A2 and GLIS3 genes are well established candidate genes for cardiometabolic diseases (CMDs) across different ethnic populations. We investigated their association with CMDs in a mixed ancestry population of South Africa. rs10830963, rs1111875, rs11920090, rs13266634, rs7034200 and rs7903146 SNPs were genotyped by quantitative real time PCR in 1650 participants and Hardy-Weinberg equilibrium (HWE) analyses performed on the SNPs. Diabetes, obesity, hypertension and cardiometabolic traits were compared across genotypes of SNPs in HWE. Linear and logistic regressions adjusting for age, gender and body mass index were used to determine the risk of T2DM, obesity and hypertension. rs7903146 (p = 0.055), rs1111875 (p = 0.465), rs13266634 (p = 0.828), and rs10830963 (p = 0.158) were in HWE. The rs10830963 recessive genotype was able to predict FPG, insulin and HOMA-IR, while the rs1111875 recessive genotype was able to predict total cholesterol, triglyceride, LDL cholesterol and FPG. The rs7903146 recessive genotype was able to predict SBP and LDL cholesterol. The recessive genotypes of MTNRIB and HHEX SNPs were associated with T2DM traits in the study population and could partially explain the high prevalence of T2DM. Further studies are required to confirm these findings and establish candidate genes in the African population.
Collapse
Affiliation(s)
- Ndonwi Elvis Ngwa
- South African Medical Research Council/Cape Peninsula University of Technology, Cardio-Metabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Old Science Building, Cape Town, South Africa.
- Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé 1, Yaoundé, Cameroon.
| | - Don Makwakiwe Matshazi
- South African Medical Research Council/Cape Peninsula University of Technology, Cardio-Metabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Old Science Building, Cape Town, South Africa
| | - Glenda Mary Davison
- South African Medical Research Council/Cape Peninsula University of Technology, Cardio-Metabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Old Science Building, Cape Town, South Africa
| | - Andre Pascal Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Tandi Edith Matsha
- South African Medical Research Council/Cape Peninsula University of Technology, Cardio-Metabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Old Science Building, Cape Town, South Africa
- Sefako Makgatho Health Sciences University, Ga-Rankuwa, South Africa
| |
Collapse
|
6
|
Dallatana A, Cremonesi L, Trombetta M, Fracasso G, Nocini R, Giacomello L, Innamorati G. G Protein-Coupled Receptors and the Rise of Type 2 Diabetes in Children. Biomedicines 2023; 11:1576. [PMID: 37371671 DOI: 10.3390/biomedicines11061576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
The human genome counts hundreds of GPCRs specialized to sense thousands of different extracellular cues, including light, odorants and nutrients in addition to hormones. Primordial GPCRs were likely glucose transporters that became sensors to monitor the abundance of nutrients and direct the cell to switch from aerobic metabolism to fermentation. Human β cells express multiple GPCRs that contribute to regulate glucose homeostasis, cooperating with many others expressed by a variety of cell types and tissues. These GPCRs are intensely studied as pharmacological targets to treat type 2 diabetes in adults. The dramatic rise of type 2 diabetes incidence in pediatric age is likely correlated to the rapidly evolving lifestyle of children and adolescents of the new century. Current pharmacological treatments are based on therapies designed for adults, while youth and puberty are characterized by a different hormonal balance related to glucose metabolism. This review focuses on GPCRs functional traits that are relevant for β cells function, with an emphasis on aspects that could help to differentiate new treatments specifically addressed to young type 2 diabetes patients.
Collapse
Affiliation(s)
- Alessia Dallatana
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| | - Linda Cremonesi
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| | - Maddalena Trombetta
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, 37124 Verona, Italy
| | - Giulio Fracasso
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Riccardo Nocini
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| | - Luca Giacomello
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| | - Giulio Innamorati
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| |
Collapse
|
7
|
Patel R, Parmar N, Palit SP, Rathwa N, Begum R. A novel combination of sitagliptin and melatonin ameliorates T2D manifestations: studies on experimental diabetic models. J Endocrinol Invest 2023:10.1007/s40618-023-02014-6. [PMID: 36692817 DOI: 10.1007/s40618-023-02014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Type 2 diabetes (T2D) is an endocrine disorder characterized by hyperglycemia, insulin resistance, dysregulated glucose and lipid metabolism, reduced pancreatic β-cell function and mass, and a reduced incretin effect. Circadian rhythm disruption is associated with increased T2D risk. We have investigated the therapeutic potential of a combination of melatonin (M) and sitagliptin (S), a dipeptidyl peptidase IV (DPP-IV) inhibitor, in the amelioration of T2D manifestations in high-fat diet (HFD) induced T2D mouse model and also on β-cell proliferation under gluco-lipotoxicity stress in vitro. METHODS For in vivo study, mice were fed with HFD for 25 weeks to induce T2D and were treated with monotherapies and S + M for four weeks. For the in vitro study, primary mouse islets were exposed to normal glucose and high glucose + palmitate to induce gluco-lipotoxic stress. RESULTS Our results suggest that monotherapies and S + M improve metabolic parameters and glyco-lipid metabolism in the liver and adipose tissue, respectively, and improve mitochondrial function in the skeletal muscle. Moreover, it increases peripheral insulin sensitivity. Our in vitro and in vivo studies suggest that β-cell mass was preserved in all the drug-treated groups. CONCLUSION The combination treatment is superior to monotherapies in the management of T2D.
Collapse
Affiliation(s)
- R Patel
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - N Parmar
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - S P Palit
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - N Rathwa
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - R Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
8
|
Li YY, Wang H, Zhang YY. Melatonin receptor 1B gene rs10830963 C/G polymorphism associated with type 2 diabetes mellitus: An updated meta-analysis of 13,752 participants. Heliyon 2022; 8:e11786. [DOI: 10.1016/j.heliyon.2022.e11786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/13/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
|
9
|
Patel R, Parmar N, Rathwa N, Palit SP, Li Y, Garcia-Ocaña A, Begum R. A novel therapeutic combination of sitagliptin and melatonin regenerates pancreatic β-cells in mouse and human islets. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119263. [PMID: 35364117 DOI: 10.1016/j.bbamcr.2022.119263] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023]
Abstract
Autoimmune-led challenge resulting in β-cell loss is responsible for the development of type 1 diabetes (T1D). Melatonin, a pineal hormone or sitagliptin, a dipeptidyl peptidase IV (DPP-IV) inhibitor, has increased β-cell mass in various diabetic models and has immunoregulatory property. Both β-cell regenerative capacity and melatonin secretion decrease with ageing. Thus, we aimed to investigate the therapeutic potential of melatonin combined with sitagliptin on β-cell regeneration under glucotoxic stress, in the streptozotocin-induced young and old diabetic mouse models, and euglycemic humanized islet transplant mouse model. Our results suggest that combination therapy of sitagliptin and melatonin show an additive effect in inducing mouse β-cell regeneration under glucotoxic stress, and in the human islet transplant mouse model. Further, in the young diabetic mouse model, the monotherapies induce β-cell transdifferentiation and reduce β-cell apoptosis whereas, in the old diabetic mouse model, melatonin and sitagliptin induce β-cell proliferation and β-cell transdifferentiation, and it also reduces β-cell apoptosis. Further, in both the models, combination therapy reduces fasting blood glucose levels, increases plasma insulin levels and glucose tolerance and promotes β-cell proliferation, β-cell transdifferentiation, and reduces β-cell apoptosis. It can be concluded that combination therapy is superior to monotherapies in ameliorating diabetic manifestations, and it can be used as a future therapy for β-cell regeneration in diabetes patients.
Collapse
Affiliation(s)
- Roma Patel
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara- 390002, Gujarat, India
| | - Nishant Parmar
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara- 390002, Gujarat, India
| | - Nirali Rathwa
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara- 390002, Gujarat, India
| | - Sayantani Pramanik Palit
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara- 390002, Gujarat, India
| | - Yansui Li
- Diabetes, Obesity and Metabolism Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity and Metabolism Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara- 390002, Gujarat, India.
| |
Collapse
|
10
|
Hagström A, Kal Omar R, Williams PA, Stålhammar G. The rationale for treating uveal melanoma with adjuvant melatonin: a review of the literature. BMC Cancer 2022; 22:398. [PMID: 35413810 PMCID: PMC9006630 DOI: 10.1186/s12885-022-09464-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/28/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Uveal melanoma is a rare form of cancer with high mortality. The incidence of metastases is attributed to early seeding of micrometastases from the eye to distant organs, primarily the liver. Once these seeded clusters of dormant tumor cells grow into larger radiologically detectable macrometastases, median patient survival is about 1 year. Melatonin is an important hormone for synchronizing circadian rhythms. It is also involved in other aspects of human physiology and may offer therapeutic benefits for a variety of diseases including cancer. METHODS Articles involving the physiological effects of melatonin, pharmacokinetics, and previous use in cancer studies were acquired using a comprehensive literature search in the Medline (PubMed) and Web of Science databases. In total, 147 publications were selected and included in the review. RESULTS Melatonin has been observed to suppress the growth of cancer cells, inhibit metastatic spread, enhance immune system functions, and act as an anti-inflammatory in both in vitro and in vivo models. Melatonin may also enhance the efficacy of cancer treatments such as immuno- and chemotherapy. Numerous studies have shown promising results for oral melatonin supplementation in patients with other forms of cancer including cutaneous malignant melanoma. Cell line and animal studies support a hypothesis in which similar benefits may exist for uveal melanoma. CONCLUSIONS Given its low cost, good safety profile, and limited side effects, there may be potential for the use of melatonin as an adjuvant oncostatic treatment. Future avenues of research could include clinical trials to evaluate the effect of melatonin in prevention of macrometastases of uveal melanoma.
Collapse
Affiliation(s)
- Anna Hagström
- Department of Medicine, Karolinska Institutet, D1:04, 171 76, Stockholm, Sweden.
| | - Ruba Kal Omar
- Department of Medicine, Karolinska Institutet, D1:04, 171 76, Stockholm, Sweden.
