1
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
2
|
Amrollahi-Sharifabadi M, Oladejo TO, Ibrahim AS, Shakoor B, Mehrpour O, Sadeghi-Hashjin G, Gonçalves S. Melatonin's paradox: From therapeutic benefits to toxicity warnings. Chem Biol Interact 2025; 417:111556. [PMID: 40383469 DOI: 10.1016/j.cbi.2025.111556] [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: 03/28/2025] [Revised: 05/03/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Melatonin is an endogenous chemical predominantly synthesized in the pineal gland, widely recognized for its hormonal roles, such as regulating sleep and circadian rhythms. Through mechanisms such as anti-oxidative reduction, anti-inflammatory, and immunomodulation, it is suggested that melatonin exhibits biochemical properties in in vitro conditions. Beyond these functions, melatonin has garnered attention for its pharmacological benefits, particularly as a therapeutic agent that is exogenously administered as a supplement in various diseases ranging from insomnia to immunological and gastrointestinal disorders. However, emerging studies highlight potential toxicological concerns associated with exogenous melatonin use, especially in specific populations. This review provided a comprehensive exploration of melatonin's dual role as a therapeutic and potentially toxic agent. It summarized what is currently known about its pharmacological, toxicological, and biochemical characteristics as well as toxicity mechanisms, and safety concerns in susceptible groups. By highlighting new knowledge gaps about melatonin's clinical uses, the study opens the door for further studies to maximize its therapeutic benefits while maintaining its safety.
Collapse
Affiliation(s)
| | - Toheeb Olalekan Oladejo
- Department of Pharmacology and Toxicology, Nazarbayev School of Medicine, Astana, Kazakhstan
| | - Adedayo Sheu Ibrahim
- Department of Pharmacology and Toxicology, Nazarbayev School of Medicine, Astana, Kazakhstan
| | - Bushra Shakoor
- Synthetic and Natural Product Drug Discovery Laboratory, Department of Chemistry, Government College Women University Faisalabad, Faisalabad, 38000, Pakistan
| | - Omid Mehrpour
- Michigan Poison & Drug Information Center, School of Medicine, Wayne State University, Detroit, MI, United States of America
| | - Goudarz Sadeghi-Hashjin
- Department of Comparative Biosciences, College of Veterinary Medicine & Biomedical Science, University of Tehran, Tehran, Iran
| | - Sara Gonçalves
- Academic Clinical Center of Trás-os-Montes and Alto Douro (CACTMAD), University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; School of Health, University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal
| |
Collapse
|
3
|
Zheng TT, Liu JH, Huang WT, Hong B, Wang D, Liu CY, Zhang J, Li SS, Wu SW, Wang Q, Chen L, Jin L. Single-nucleotide polymorphisms in genes involved in folate metabolism or selected other metabolites and risk for gestational diabetes mellitus. World J Diabetes 2025; 16:103602. [DOI: 10.4239/wjd.v16.i5.103602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/09/2025] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND There are conflicting results on the potential correlation between folic acid and gestational diabetes mellitus (GDM), and the correlation between genetic factors related to folic acid metabolism pathways and GDM remains to be revealed.
AIM To examine the association between single-nucleotide polymorphisms (SNPs) of enzyme genes in the folate metabolite pathway as well as that between GDM-related genes and risk for GDM.
METHODS A nested case-control study was conducted with GDM cases (n = 412) and healthy controls (n = 412). DNA was extracted blood samples and SNPs were genotyped using Agena Bioscience’s MassARRAY gene mass spectrometry system. The associations between different SNPs of genes and the risk for GDM were estimated using logistic regression models. The generalized multi-factor dimensionality reduction (GMDR) method was used to analyze gene-gene and gene-environment interactions using the GMDR 0.9 software.
RESULTS The variation allele frequency of melatonin receptor 1B (MTNR1B) rs10830963 was higher in the GDM group than in controls (P < 0.05). MTNR1B rs10830963 mutant G was associated with risk for GDM [adjusted odds ratio (aOR): 1.43; 95% confidence interval (95%CI): 1.13-1.80] in the additive model. MTNR1B rs10830963 GG + GC was significantly associated with the risk for GDM (aOR: 1.65; 95%CI: 1.23-2.22) in the dominant model. The two-locus model of MTNR1B rs10830963 and CHEMERIN rs4721 was the best model (P < 0.05) for gene-gene interactions in the GMDR results. The high-risk rs10830963 × rs4721 type of interaction was a risk factor for GDM (aOR: 2.09; 95%CI: 1.49-2.93).