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - Gustav Stålhammar
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden
- St. Erik Eye Hospital, Box 4078, 171 04, Stockholm, Sweden
| |
Collapse
|
11
|
Pfeffer M, von Gall C, Wicht H, Korf HW. The Role of the Melatoninergic System in Circadian and Seasonal Rhythms—Insights From Different Mouse Strains. Front Physiol 2022; 13:883637. [PMID: 35492605 PMCID: PMC9039042 DOI: 10.3389/fphys.2022.883637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 01/01/2023] Open
Abstract
The melatoninergic system comprises the neurohormone melatonin and its molecular targets. The major source of melatonin is the pineal organ where melatonin is rhythmically produced during darkness. In mammals, melatonin biosynthesis is controlled by the central circadian rhythm generator in the suprachiasmatic nucleus (SCN) and photoreceptors in the retina. Melatonin elicits its function principally through two specific receptors called MT1 and MT2. MT1 is highly expressed in the SCN and the hypophysial pars tuberalis (PT), an important interface for control of seasonal functions. The expression of the MT2 is more widespread. The role of the melatoninergic system in the control of seasonal functions, such as reproduction, has been known for more than 4 decades, but investigations on its impact on the circadian system under normal (entrained) conditions started 2 decades later by comparing mouse strains with a fully functional melatoninergic system with mouse strains which either produce insufficient amounts of melatonin or lack the melatonin receptors MT1 and MT2. These studies revealed that an intact melatoninergic system is not required for the generation or maintenance of rhythmic behavior under physiological entrained conditions. As shown by jet lag experiments, the melatoninergic system facilitated faster re-entrainment of locomotor activity accompanied by a more rapid adaptation of the molecular clock work in the SCN. This action depended on MT2. Further studies indicated that the endogenous melatoninergic system stabilizes the locomotor activity under entrained conditions. Notably, these effects of the endogenous melatoninergic system are subtle, suggesting that other signals such as corticosterone or temperature contribute to the synchronization of locomotor activity. Outdoor experiments lasting for a whole year indicate a seasonal plasticity of the chronotype which depends on the melatoninergic system. The comparison between mice with an intact or a compromised melatoninergic system also points toward an impact of this system on sleep, memory and metabolism.
Collapse
Affiliation(s)
- Martina Pfeffer
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- *Correspondence: Martina Pfeffer,
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Helmut Wicht
- Dr. Senckenbergische Anatomie II, Fachbereich Medizin der Goethe-Universität, Frankfurt am Main, Germany
| | - Horst-Werner Korf
- Institute of Anatomy I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
12
|
Zibolka J, Bähr I, Peschke E, Mühlbauer E, Bazwinsky-Wutschke I. Human and Rodent Cell Lines as Models of Functional Melatonin-Responsive Pancreatic Islet Cells. Methods Mol Biol 2022; 2550:329-352. [PMID: 36180704 DOI: 10.1007/978-1-0716-2593-4_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cell culture of different pancreatic islet cell lines, like the murine α-cell line αTC1.9, the rat β-cell lines INS-1 and INS-1 832/13, and the human δ-cell line QGP-1, can serve as valuable cell models for the analysis of melatonin-dependent modulation of hormone secretion. The paper summarizes in detail the requirements of culture for each cell line and includes batch protocols to stimulate hormone secretion and to treat cells with several melatonin concentrations as previously published. We here describe the processing of collected cell pellets or cell culture supernatants as well as different methods to analyze cell experiments after melatonin treatment on the basis of our own experience. Finally, we outlined for each cell line under which conditions the melatonin treatment should be performed to gain reproducible results.
Collapse
Affiliation(s)
- Juliane Zibolka
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ina Bähr
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Elmar Peschke
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Eckhard Mühlbauer
- Saxon Academy of Sciences and Humanities in Leipzig, Leipzig, Germany
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
13
|
Obayemi MJ, Akintayo CO, Oniyide AA, Aturamu A, Badejogbin OC, Atuma CL, Saidi AO, Mahmud H, Olaniyi KS. Protective role of melatonin against adipose-hepatic metabolic comorbidities in experimentally induced obese rat model. PLoS One 2021; 16:e0260546. [PMID: 34879109 PMCID: PMC8654266 DOI: 10.1371/journal.pone.0260546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Background Adipose and hepatic metabolic dysfunctions are critical comorbidities that
also aggravate insulin resistance in obese individuals. Melatonin is a
low-cost agent and previous studies suggest that its use may promote
metabolic health. However, its effects on some comorbidities associated with
obesity are unknown. Herein, we investigated the hypothesis that melatonin
supplementation would attenuate adipose-hepatic metabolic dysfunction in
high fat diet (HFD)-induced obesity in male Wistar rats. Materials and methods Twenty-four adult male Wistar rats (n = 6/group) were used: Control group
received vehicle (normal saline), obese group received 40% high fat diet,
melatonin-treated group received 4 mg/kg of melatonin, and obese plus
melatonin group received 40% HFD and melatonin. The treatment lasted for 12
weeks. Results HFD caused increased food intake, body weight, insulin level, insulin
resistance and plasma and liver lipid but decreased adipose lipid. In
addition, HFD also increased plasma, adipose and liver malondialdehyde,
IL-6, uric acid and decreased Glucose-6-phosphate dehydrogenase,
glutathione, nitric oxide and circulating obestatin concentration. However,
these deleterious effects except food intake were attenuated when
supplemented with melatonin. Conclusion Taken together, the present results indicate that HFD exposure causes
adipose-hepatic metabolic disturbance in obese animals, which are
accompanied by oxidative stress and inflammation. In addition, the present
results suggest that melatonin supplementation attenuates adipose-hepatic
metabolic dysfunction, accompanying obesity by suppression of oxidative
stress/inflammation-dependent mechanism and increasing circulating
obestatin.
Collapse
Affiliation(s)
- Mary J. Obayemi
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Christopher O. Akintayo
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Adesola A. Oniyide
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Ayodeji Aturamu
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Olabimpe C. Badejogbin
- Department of Physiology, Benjamin Carson School of Medicine, Babcock
University, Ilishan-Remo, Nigeria
| | - Chukwubueze L. Atuma
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Azeezat O. Saidi
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Hadiza Mahmud
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Kehinde S. Olaniyi
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
- * E-mail: ,
| |
Collapse
|
14
|
Lauritzen ES, Støy J, Bæch-Laursen C, Grarup N, Jessen N, Hansen T, Møller N, Hartmann B, Holst JJ, Kampmann U. The Effect of Melatonin on Incretin Hormones: Results From Experimental and Randomized Clinical Studies. J Clin Endocrinol Metab 2021; 106:e5109-e5123. [PMID: 34265066 DOI: 10.1210/clinem/dgab521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Indexed: 01/10/2023]
Abstract
CONTEXT Glucose homeostasis is under circadian control through both endocrine and intracellular mechanisms, with several lines of evidence suggesting that melatonin affects glucose homeostasis. OBJECTIVE To evaluate the acute in vivo and in situ effects of melatonin on secretion of the incretin hormones, glucagon-like-peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP), and their impact on β-cell insulin secretion. DESIGN A human randomized, double-blinded, placebo-controlled crossover study combined with a confirmatory in situ study of perfused rat intestines. SETTING Aarhus University Hospital. METHODS Fifteen healthy male participants were examined 2 × 2 times: an oral glucose tolerance test (OGTT) was performed on day 1 and an isoglycemic IV glucose infusion replicating the blood glucose profile of the OGTT day was performed on day 2. These pairs of study days were repeated on treatment with melatonin and placebo, respectively. For the in situ study, 6 rat intestines and 4 rat pancreases were perfused arterially with perfusion buffer ± melatonin. The intestines were concomitantly perfused with glucose through the luminal compartment. RESULTS In humans, melatonin treatment resulted in reduced GIP secretion compared with placebo (ANOVA P = 0.003), an effect also observed in the perfused rat intestines (ANOVA P = 0.003), in which GLP-1 secretion also was impaired by arterial melatonin infusion (ANOVA P < 0.001). Despite a decrease in GIP levels, the in vivo glucose-stimulated insulin secretion was unaffected by melatonin (P = 0.78). CONCLUSION Melatonin reduced GIP secretion during an oral glucose challenge in healthy young men but did not affect insulin secretion. Reduced GIP secretion was confirmed in an in situ model of the rat intestine.
Collapse
Affiliation(s)
- Esben Stistrup Lauritzen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Medical research laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julie Støy
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Medical research laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Cecilie Bæch-Laursen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Research Laboratory for Biochemical Pathology, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Møller
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Medical research laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Medical research laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
15
|
Jung SY. Genetic Signatures of Glucose Homeostasis: Synergistic Interplay With Long-Term Exposure to Cigarette Smoking in Development of Primary Colorectal Cancer Among African American Women. Clin Transl Gastroenterol 2021; 12:e00412. [PMID: 34608882 PMCID: PMC8500576 DOI: 10.14309/ctg.0000000000000412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/22/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Insulin resistance (IR)/glucose intolerance is a critical biologic mechanism for the development of colorectal cancer (CRC) in postmenopausal women. Whereas IR and excessive adiposity are more prevalent in African American (AA) women than in White women, AA women are underrepresented in genome-wide studies for systemic regulation of IR and the association with CRC risk. METHODS With 780 genome-wide IR single-nucleotide polymorphisms (SNPs) among 4,692 AA women, we tested for a causal inference between genetically elevated IR and CRC risk. Furthermore, by incorporating CRC-associated lifestyle factors, we established a prediction model on the basis of gene-environment interactions to generate risk profiles for CRC with the most influential genetic and lifestyle factors. RESUTLS In the pooled Mendelian randomization analysis, the genetically elevated IR was associated with 9 times increased risk of CRC, but with lack of analytic power. By addressing the variation of individual SNPs in CRC in the prediction model, we detected 4 fasting glucose-specific SNPs in GCK, PCSK1, and MTNR1B and 4 lifestyles, including smoking, aging, prolonged lifetime exposure to endogenous estrogen, and high fat intake, as the most predictive markers of CRC risk. Our joint test for those risk genotypes and lifestyles with smoking revealed the synergistically increased CRC risk, more substantially in women with longer-term exposure to cigarette smoking. DISCUSSION Our findings may improve CRC prediction ability among medically underrepresented AA women and highlight genetically informed preventive interventions (e.g., smoking cessation; CRC screening to longer-term smokers) for those women at high risk with risk genotypes and behavioral patterns.
Collapse
Affiliation(s)
- Su Yon Jung
- Translational Sciences Section, School of Nursing, University of California, Los Angeles, Los Angeles, California, USA; and
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
16
|
Gomes PRL, Vilas-Boas EA, Leite EDA, Munhoz AC, Lucena CF, Amaral FGD, Carpinelli AR, Cipolla-Neto J. Melatonin regulates maternal pancreatic remodeling and B-cell function during pregnancy and lactation. J Pineal Res 2021; 71:e12717. [PMID: 33460489 DOI: 10.1111/jpi.12717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 01/23/2023]
Abstract
The endocrine pancreas of pregnant rats shows evident plasticity, which allows the morphological structures to return to the nonpregnant state right after delivery. Furthermore, it is well-known the role of melatonin in the maintenance of the endocrine pancreas and its tropism. Studies indicate increasing nocturnal serum concentrations of maternal melatonin during pregnancy in both humans and rodents. The present study investigated the role of melatonin on energy metabolism and in pancreatic function and remodeling during pregnancy and early lactation in rats. The results confirm that the absence of melatonin during pregnancy impairs glucose metabolism. In addition, there is a dysregulation in insulin secretion at various stages of the development of pregnancy and an apparent failure in the glucose-stimulated insulin secretion during the lactation period, evidencing the role of melatonin on the regulation of insulin secretion. This mechanism seems not to be dependent on the antioxidant effect of melatonin and probably dependent on MT2 receptors. We also observed changes in the mechanisms of death and cell proliferation at the end of pregnancy and beginning of lactation, crucial periods for pancreatic remodeling. The present observations strongly suggest that both functionality and remodeling of the endocrine pancreas are impaired in the absence of melatonin and its adequate replacement, mimicking the physiological increase seen during pregnancy, is able to reverse some of the damage observed. Thus, we conclude that pineal melatonin is important to metabolic adaptation to pregnancy and both the functionality of the beta cells and the remodeling of the pancreas during pregnancy and early lactation, ensuring the return to nonpregnancy conditions.
Collapse
Affiliation(s)
| | - Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Eduardo de Almeida Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Camila Ferraz Lucena
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
The Melatonin Receptor Agonist Ramelteon Induces Cardioprotection that Requires MT2 Receptor Activation and Release of Reactive Oxygen Species. Cardiovasc Drugs Ther 2020; 34:303-310. [PMID: 32236860 PMCID: PMC7242242 DOI: 10.1007/s10557-020-06972-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Purpose The melatonin receptor (MT) agonist ramelteon has a higher affinity to MT1 than for MT2 receptors and induces cardioprotection by involvement of mitochondrial potassium channels. Activation of mitochondrial potassium channels leads to release of free radicals. We investigated whether (1) ramelteon-induced cardioprotection is MT2 receptor specific and (2) if free radicals are involved in ramelteon-induced cardioprotection. Methods Hearts of male Wistar rats were randomized, placed on a Langendorff system, and perfused with Krebs-Henseleit buffer at a constant pressure of 80 mmHg. All hearts were subjected to 33 min of global ischemia and 60 min of reperfusion. Before ischemia hearts were perfused with ramelteon (Ram) with or without the MT2 receptor inhibitor 4-phenyl-2-propionamidotetralin (4P-PDOT+Ram, 4P-PDOT). In subsequent experiments, ramelteon was administered together with the radical oxygen species (ROS) scavenger N-2-mercaptopropionylglycine (MPG+Ram). To determine whether the blockade of ramelteon-induced cardioprotection can be restored, we combined ramelteon and MPG with mitochondrial permeability transition pore (mPTP) inhibitor cyclosporine A (CsA) at different time points. Infarct size was determined by triphenyltetrazolium chloride (TTC) staining. Results Ramelteon-induced infarct size reduction was completely blocked by 4P-PDOT and MPG. Ramelteon and MPG combined with CsA before ischemia were not cardioprotective but CsA at the onset of reperfusion could restore infarct size reduction. Conclusions This study shows for the first time that despite the higher affinity to MT1 receptors, (1) ramelteon-induced cardioprotection involves MT2 receptors, (2) cardioprotection requires ROS release, and (3) inhibition of the mPTP can restore infarct size reduction.
Collapse
|
18
|
Tan X, Ciuculete DM, Schiöth HB, Benedict C. Associations between chronotype, MTNR1B genotype and risk of type 2 diabetes in UK Biobank. J Intern Med 2020; 287:189-196. [PMID: 31623012 PMCID: PMC7003850 DOI: 10.1111/joim.12994] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/14/2019] [Accepted: 10/10/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To examine the association between the MTNR1B G risk allele, type 2 diabetes (T2D) and chronotype in the UK Biobank. METHODS Data from the baseline investigation of the UK Biobank were utilized (n = 337 083 White British; mean age: 56.9 years; 54% women). MTNR1B rs10830963 was directly genotyped [CC (reference group), CG and GG]. Chronotype was divided into four categories: definitely morning (reference group); more morning than evening; more evening than morning; and definitely evening. Logistic regression analyses were performed to estimate odds ratios and 95% confidence intervals (CIs) for T2D, controlling for age, sex and other confounders. RESULTS Carriers of the rs10830963 risk allele had a higher risk of T2D [CG vs. CC: OR (95% CI) 1.10 (1.07, 1.15); GG vs. CC: 1.21 (1.14, 1.29)]. Compared with definitely morning chronotype, participants with definitely evening chronotype exhibited the highest risk of T2D [1.25 (1.17, 1.33)]. Despite a nonsignificant interaction between chronotype and the risk allele [0.98 (0.94, 1.01), P = 0.176 for interaction term], we found that definitely evening chronotype (vs. definitely morning) was linked with a higher risk of T2D amongst CC and CG but not GG carriers. Additionally, we saw that the GG genotype (vs. CC) was associated with a higher risk of T2D across all chronotype categories, except for definitely evening. CONCLUSION Our findings suggest that the MTNR1B G risk allele and late chronotype increase the risk of T2D. The association between late chronotype and higher risk of T2D appears to vary across MTNR1B rs10830963 genotypes.
Collapse
Affiliation(s)
- X Tan
- From the, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - D-M Ciuculete
- From the, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - H B Schiöth
- From the, Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - C Benedict
- From the, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
Maryam A, Vedithi SC, Khalid RR, Alsulami AF, Torres PHM, Siddiqi AR, Blundell TL. The Molecular Organization of Human cGMP Specific Phosphodiesterase 6 (PDE6): Structural Implications of Somatic Mutations in Cancer and Retinitis Pigmentosa. Comput Struct Biotechnol J 2019; 17:378-389. [PMID: 30962868 PMCID: PMC6434069 DOI: 10.1016/j.csbj.2019.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/24/2019] [Accepted: 03/03/2019] [Indexed: 01/12/2023] Open
Abstract
In the cyclic guanosine monophosphate (cGMP) signaling pathway, phosphodiesterase 6 (PDE6) maintains a critical balance of the intracellular concentration of cGMP by catalyzing it to 5′ guanosine monophosphate (5′-GMP). To gain insight into the mechanistic impacts of the PDE6 somatic mutations that are implicated in cancer and retinitis pigmentosa, we first defined the structure and organization of the human PDE6 heterodimer using computational comparative modelling. Each subunit of PDE6αβ possesses three domains connected through long α-helices. The heterodimer model indicates that the two chains are likely related by a pseudo two-fold axis. The N-terminal region of each subunit is comprised of two allosteric cGMP-binding domains (Gaf-A & Gaf-B), oriented in the same way and interacting with the catalytic domain present at the C-terminal in a way that would allow the allosteric cGMP-binding domains to influence catalytic activity. Subsequently, we applied an integrated knowledge-driven in silico mutation analysis approach to understand the structural and functional implications of experimentally identified mutations that cause various cancers and retinitis pigmentosa, as well as computational saturation mutagenesis of the dimer interface and cGMP-binding residues of both Gaf-A, and the catalytic domains. We studied the impact of mutations on the stability of PDE6αβ structure, subunit-interfaces and Gaf-cGMP interactions. Further, we discussed the changes in interatomic interactions of mutations that are destabilizing in Gaf-A (R93L, V141 M, F162 L), catalytic domain (D600N, F742 L, F776 L) and at the dimer interface (F426A, F248G, F424 N). This study establishes a possible link of change in PDE6αβ structural stability to the experimentally observed disease phenotypes.
Collapse
Affiliation(s)
- Arooma Maryam
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, Pakistan.,Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge CB2 1GA, UK
| | | | - Rana Rehan Khalid
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, Pakistan
| | - Ali F Alsulami
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge CB2 1GA, UK
| | | | - Abdul Rauf Siddiqi
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, Pakistan
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge CB2 1GA, UK
| |
Collapse
|
20
|
Abstract
Despite considerable advances in the past few years, obesity and type 2 diabetes mellitus (T2DM) remain two major challenges for public health systems globally. In the past 9 years, genome-wide association studies (GWAS) have established a major role for genetic variation within the MTNR1B locus in regulating fasting plasma levels of glucose and in affecting the risk of T2DM. This discovery generated a major interest in the melatonergic system, in particular the melatonin MT2 receptor (which is encoded by MTNR1B). In this Review, we discuss the effect of melatonin and its receptors on glucose homeostasis, obesity and T2DM. Preclinical and clinical post-GWAS evidence of frequent and rare variants of the MTNR1B locus confirmed its importance in regulating glucose homeostasis and T2DM risk with minor effects on obesity. However, these studies did not solve the question of whether melatonin is beneficial or detrimental, an issue that will be discussed in the context of the peculiarities of the melatonergic system. Melatonin receptors might have therapeutic potential as they belong to the highly druggable G protein-coupled receptor superfamily. Clarifying the precise role of melatonin and its receptors on glucose homeostasis is urgent, as melatonin is widely used for other indications, either as a prescribed medication or as a supplement without medical prescription, in many countries in Europe and in the USA.
Collapse
Affiliation(s)
- Angeliki Karamitri
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France.
- CNRS UMR 8104, Paris, France.
- Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
21
|
Tse LH, Wong YH. GPCRs in Autocrine and Paracrine Regulations. Front Endocrinol (Lausanne) 2019; 10:428. [PMID: 31354618 PMCID: PMC6639758 DOI: 10.3389/fendo.2019.00428] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest superfamily of integral membrane protein receptors. As signal detectors, the several 100 known GPCRs are responsible for sensing the plethora of endogenous ligands that are critical for the functioning of our endocrine system. Although GPCRs are typically considered as detectors for first messengers in classical signal transduction pathways, they seldom operate in isolation in complex biological systems. Intercellular communication between identical or different cell types is often mediated by autocrine or paracrine signals that are generated upon activation of specific GPCRs. In the context of energy homeostasis, the distinct complement of GPCRs in each cell type bridges the autocrine and paracrine communication within an organ, and the various downstream signaling mechanisms regulated by GPCRs can be integrated in a cell to produce an ultimate output. GPCRs thus act as gatekeepers that coordinate and fine-tune a response. By examining the role of GPCRs in activating and receiving autocrine and paracrine signals, one may have a better understanding of endocrine diseases that are associated with GPCR mutations, thereby providing new insights for treatment regimes.
Collapse
Affiliation(s)
- Lap Hang Tse
- Division of Life Science, Biotechnology Research Institute, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Yung Hou Wong
- Division of Life Science, Biotechnology Research Institute, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
- State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
- *Correspondence: Yung Hou Wong
| |
Collapse
|
22
|
Tamtaji OR, Mirhosseini N, Reiter RJ, Behnamfar M, Asemi Z. Melatonin and pancreatic cancer: Current knowledge and future perspectives. J Cell Physiol 2018; 234:5372-5378. [PMID: 30229898 DOI: 10.1002/jcp.27372] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer has a high mortality rate due to the absence of early symptoms and subsequent late diagnosis; additionally, pancreatic cancer has a high resistance to radio- and chemotherapy. Multiple inflammatory pathways are involved in the pathophysiology of pancreatic cancer. Melatonin an indoleamine produced in the pineal gland mediated and receptor-independent action is the pancreas and other where has both receptors. Melatonin is a potent antioxidant and tissue protector against inflammation and oxidative stress. In vivo and in vitro studies have shown that melatonin supplementation is an appropriate therapeutic approach for pancreatic cancer. Melatonin may be an effective apoptosis inducer in cancer cells through regulation of a large number of molecular pathways including oxidative stress, heat shock proteins, and vascular endothelial growth factor. Limited clinical studies, however, have evaluated the role of melatonin in pancreatic cancer. This review summarizes what is known regarding the effects of melatonin on pancreatic cancer and the mechanisms involved.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science, Center, San Antonio, Texas
| | - Morteza Behnamfar
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
23
|
Melatonin Uptake by Cells: An Answer to Its Relationship with Glucose? Molecules 2018; 23:molecules23081999. [PMID: 30103453 PMCID: PMC6222335 DOI: 10.3390/molecules23081999] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 02/06/2023] Open
Abstract
Melatonin, N-acetyl-5-methoxytryptamine, is an indole mainly synthesized from tryptophan in the pineal gland and secreted exclusively during the night in all the animals reported to date. While the pineal gland is the major source responsible for this night rise, it is not at all the exclusive production site and many other tissues and organs produce melatonin as well. Likewise, melatonin is not restricted to vertebrates, as its presence has been reported in almost all the phyla from protozoa to mammals. Melatonin displays a large set of functions including adaptation to light: dark cycles, free radical scavenging ability, antioxidant enzyme modulation, immunomodulatory actions or differentiation–proliferation regulatory effects, among others. However, in addition to those important functions, this evolutionary ‘ancient’ molecule still hides further tools with important cellular implications. The major goal of the present review is to discuss the data and experiments that have addressed the relationship between the indole and glucose. Classically, the pineal gland and a pinealectomy were associated with glucose homeostasis even before melatonin was chemically isolated. Numerous reports have provided the molecular components underlying the regulatory actions of melatonin on insulin secretion in pancreatic beta-cells, mainly involving membrane receptors MTNR1A/B, which would be partially responsible for the circadian rhythmicity of insulin in the organism. More recently, a new line of evidence has shown that glucose transporters GLUT/SLC2A are linked to melatonin uptake and its cellular internalization. Beside its binding to membrane receptors, melatonin transportation into the cytoplasm, required for its free radical scavenging abilities, still generates a great deal of debate. Thus, GLUT transporters might constitute at least one of the keys to explain the relationship between glucose and melatonin. These and other potential mechanisms responsible for such interaction are also discussed here.
Collapse
|
24
|
Al-Sarraf IAK, Kasabri V, Akour A, Naffa R. Melatonin and cryptochrome 2 in metabolic syndrome patients with or without diabetes: a cross-sectional study. Horm Mol Biol Clin Investig 2018; 35:/j/hmbci.ahead-of-print/hmbci-2018-0016/hmbci-2018-0016.xml. [PMID: 29813030 DOI: 10.1515/hmbci-2018-0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/25/2018] [Indexed: 12/19/2022]
Abstract
Background Metabolic syndrome (MetS) is a cluster of metabolic risk factors which increases the chances for future cardiovascular diseases, as well as diabetes. The underlying causes of MetS include overweight and obesity, physical inactivity and genetic factors. Our intension here was to focus in this study on the importance of the chronobiology, represented by melatonin (MT) and cryptochrome 2 (CRY2), in developing MetS and type 2 diabetes mellitus (T2DM). Thus, we aimed to compare MT and CRY2 plasma levels and correlate both biomarkers with adiposity, atherogenicity and hematological indices in MetS and T2DM cohorts. Methods In a cross-sectional study, 28 normoglycemic lean subjects (controls), 29 normoglycemic MetS subjects and 30 MetS (pre-diabetic/diabetic) were recruited. Results MT (pg/mL) was elevated significantly in MetS arm p-value < 0.05, whereas CRY2 levels (ng/mL) were markedly higher in both MetS groups (non-diabetic and pre-diabetic/diabetic) (all with p-value < 0.001). A reciprocal MT-CRY2 relationship was observed in the MetS (non-diabetic) group (p-value = 0.003). Of note in the total study population, both MT and CRY2 proportionally correlated with each of the following: atherogenicity index of plasma (AIP), waist circumference (WC) and systolic blood pressure (SBP) (all with p-value < 0.05) for MT and CRY2, respectively). Whereas MT correlated inversely with high-density lipoprotein-cholesterol (HDL-C) (p-value < 0.05). Additionally, CRY2 correlated directly with each of the following: diastolic blood pressure (DBP), total cholesterol (TC), low-density lipoprotein (LDL-C), hip circumference (HC), body adiposity index (BAI), weight-to-height (WHtR) ratio, mean platelet volume (MPV) and platelet/lymphocyte ratio (PLR) (p-value < 0.05). Conclusion These findings substantiate that both metabolic risk biomarkers can be prognostic tools and pharmacotherapeutic targets to slowdown the accelerated nature of T2DM.
Collapse
Affiliation(s)
| | - Violet Kasabri
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan, Phone: +96265355000, Fax: +9625300520
| | - Amal Akour
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Randa Naffa
- School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
25
|
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
|
26
|
Forrestel AC, Miedlich SU, Yurcheshen M, Wittlin SD, Sellix MT. Chronomedicine and type 2 diabetes: shining some light on melatonin. Diabetologia 2017; 60:808-822. [PMID: 27981356 DOI: 10.1007/s00125-016-4175-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/18/2016] [Indexed: 12/19/2022]
Abstract
In mammals, the circadian timing system drives rhythms of physiology and behaviour, including the daily rhythms of feeding and activity. The timing system coordinates temporal variation in the biochemical landscape with changes in nutrient intake in order to optimise energy balance and maintain metabolic homeostasis. Circadian disruption (e.g. as a result of shift work or jet lag) can disturb this continuity and increase the risk of cardiometabolic disease. Obesity and metabolic disease can also disturb the timing and amplitude of the clock in multiple organ systems, further exacerbating disease progression. As our understanding of the synergy between the timing system and metabolism has grown, an interest has emerged in the development of novel clock-targeting pharmaceuticals or nutraceuticals for the treatment of metabolic dysfunction. Recently, the pineal hormone melatonin has received some attention as a potential chronotherapeutic drug for metabolic disease. Melatonin is well known for its sleep-promoting effects and putative activity as a chronobiotic drug, stimulating coordination of biochemical oscillations through targeting the internal timing system. Melatonin affects the insulin secretory activity of the pancreatic beta cell, hepatic glucose metabolism and insulin sensitivity. Individuals with type 2 diabetes mellitus have lower night-time serum melatonin levels and increased risk of comorbid sleep disturbances compared with healthy individuals. Further, reduced melatonin levels, and mutations and/or genetic polymorphisms of the melatonin receptors are associated with an increased risk of developing type 2 diabetes. Herein we review our understanding of molecular clock control of glucose homeostasis, detail the influence of circadian disruption on glucose metabolism in critical peripheral tissues, explore the contribution of melatonin signalling to the aetiology of type 2 diabetes, and discuss the pros and cons of melatonin chronopharmacotherapy in disease management.
Collapse
Affiliation(s)
- Andrew C Forrestel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Susanne U Miedlich
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Michael Yurcheshen
- UR Medicine Sleep Center, Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steven D Wittlin
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Michael T Sellix
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA.
| |
Collapse
|
27
|
Simões D, Riva P, Peliciari-Garcia RA, Cruzat VF, Graciano MF, Munhoz AC, Taneda M, Cipolla-Neto J, Carpinelli AR. Melatonin modifies basal and stimulated insulin secretion via NADPH oxidase. J Endocrinol 2016; 231:235-244. [PMID: 27803236 DOI: 10.1530/joe-16-0259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/04/2016] [Indexed: 01/10/2023]
Abstract
Melatonin is a hormone synthesized in the pineal gland, which modulates several functions within the organism, including the synchronization of glucose metabolism and glucose-stimulated insulin secretion (GSIS). Melatonin can mediate different signaling pathways in pancreatic islets through two membrane receptors and via antioxidant or pro-oxidant enzymes modulation. NADPH oxidase (NOX) is a pro-oxidant enzyme responsible for the production of the reactive oxygen specie (ROS) superoxide, generated from molecular oxygen. In pancreatic islets, NOX-derived ROS can modulate glucose metabolism and regulate insulin secretion. Considering the roles of both melatonin and NOX in islets, the aim of this study was to evaluate the association of NOX and ROS production on glucose metabolism, basal and GSIS in pinealectomized rats (PINX) and in melatonin-treated isolated pancreatic islets. Our results showed that ROS content derived from NOX activity was increased in PINX at baseline (2.8 mM glucose), which was followed by a reduction in glucose metabolism and basal insulin secretion in this group. Under 16.7 mM glucose, an increase in both glucose metabolism and GSIS was observed in PINX islets, without changes in ROS content. In isolated pancreatic islets from control animals incubated with 2.8 mM glucose, melatonin treatment reduced ROS content, whereas in 16.7 mM glucose, melatonin reduced ROS and GSIS. In conclusion, our results demonstrate that both basal and stimulated insulin secretion can be regulated by melatonin through the maintenance of ROS homeostasis in pancreatic islets.
Collapse
Affiliation(s)
- Daniel Simões
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Patrícia Riva
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Rodrigo Antonio Peliciari-Garcia
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
- Department of Biological SciencesLaboratory of Biosystems, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Vinicius Fernandes Cruzat
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Maria Fernanda Graciano
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Munhoz
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Marco Taneda
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - José Cipolla-Neto
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and BiophysicsInstitute of Biomedical Sciences-I, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
28
|
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: 298] [Impact Index Per Article: 33.1] [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
|
29
|
Lane JM, Chang AM, Bjonnes AC, Aeschbach D, Anderson C, Cade BE, Cain SW, Czeisler CA, Gharib SA, Gooley JJ, Gottlieb DJ, Grant SFA, Klerman EB, Lauderdale DS, Lockley SW, Munch M, Patel S, Punjabi NM, Rajaratnam SMW, Rueger M, St Hilaire MA, Santhi N, Scheuermaier K, Van Reen E, Zee PC, Shea SA, Duffy JF, Buxton OM, Redline S, Scheer FAJL, Saxena R. Impact of Common Diabetes Risk Variant in MTNR1B on Sleep, Circadian, and Melatonin Physiology. Diabetes 2016; 65:1741-51. [PMID: 26868293 PMCID: PMC4878414 DOI: 10.2337/db15-0999] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 02/07/2016] [Indexed: 12/23/2022]
Abstract
The risk of type 2 diabetes (T2D) is increased by abnormalities in sleep quantity and quality, circadian alignment, and melatonin regulation. A common genetic variant in a receptor for the circadian-regulated hormone melatonin (MTNR1B) is associated with increased fasting blood glucose and risk of T2D, but whether sleep or circadian disruption mediates this risk is unknown. We aimed to test if MTNR1B diabetes risk variant rs10830963 associates with measures of sleep or circadian physiology in intensive in-laboratory protocols (n = 58-96) or cross-sectional studies with sleep quantity and quality and timing measures from self-report (n = 4,307-10,332), actigraphy (n = 1,513), or polysomnography (n = 3,021). In the in-laboratory studies, we found a significant association with a substantially longer duration of elevated melatonin levels (41 min) and delayed circadian phase of dim-light melatonin offset (1.37 h), partially mediated through delayed offset of melatonin synthesis. Furthermore, increased T2D risk in MTNR1B risk allele carriers was more pronounced in early risers versus late risers as determined by 7 days of actigraphy. Our results provide the surprising insight that the MTNR1B risk allele influences dynamics of melatonin secretion, generating a novel hypothesis that the MTNR1B risk allele may extend the duration of endogenous melatonin production later into the morning and that early waking may magnify the diabetes risk conferred by the risk allele.
Collapse
Affiliation(s)
- Jacqueline M Lane
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| | - Anne-Marie Chang
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Department of Biobehavioral Health, Pennsylvania State University, University Park, PA
| | - Andrew C Bjonnes
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| | - Daniel Aeschbach
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Clare Anderson
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Sean W Cain
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Charles A Czeisler
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA
| | - Joshua J Gooley
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Struan F A Grant
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | | | - Steven W Lockley
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Miriam Munch
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Sanjay Patel
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Naresh M Punjabi
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD
| | - Shanthakumar M W Rajaratnam
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Melanie Rueger
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Melissa A St Hilaire
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Nayantara Santhi
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Karin Scheuermaier
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Eliza Van Reen
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Phyllis C Zee
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Steven A Shea
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Jeanne F Duffy
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Orfeu M Buxton
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Department of Biobehavioral Health, Pennsylvania State University, University Park, PA Department of Social and Behavioral Sciences, Harvard School of Public Health, Boston, MA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Richa Saxena
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
30
|
Affiliation(s)
- Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
31
|
Li X, Cai S, Yin W, Hu X, Zhang S, Li Z, Li X, Laudon M. Role of Neu-p11/luzindole in the regulation of insulin signaling pathways and insulin resistance. Acta Biochim Biophys Sin (Shanghai) 2016; 48:485-6. [PMID: 27025603 DOI: 10.1093/abbs/gmw015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Xiuping Li
- Department of Laboratory, Hunan University of Medicine, Huaihua 418000, China
| | - Shichang Cai
- Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Weidong Yin
- Institute of Cardiovascular Disease, Key Laboratory Arteriosclerology of Hunan Province, University of South China, Hengyang 412000, China
| | - Xiaobo Hu
- Institute of Cardiovascular Disease, Key Laboratory Arteriosclerology of Hunan Province, University of South China, Hengyang 412000, China
| | - Sujun Zhang
- Department of Experimental Animal, University of South China, Hengyang 412000, China
| | - Zhengming Li
- Department of Laboratory, Hunan University of Medicine, Huaihua 418000, China
| | - Xing Li
- Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Moshe Laudon
- Neurim Pharmaceuticals Ltd., Tel-Aviv 69710, Israel
| |
Collapse
|
32
|
Owino S, Contreras-Alcantara S, Baba K, Tosini G. Melatonin Signaling Controls the Daily Rhythm in Blood Glucose Levels Independent of Peripheral Clocks. PLoS One 2016; 11:e0148214. [PMID: 26824606 PMCID: PMC4732609 DOI: 10.1371/journal.pone.0148214] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/14/2016] [Indexed: 01/01/2023] Open
Abstract
Melatonin is rhythmically secreted by both the pineal gland and retina in a circadian fashion, with its peak synthesis occurring during the night. Once synthesized, melatonin exerts its effects by binding to two specific G-protein coupled receptors-melatonin receptor type 1(MT1) and melatonin receptor type 2(MT2). Recent studies suggest the involvement of MT1 and MT2 in the regulation of glucose homeostasis; however the ability of melatonin signaling to impart timing cues on glucose metabolism remains poorly understood. Here we report that the removal of MT1 or MT2 in mice abolishes the daily rhythm in blood glucose levels. Interestingly, removal of melatonin receptors produced small effects on the rhythmic expression patterns of clock genes within skeletal muscle, liver, and adipose tissue. Taken together, our data suggest that the loss of the daily rhythm in blood glucose observed in MT1(-/-) and MT2(-/-) mice does not occur as a consequence of 'disrupted' clocks within insulin sensitive tissues. Finally our results highlight a diurnal contribution of melatonin receptor signaling in the daily regulation of blood glucose levels.
Collapse
MESH Headings
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/metabolism
- CLOCK Proteins/genetics
- CLOCK Proteins/metabolism
- Circadian Rhythm/genetics
- Gene Expression Regulation
- Homeostasis
- Liver/metabolism
- Male
- Melatonin/metabolism
- Mice
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Pineal Gland/metabolism
- Receptor, Melatonin, MT1/deficiency
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT2/deficiency
- Receptor, Melatonin, MT2/genetics
- Retina/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Sharon Owino
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Susana Contreras-Alcantara
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Kenkichi Baba
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (KB); (GT)
| | - Gianluca Tosini
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (KB); (GT)
| |
Collapse
|
33
|
Sharma S, Singh H, Ahmad N, Mishra P, Tiwari A. The role of melatonin in diabetes: therapeutic implications. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2015; 59:391-9. [PMID: 26331226 DOI: 10.1590/2359-3997000000098] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/06/2015] [Indexed: 01/27/2023]
Abstract
Melatonin referred as the hormone of darkness is mainly secreted by pineal gland, its levels being elevated during night and low during the day. The effects of melatonin on insulin secretion are mediated through the melatonin receptors (MT1 and MT2). It decreases insulin secretion by inhibiting cAMP and cGMP pathways but activates the phospholipaseC/IP3 pathway, which mobilizes Ca2+from organelles and, consequently increases insulin secretion. Both in vivo and in vitro, insulin secretion by the pancreatic islets in a circadian manner, is due to the melatonin action on the melatonin receptors inducing a phase shift in the cells. Melatonin may be involved in the genesis of diabetes as a reduction in melatonin levels and a functional interrelationship between melatonin and insulin was observed in diabetic patients. Evidences from experimental studies proved that melatonin induces production of insulin growth factor and promotes insulin receptor tyrosine phosphorylation. The disturbance of internal circadian system induces glucose intolerance and insulin resistance, which could be restored by melatonin supplementation. Therefore, the presence of melatonin receptors on human pancreatic islets may have an impact on pharmacotherapy of type 2 diabetes.
Collapse
Affiliation(s)
- Shweta Sharma
- School of Biotechnology, Rajiv Gandhi Technical University, Bhopal, Madhya Pradesh, India
| | - Hemant Singh
- School of Biotechnology, Rajiv Gandhi Technical University, Bhopal, Madhya Pradesh, India
| | - Nabeel Ahmad
- School of Biotechnology, IFTM University, Uttar Pradesh, India
| | - Priyanka Mishra
- School of Biotechnology, Rajiv Gandhi Technical University, Bhopal, Madhya Pradesh, India
| | - Archana Tiwari
- School of Biotechnology, Rajiv Gandhi Technical University, Bhopal, Madhya Pradesh, India
| |
Collapse
|
34
|
Peschke E, Bähr I, Mühlbauer E. Experimental and clinical aspects of melatonin and clock genes in diabetes. J Pineal Res 2015; 59:1-23. [PMID: 25904189 DOI: 10.1111/jpi.12240] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 12/15/2022]
Abstract
The pineal hormone melatonin influences insulin secretion, as well as glucagon and somatostatin secretion, both in vivo and in vitro. These effects are mediated by two specific, high-affinity, seven transmembrane, pertussis toxin-sensitive, Gi-protein-coupled melatonin receptors, MT1 and MT2. Both isoforms are expressed in the β-cells, α-cells as well as δ-cells of the pancreatic islets of Langerhans and are involved in the modulation of insulin secretion, leading to inhibition of the adenylate cyclase-dependent cyclic adenosine monophosphate as well as cyclic guanosine monophosphate formation in pancreatic β-cells by inhibiting the soluble guanylate cyclase, probably via MT2 receptors. In this way, melatonin also likely inhibits insulin secretion, whereas using the inositol triphosphate pathway after previous blocking of Gi-proteins by pertussis toxin, melatonin increases insulin secretion. Desynchrony of receptor signaling may lead to the development of type 2 diabetes. This notion has recently been supported by genomewide association studies pinpointing variances of the MT2 receptor as a risk factor for this rapidly spreading metabolic disturbance. As melatonin is secreted in a clearly diurnal fashion, it is safe to assume that it also has a diurnal impact on the blood-glucose-regulating function of the islet. Observations of the circadian expression of clock genes (Clock, Bmal1, Per1,2,3, and Cry1,2) in pancreatic islets, as well as in INS1 rat insulinoma cells, may indicate that circadian rhythms are generated in the β-cells themselves. The circadian secretion of insulin from pancreatic islets is clock-driven. Disruption of circadian rhythms and clock function leads to metabolic disturbances, for example, type 2 diabetes. The study of melatonin-insulin interactions in diabetic rat models has revealed an inverse relationship between these two hormones. Both type 2 diabetic rats and patients exhibit decreased melatonin levels and slightly increased insulin levels, whereas type 1 diabetic rats show extremely reduced levels or the absence of insulin, but statistically significant increases in melatonin levels. Briefly, an increase in melatonin levels leads to a decrease in stimulated insulin secretion and vice versa. Melatonin levels in blood plasma, as well as the activity of the key enzyme of melatonin synthesis, AA-NAT (arylalkylamine-N-acetyltransferase) in pineal, are lower in type 2 diabetic rats compared to controls. In contrast, melatonin and pineal AA-NAT mRNA are increased and insulin receptor mRNA is decreased in type 1 diabetic rats, which also indicates a close relationship between insulin and melatonin. As an explanation, it was hypothesized that catecholamines, which reduce insulin levels and stimulate melatonin synthesis, control insulin-melatonin interactions. This conviction stems from the observation that catecholamines are increased in type 1 but are diminished in type 2 diabetes. In this context, another important line of inquiry involves the fact that melatonin protects β-cells against functional overcharge and, consequently, hinders the development of type 2 diabetes.
Collapse
Affiliation(s)
| | - Ina Bähr
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | |
Collapse
|
35
|
Salman M, Dasgupta S, Cholendra A, Venugopal PN, Lakshmi GL, Xaviour D, Rao J, D'Souza CJM. MTNR1B gene polymorphisms and susceptibility to Type 2 Diabetes: A pilot study in South Indians. Gene 2015; 566:189-93. [PMID: 25922310 DOI: 10.1016/j.gene.2015.04.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/26/2015] [Accepted: 04/17/2015] [Indexed: 01/23/2023]
Abstract
Type 2 Diabetes (T2D) is the major health concern in the Indian subcontinent. A genome-wide association study carried out with non-diabetic Indians showed association of MTNR1B variants with fasting glucose. MTNR1B mediates the effect of melatonin on insulin secretion. In light of the growing importance of MTNR1B in the etiology of T2D, we sought to test its association with the disease in the south Indian type 2 diabetics. Five single nucleotide polymorphisms of MTNR1B (rs10830962, rs10830963, rs3847554, rs1387153 and rs2166706) were genotyped in 346 T2D patients and 341 non-diabetic controls. None of the SNPs differed significantly between patients and controls with respect to allele and genotype frequencies. Linear regression analysis after adjustment for age, sex and BMI showed a significant positive association of rs3847554 with fasting glucose under recessive model (β=14.98, p=0.012). Haplotypes constituted by minor alleles of rs3847554, rs1387153, rs2166706, rs10830963 and major allele of rs10830962 showed significant positive correlation with fasting glucose (p<0.05). Though the results obtained are suggestive of MTNR1B role in T2D etiology, they need to be confirmed with much larger sample sizes.
Collapse
Affiliation(s)
- Mohammed Salman
- Anthropological Survey of India, Southern Regional Centre, Mysore 570026, Karnataka, India; Department of Studies in Biochemistry, University of Mysore, Mysore 570006, Karnataka, India.
| | - Shruti Dasgupta
- Department of Studies in Biotechnology, University of Mysore, Mysore 570006, Karnataka, India.
| | - A Cholendra
- Department of Anthropology, Division of Human Genetics, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India.
| | - P N Venugopal
- Anthropological Survey of India, North-West Regional Center, Dehradun 248 195, India.
| | - G L Lakshmi
- Anthropological Survey of India, Southern Regional Centre, Mysore 570026, Karnataka, India.
| | - D Xaviour
- Anthropological Survey of India, Southern Regional Centre, Mysore 570026, Karnataka, India.
| | - Jayashankar Rao
- Anthropological Survey of India, Southern Regional Centre, Mysore 570026, Karnataka, India.
| | - Cletus J M D'Souza
- Department of Studies in Biochemistry, University of Mysore, Mysore 570006, Karnataka, India.
| |
Collapse
|
36
|
Zibolka J, Mühlbauer E, Peschke E. Melatonin influences somatostatin secretion from human pancreatic δ-cells via MT1 and MT2 receptors. J Pineal Res 2015; 58:198-209. [PMID: 25585597 DOI: 10.1111/jpi.12206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/09/2015] [Indexed: 12/20/2022]
Abstract
Melatonin is an effector of the diurnal clock on pancreatic islets. The membrane receptor-transmitted inhibitory influence of melatonin on insulin secretion is well established and contrasts with the reported stimulation of glucagon release from α-cells. Virtually, nothing is known concerning the melatonin-mediated effects on islet δ-cells. Analysis of a human pancreatic δ-cell model, the cell line QGP-1, and the use of a somatostatin-specific radioimmunoassay showed that melatonin primarily has an inhibitory effect on somatostatin secretion in the physiological concentration range. In the pharmacological range, melatonin elicited slightly increased somatostatin release from δ-cells. Cyclic adenosine monophosphate (cAMP) is the major second messenger dose-dependently stimulating somatostatin secretion, in experiments employing the membrane-permeable 8-Br-cAMP. 8-Br-cyclic guanosine monophosphate proved to be of only minor relevance to somatostatin release. As the inhibitory effect of 1 nm melatonin was reversed after incubation of QGP-1 cells with the nonselective melatonin receptor antagonist luzindole, but not with the MT2-selective antagonist 4-P-PDOT (4-phenyl-2-propionamidotetraline), an involvement of the MT1 receptor can be assumed. Somatostatin release from the δ-cells at low glucose concentrations was significantly inhibited during co-incubation with 1 nm melatonin, an effect which was less pronounced at higher glucose levels. Transient expression experiments, overexpressing MT1, MT2, or a deletion variant as a control, indicated that the MT1 and not the MT2 receptor was the major transmitter of the inhibitory melatonin effect. These data point to a significant influence of melatonin on pancreatic δ-cells and on somatostatin release.
Collapse
Affiliation(s)
- Juliane Zibolka
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | | |
Collapse
|
37
|
Tosini G, Owino S, Guillaume JL, Jockers R. Understanding melatonin receptor pharmacology: latest insights from mouse models, and their relevance to human disease. Bioessays 2014; 36:778-87. [PMID: 24903552 PMCID: PMC4151498 DOI: 10.1002/bies.201400017] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Melatonin, the neuro-hormone synthesized during the night, has recently seen an unexpected extension of its functional implications toward type 2 diabetes development, visual functions, sleep disturbances, and depression. Transgenic mouse models were instrumental for the establishment of the link between melatonin and these major human diseases. Most of the actions of melatonin are mediated by two types of G protein-coupled receptors, named MT1 and MT2 , which are expressed in many different organs and tissues. Understanding the pharmacology and function of mouse MT1 and MT2 receptors, including MT1 /MT2 heteromers, will be of crucial importance to evaluate the relevance of these mouse models for future therapeutic developments. This review will critically discuss these aspects, and give some perspectives including the generation of new mouse models.
Collapse
Affiliation(s)
- Gianluca Tosini
- Neuroscience Institute and Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | | | | | | |
Collapse
|
38
|
Cipolla-Neto J, Amaral FG, Afeche SC, Tan DX, Reiter RJ. Melatonin, energy metabolism, and obesity: a review. J Pineal Res 2014; 56:371-81. [PMID: 24654916 DOI: 10.1111/jpi.12137] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 03/17/2014] [Indexed: 12/15/2022]
Abstract
Melatonin is an old and ubiquitous molecule in nature showing multiple mechanisms of action and functions in practically every living organism. In mammals, pineal melatonin functions as a hormone and a chronobiotic, playing a major role in the regulation of the circadian temporal internal order. The anti-obesogen and the weight-reducing effects of melatonin depend on several mechanisms and actions. Experimental evidence demonstrates that melatonin is necessary for the proper synthesis, secretion, and action of insulin. Melatonin acts by regulating GLUT4 expression and/or triggering, via its G-protein-coupled membrane receptors, the phosphorylation of the insulin receptor and its intracellular substrates mobilizing the insulin-signaling pathway. Melatonin is a powerful chronobiotic being responsible, in part, by the daily distribution of metabolic processes so that the activity/feeding phase of the day is associated with high insulin sensitivity, and the rest/fasting is synchronized to the insulin-resistant metabolic phase of the day. Furthermore, melatonin is responsible for the establishment of an adequate energy balance mainly by regulating energy flow to and from the stores and directly regulating the energy expenditure through the activation of brown adipose tissue and participating in the browning process of white adipose tissue. The reduction in melatonin production, as during aging, shift-work or illuminated environments during the night, induces insulin resistance, glucose intolerance, sleep disturbance, and metabolic circadian disorganization characterizing a state of chronodisruption leading to obesity. The available evidence supports the suggestion that melatonin replacement therapy might contribute to restore a more healthy state of the organism.
Collapse
Affiliation(s)
- J Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
39
|
Karamitri A, Jockers R. Exon Sequencing of G Protein-Coupled Receptor Genes and Perspectives for Disease Treatment. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-62703-779-2_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
40
|
Santofimia-Castaño P, Ruy DC, Salido GM, González A. Melatonin modulates Ca2+ mobilization and amylase release in response to cholecystokinin octapeptide in mouse pancreatic acinar cells. J Physiol Biochem 2013; 69:897-908. [PMID: 23904230 DOI: 10.1007/s13105-013-0267-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/12/2013] [Indexed: 01/01/2023]
Abstract
In the present work, we have evaluated the effect of an acute addition of melatonin on cholecystokinin octapeptide (CCK-8)-evoked Ca(2+) signals and amylase secretion in mouse pancreatic acinar cells. For this purpose, freshly isolated mouse pancreatic acinar cells were loaded with fura-2 to study intracellular free Ca(2+) concentration ([Ca(2+)](c)). Amylase release and cell viability were studied employing colorimetric methods. Our results show that CCK-8 evoked a biphasic effect on amylase secretion, finding a maximum at a concentration of 0.1 nM and a reduction of secretion at higher concentrations. Pre-incubation of cells with melatonin (1 μM-1 mM) significantly attenuated enzyme secretion in response to high concentrations of CCK-8. Stimulation of cells with 1 nM CCK-8 led to a transient increase in [Ca(2+)](c), followed by a decrease towards a constant level. In the presence of 1 mM melatonin, stimulation of cells with CCK-8 resulted in a smaller [Ca(2+)](c) peak response, a faster rate of decay of [Ca(2+)](c) and lower values for the steady state of [Ca(2+)](c), compared with the effect of CCK-8 alone. Melatonin also reduced the oscillatory pattern of Ca(2+) mobilization evoked by a physiological concentration of CCK-8 (20 pM), and completely inhibited Ca(2+) mobilization induced by 10 pM CCK-8. On the other hand, Ca(2+) entry from the extracellular space was not affected in the presence of melatonin. Finally, melatonin alone did not change cell viability. We conclude that melatonin, at concentrations higher than those found in blood, might regulate exocrine pancreatic function via modulation of Ca(2+) signals.
Collapse
Affiliation(s)
- Patricia Santofimia-Castaño
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Avenida Universidad s/n, 10003, Caceres, Spain
| | | | | | | |
Collapse
|
41
|
Li C, Qiao B, Zhan Y, Peng W, Chen ZJ, Sun L, Zhang J, Zhao L, Gao Q. Association between genetic variations in MTNR1A and MTNR1B genes and gestational diabetes mellitus in Han Chinese women. Gynecol Obstet Invest 2013; 76:221-7. [PMID: 24157813 DOI: 10.1159/000355521] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/07/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS The purpose of this study was to investigate the association between MTNR1A and MTNR1B gene polymorphisms and gestational diabetes mellitus (GDM) in Han Chinese women. METHODS Study participants included 350 patients with GDM and 480 control subjects. Three single-nucleotide polymorphisms (SNPs; rs2119882 in MTNR1A and rs10830963 and rs10830962 in MTNR1B) were genotyped using direct sequencing. Genotype and allele distributions of SNPs in cases of GDM and controls were analyzed. Association of the MTNR1A and MTNR1B gene variants with plasma glucose and insulin levels as well as blood lipid levels was further investigated. RESULTS The frequencies of genotypes and allele types of rs2119882 in MTNR1A and rs10830963 in MTNR1B were significantly different between women with GDM and controls (p < 0.05). Moreover, in the GDM group, these SNPs were associated with increased fasting plasma glucose concentrations (p < 0.001) and increased homeostasis model assessment of insulin resistance (p < 0.001). The rs10830962 polymorphism in MTNR1B was not associated with an increased risk of developing GDM or any of the clinical or metabolic characteristics in patients with GDM (p > 0.05). CONCLUSION The genetic polymorphisms rs2119882 in MTNR1A and rs10830963 in MTNR1B are associated with an increased risk of developing GDM and insulin resistance in Han Chinese women.
Collapse
Affiliation(s)
- Chao Li
- Department of Obstetrics, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Li X, Cai S, Yin W, Hu X. Neu-p11 reduces clock/apelin expression in insulin-resistant mouse adipocyte model. Acta Biochim Biophys Sin (Shanghai) 2013; 45:798-800. [PMID: 23963596 DOI: 10.1093/abbs/gmt077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
43
|
Karamitri A, Renault N, Clement N, Guillaume JL, Jockers R. Minireview: Toward the establishment of a link between melatonin and glucose homeostasis: association of melatonin MT2 receptor variants with type 2 diabetes. Mol Endocrinol 2013; 27:1217-33. [PMID: 23798576 DOI: 10.1210/me.2013-1101] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The existence of interindividual variations in G protein-coupled receptor sequences has been recognized early on. Recent advances in large-scale exon sequencing techniques are expected to dramatically increase the number of variants identified in G protein-coupled receptors, giving rise to new challenges regarding their functional characterization. The current minireview will illustrate these challenges based on the MTNR1B gene, which encodes the melatonin MT2 receptor, for which exon sequencing revealed 40 rare nonsynonymous variants in the general population and in type 2 diabetes (T2D) cohorts. Functional characterization of these MT2 mutants revealed 14 mutants with loss of Gi protein activation that associate with increased risk of T2D development. This repertoire of disease-associated mutants is a rich source for structure-activity studies and will help to define the still poorly understood role of melatonin in glucose homeostasis and T2D development in humans. Defining the functional defects in carriers of rare MT2 mutations will help to provide personalized therapies to these patients in the future.
Collapse
Affiliation(s)
- Angeliki Karamitri
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France
| | | | | | | | | |
Collapse
|
44
|
Lin GJ, Huang SH, Chen SJ, Wang CH, Chang DM, Sytwu HK. Modulation by melatonin of the pathogenesis of inflammatory autoimmune diseases. Int J Mol Sci 2013; 14:11742-66. [PMID: 23727938 PMCID: PMC3709754 DOI: 10.3390/ijms140611742] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 12/14/2022] Open
Abstract
Melatonin is the major secretory product of the pineal gland during the night and has multiple activities including the regulation of circadian and seasonal rhythms, and antioxidant and anti-inflammatory effects. It also possesses the ability to modulate immune responses by regulation of the T helper 1/2 balance and cytokine production. Autoimmune diseases, which result from the activation of immune cells by autoantigens released from normal tissues, affect around 5% of the population. Activation of autoantigen-specific immune cells leads to subsequent damage of target tissues by these activated cells. Melatonin therapy has been investigated in several animal models of autoimmune disease, where it has a beneficial effect in a number of models excepting rheumatoid arthritis, and has been evaluated in clinical autoimmune diseases including rheumatoid arthritis and ulcerative colitis. This review summarizes and highlights the role and the modulatory effects of melatonin in several inflammatory autoimmune diseases including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes mellitus, and inflammatory bowel disease.
Collapse
Affiliation(s)
- Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, No. 161, Section 6, MinChuan East Road, Neihu, Taipei City 114, Taiwan; E-Mail:
| | - Shing-Hwa Huang
- Department of General Surgery, Tri-Service General Hospital, No.325, Section 2, Chenggong Rd., Neihu District, Taipei City 114, Taiwan; E-Mail:
| | - Shyi-Jou Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, No. 161, Section 6, MinChuan East Road, Neihu, Taipei City 114, Taiwan; E-Mails: (S.-J.C.); (C.-H.W.)
- Department of Pediatrics, Tri-Service General Hospital, No.325, Section 2, Chenggong Rd., Neihu District, Taipei City 114, Taiwan
| | - Chih-Hung Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, No. 161, Section 6, MinChuan East Road, Neihu, Taipei City 114, Taiwan; E-Mails: (S.-J.C.); (C.-H.W.)
- Department of Otolaryngology—Head and Neck Surgery, Tri-Service General Hospital, No.325, Section 2, Chenggong Rd., Neihu District, Taipei City 114, Taiwan
- Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, No. 161, Section 6, MinChuan East Road, Neihu, Taipei City 114, Taiwan
- Department of Microbiology and Immunology, National Defense Medical Center, No. 161, Section 6, MinChuan East Road, Neihu, Taipei City 114, Taiwan
| | - Deh-Ming Chang
- Rheumatology/Immunology/Allergy, Tri-Service General Hospital, No.325, Section 2, Chenggong Rd., Neihu District, Taipei City 114, Taiwan; E-Mail:
| | - Huey-Kang Sytwu
- Department of Microbiology and Immunology, National Defense Medical Center, No. 161, Section 6, MinChuan East Road, Neihu, Taipei City 114, Taiwan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-2-8792-3100 (ext. 18540); Fax: +886-2-8792-1774
| |
Collapse
|
45
|
Melatonin and pancreatic islets: interrelationships between melatonin, insulin and glucagon. Int J Mol Sci 2013; 14:6981-7015. [PMID: 23535335 PMCID: PMC3645673 DOI: 10.3390/ijms14046981] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 03/07/2013] [Accepted: 03/11/2013] [Indexed: 12/15/2022] Open
Abstract
The pineal hormone melatonin exerts its influence in the periphery through activation of two specific trans-membrane receptors: MT1 and MT2. Both isoforms are expressed in the islet of Langerhans and are involved in the modulation of insulin secretion from β-cells and in glucagon secretion from α-cells. De-synchrony of receptor signaling may lead to the development of type 2 diabetes. This notion has recently been supported by genome-wide association studies identifying particularly the MT2 as a risk factor for this rapidly spreading metabolic disturbance. Since melatonin is secreted in a clearly diurnal fashion, it is safe to assume that it also has a diurnal impact on the blood-glucose-regulating function of the islet. This factor has hitherto been underestimated; the disruption of diurnal signaling within the islet may be one of the most important mechanisms leading to metabolic disturbances. The study of melatonin–insulin interactions in diabetic rat models has revealed an inverse relationship: an increase in melatonin levels leads to a down-regulation of insulin secretion and vice versa. Elucidation of the possible inverse interrelationship in man may open new avenues in the therapy of diabetes.
Collapse
|
46
|
Abstract
Circadian rhythms are ubiquitous in biological systems and regulate metabolic processes throughout the body. Misalliance of these circadian rhythms and the systems they regulate has a profound impact on hormone levels and increases risk of developing metabolic diseases. Melatonin, a hormone secreted by the pineal gland, is one of the major signaling molecules used by the master circadian oscillator to entrain downstream circadian rhythms. Several recent genetic studies have pointed out that a common variant in the gene that encodes the melatonin receptor 2 (MTNR1B) is associated with impaired glucose homeostasis, reduced insulin secretion, and an increased risk of developing type 2 diabetes. Here, we try to review the role of this receptor and its signaling pathways in respect to glucose homeostasis and development of the disease.
Collapse
MESH Headings
- Circadian Rhythm/genetics
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Female
- Genetic Variation
- Humans
- Insulin/blood
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells
- Male
- Melatonin/biosynthesis
- Receptor, Melatonin, MT1/blood
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT2/blood
- Receptor, Melatonin, MT2/genetics
- Risk Factors
- Signal Transduction
Collapse
Affiliation(s)
- Cecilia Nagorny
- Unit of Molecular Metabolism, Department of Clinical Sciences in Malmoe, Lund University Diabetes Centre, 20502, Malmoe, Sweden.
| | | |
Collapse
|
47
|
Bähr I, Mühlbauer E, Albrecht E, Peschke E. Evidence of the receptor-mediated influence of melatonin on pancreatic glucagon secretion via the Gαq protein-coupled and PI3K signaling pathways. J Pineal Res 2012; 53:390-8. [PMID: 22672634 DOI: 10.1111/j.1600-079x.2012.01009.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Melatonin has been shown to modulate glucose metabolism by influencing insulin secretion. Recent investigations have also indicated a regulatory function of melatonin on the pancreatic α-cells. The present in vitro and in vivo studies evaluated whether melatonin mediates its effects via melatonin receptors and which signaling cascade is involved. Incubation experiments using the glucagon-producing mouse pancreatic α-cell line αTC1 clone 9 (αTC1.9) as well as isolated pancreatic islets of rats and mice revealed that melatonin increases glucagon secretion. Preincubation of αTC1.9 cells with the melatonin receptor antagonists luzindole and 4P-PDOT abolished the glucagon-stimulatory effect of melatonin. In addition, glucagon secretion was lower in the pancreatic islets of melatonin receptor knockout mice than in the islets of the wild-type (WT) control animals. Investigations of melatonin receptor knockout mice revealed decreased plasma glucagon concentrations and elevated mRNA expression levels of the hepatic glucagon receptor when compared to WT mice. Furthermore, studies using pertussis toxin, as well as measurements of cAMP concentrations, ruled out the involvement of Gαi- and Gαs-coupled signaling cascades in mediating the glucagon increase induced by melatonin. In contrast, inhibition of phospholipase C in αTC1.9 cells prevented the melatonin-induced effect, indicating the physiological relevance of the Gαq-coupled pathway. Our data point to the involvement of the phosphatidylinositol 3-kinase signaling cascade in mediating melatonin effects in pancreatic α-cells. In conclusion, these findings provide evidence that the glucagon-stimulatory effect of melatonin in pancreatic α-cells is melatonin receptor mediated, thus supporting the concept of melatonin-modulated and diurnal glucagon release.
Collapse
MESH Headings
- Animals
- Cell Line
- Cyclic AMP/metabolism
- Diabetes Mellitus, Type 2/enzymology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Gene Expression Regulation
- Glucagon/blood
- Glucagon/metabolism
- Glucagon-Secreting Cells/drug effects
- Glucagon-Secreting Cells/enzymology
- Glucagon-Secreting Cells/metabolism
- Liver/drug effects
- Liver/metabolism
- Male
- Melanins/pharmacology
- Mice
- Mice, Knockout
- Pertussis Toxin/pharmacology
- Phosphatidylinositol 3-Kinase/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptor, Melatonin, MT1/deficiency
- Receptor, Melatonin, MT1/drug effects
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT2/deficiency
- Receptor, Melatonin, MT2/drug effects
- Receptor, Melatonin, MT2/genetics
- Receptors, Glucagon/drug effects
- Receptors, Glucagon/genetics
- Receptors, Glucagon/metabolism
- Signal Transduction/drug effects
- Tetrahydronaphthalenes/pharmacology
- Tissue Culture Techniques
- Tryptamines/pharmacology
- Type C Phospholipases/metabolism
Collapse
Affiliation(s)
- Ina Bähr
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | | | | | | |
Collapse
|
48
|
Bazwinsky-Wutschke I, Wolgast S, Mühlbauer E, Albrecht E, Peschke E. Phosphorylation of cyclic AMP-response element-binding protein (CREB) is influenced by melatonin treatment in pancreatic rat insulinoma β-cells (INS-1). J Pineal Res 2012; 53:344-57. [PMID: 22616931 DOI: 10.1111/j.1600-079x.2012.01004.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The pineal hormone melatonin exerts its influence on the insulin secretion of pancreatic islets by a variety of signalling pathways. The purpose of the present study was to analyse the impact of melatonin on the phosphorylated transcription factor cAMP-response element-binding protein (pCREB). In pancreatic rat insulinoma β-cells (INS-1), pCREB immunofluorescence intensities in cell nuclei using digitised confocal image analysis were measured to semi-quantify differences in the pCREB immunoreactivity (pCREB-ir) caused by different treatments. Increasing concentrations of forskolin or 3-isobutyl-1-methylxanthine (IBMX) resulted in a dose-dependent rise of the mean fluorescence intensity in pCREB-ir nuclear staining. Concomitant melatonin application significantly decreased pCREB-ir in INS-1 cells after 30-min, 1-hr and 3-hr treatment. The melatonin receptor antagonists luzindole and 4-phenyl-2-propionamidotetraline (4P-PDOT) completely abolished the pCREB phosphorylation-decreasing effect of melatonin, indicating that both melatonin receptor isoforms (MT(1) and MT(2)) are involved. In a transfected INS-1 cell line expressing the human MT(2) receptor, melatonin caused the greatest reduction in pCREB after IBMX treatment compared with nontransfected INS-1 cells, indicating a crucial influence of melatonin receptor density on pCREB regulation. Furthermore, the downregulation of pCREB by melatonin is concomitantly associated with a statistically significant downregulation of Camk2d transcript levels, as measured after 3 hr. In conclusion, the present study provides evidence that the phosphorylation level of CREB is modulated in pancreatic β-cells by melatonin. Mediated via CREB, melatonin regulates the expression of genes that play an important functional role in the regulation of β-cell signalling pathways.
Collapse
MESH Headings
- 1-Methyl-3-isobutylxanthine/pharmacology
- Animals
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cell Line, Tumor
- Colforsin/pharmacology
- Cyclic AMP Response Element-Binding Protein/metabolism
- Dose-Response Relationship, Drug
- Fluorescent Antibody Technique
- Humans
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulinoma/genetics
- Insulinoma/metabolism
- Melatonin/pharmacology
- Microscopy, Confocal
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Phosphorylation
- Rats
- Receptor, Melatonin, MT1/drug effects
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT2/drug effects
- Receptor, Melatonin, MT2/genetics
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction/drug effects
- Tetrahydronaphthalenes/pharmacology
- Time Factors
- Transfection
- Tryptamines/pharmacology
Collapse
|
49
|
Simsek N, Kaya M, Kara A, Can I, Karadeniz A, Kalkan Y. Effects of melatonin on islet neogenesis and beta cell apoptosis in streptozotocin-induced diabetic rats: an immunohistochemical study. Domest Anim Endocrinol 2012; 43:47-57. [PMID: 22541933 DOI: 10.1016/j.domaniend.2012.02.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 01/30/2012] [Accepted: 02/01/2012] [Indexed: 01/19/2023]
Abstract
This investigation was carried out to explore the antidiabetic, antiapoptotic and neogenetic effects of melatonin (MLT) in streptozotocin-induced diabetic rats. Sixty-four male rats were assigned randomly to one of four groups for periods of 21 and 42 d as follows; i) control, ii) MLT, iii) diabetic (DM), and iv) DM + MLT. Immunohistochemical methods were used -with pancreatic tissue to determine the intensity of insulin, caspase-3 and Bcl-x(L) immune reactivities, and new islet formation. In untreated DM rats, BW loss, increased plasma glucose and MLT concentrations, as well as cytoplasmic degranulation and vacuolization were observed. We also observed a marked increase in the number of apoptotic caspase-3 positive cells and a few insulin- positive cells, but not antiapoptotic Bcl-x(L) positive cells. Observations in the DM + MLT-treated group revealed a high intensity of insulin- and antiapoptotic Bcl-x(L) immune reactivities at 21 and 42 d. Moreover, data indicated that MLT may cause beta cell proliferation and that new small islets originate from cells associated with ductal epithelium and from centroacinar cells by day 21. These data indicate that; i) MLT treatment may stimulate neogenesis in the pancreas of diabetic rats, and ii) MLT's antiapoptotic action may increase beta cell differentiation and caspase-3 inactivation or Bcl-x(L) activation.
Collapse
Affiliation(s)
- N Simsek
- Department of Histology and Embryology, Faculty of Veterinary Sciences, Atatürk University, 25240, Erzurum, Turkey.
| | | | | | | | | | | |
Collapse
|
50
|
Mühlbauer E, Albrecht E, Bazwinsky-Wutschke I, Peschke E. Melatonin influences insulin secretion primarily via MT(1) receptors in rat insulinoma cells (INS-1) and mouse pancreatic islets. J Pineal Res 2012; 52:446-59. [PMID: 22288848 DOI: 10.1111/j.1600-079x.2012.00959.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several studies have revealed that melatonin affects the insulin secretion via MT(1) and MT(2) receptor isoforms. Owing to the lack of selective MT(1) receptor antagonists, we used RNA interference technology to generate an MT(1) knockdown in a clonal β-cell line to evaluate whether melatonin modulates insulin secretion specifically via the MT(1) receptor. Incubation experiments were carried out, and the insulin concentration in supernatants was measured using a radioimmunoassay. Furthermore, the intracellular cAMP was determined using an enzyme-linked immunosorbent assay. Real-time RT-PCR indicated that MT(1) knockdown resulted in a significant increase in the rIns1 mRNA and a significantly elevated basal insulin secretion of INS-1 cells. Incubation with melatonin decreased the amount of glucagon-like peptide 1 or inhibited the glucagon-stimulated insulin release of INS-1 cells, while, in MT(1) -knockdown cells, no melatonin-induced reduction in insulin secretion could be found. No decrease in 3-isobutyl-1-methylxanthine-stimulated intracellular cAMP in rMT(1) -knockdown cells was detectable after treatment with melatonin either, and immunocytochemistry proved that MT(1) knockdown abolished phosphorylation of cAMP-response-element-binding protein. In contrast to the INS-1 cells, preincubation with melatonin did not sensitize the insulin secretion of rMT(1) -knockdown cells. We also monitored insulin secretion from isolated islets of wild-type and melatonin-receptor knockout mice ex vivo. In islets of wild-type mice, melatonin treatment resulted in a decrease in insulin release, whereas melatonin treatment of islets from MT(1) knockout and MT(1/2) double-knockout mice did not show a significant effect. The data indicate that melatonin inhibits insulin secretion, primarily via the MT(1) receptor in rat INS-1 cells and isolated mouse islets.
Collapse
|