CONCLUSION This study does not find an association between SNPs of folate metabolic enzymes and risk for GDM. The G mutant allele of MTNR1B rs10830963 is identified as a risk factor for GDM in the additive model, and there may be gene-gene interactions between MTNR1B rs10830963 and CHEMERIN rs4721. It is conducive to studying the causes of GDM and provides a new perspective for the precise prevention of this disease.
Collapse
Affiliation(s)
- Ting-Ting Zheng
- Department of Obstetrics and Gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Department of Gynecology, Jinan Maternal and Child Care Hospital, Jinan 250000, Shandong Province, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing 100191, China
| | - Jia-He Liu
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Wan-Tong Huang
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Bo Hong
- Department of Obstetrics and Gynecology, Haidian Maternal and Child Health Care Hospital of Beijing, Beijing 100191, China
| | - Di Wang
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Chun-Yi Liu
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Jie Zhang
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Si-Si Li
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Shao-Wei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University Health Science Center, Xi'an 710000, Shaanxi Province, China
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Lei Chen
- Department of Obstetrics and Gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing 100191, China
| | - Lei Jin
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, China
- Key Laboratory of Reproductive Health, National Health Commission of the People’s Republic of China, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
4
|
Sørensen KV, Justesen JM, Ängquist L, Bork-Jensen J, Hartmann B, Jørgensen NR, Rungby J, Sørensen HT, Vaag A, Nielsen JS, Holst JJ, Pedersen O, Linneberg A, Hansen T, Grarup N. Rare MTNR1B variants causing diminished MT2 signalling associate with elevated HbA 1c levels but not with type 2 diabetes. Diabetologia 2025; 68:1016-1030. [PMID: 40064676 PMCID: PMC12021717 DOI: 10.1007/s00125-025-06381-y] [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: 06/28/2024] [Accepted: 12/10/2024] [Indexed: 04/25/2025]
Abstract
AIMS/HYPOTHESIS An intronic variant (rs10830963) in MTNR1B (encoding the melatonin receptor type 2 [MT2]) has been shown to strongly associate with impaired glucose regulation and elevated type 2 diabetes prevalence. However, MTNR1B missense variants have shown conflicting results on type 2 diabetes. Thus, we aimed to gain further insights into the impact of MTNR1B coding variants on type 2 diabetes prevalence and related phenotypes. METHODS We conducted a cross-sectional study, performing MTNR1B variant burden testing of glycaemic phenotypes (N=248,454, without diabetes), other cardiometabolic phenotypes (N=330,453) and type 2 diabetes prevalence (case-control study; N=263,739) in the UK Biobank. Similar burden testing with glycaemic phenotypes was performed in Danish Inter99 participants without diabetes (N=5711), and type 2 diabetes prevalence (DD2 cohort serving as cases [N=2930] and Inter99 serving as controls [N=4243]). Finally, we evaluated the effects of MTNR1B variants on the melatonin-induced glucose regulation response in a recall-by-genotype study of individuals without diabetes. RESULTS In the UK Biobank, MTNR1B variants were not associated with cardiometabolic phenotypes, including type 2 diabetes prevalence, except that carriers of missense MTNR1B variants causing impaired MT2 signalling exhibited higher HbA1c levels compared with non-carriers (effect size, β, 0.087 SD [95% CI 0.039, 0.135]). Similarly, no significant associations were observed with phenotypes associated with glycaemic phenotypes in the Inter99 population. However, carriers of variants impairing MT2 signalling demonstrated a nominally significant lower glucose-stimulated insulin response (β -0.47 SD [95% CI -0.82, -0.11]). A reduced insulin response was also observed in carriers of variants impairing MT2 signalling (β -476.0 [95% CI -928.6, -24.4]) or the rs10830963 variant (β -390.8 [95% CI -740.1, -41.6]) compared with non-carriers after melatonin treatment. CONCLUSIONS/INTERPRETATION The higher type 2 diabetes prevalence previously observed in carriers of missense MTNR1B variants causing impairment in MT2 signalling was not replicated in the UK Biobank, yet carriers had elevated HbA1c levels. DATA AVAILABILITY Data (Inter99 cohort and recall-by-genotype study) are available on reasonable request from the corresponding author. Requests for DD2 data are through the application form at https://dd2.dk/forskning/ansoeg-om-data . Access to UK Biobank data can be requested through the UK Biobank website ( https://www.ukbiobank.ac.uk/enable-your-research ).
Collapse
Affiliation(s)
- Kimmie V Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johanne M Justesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niklas R Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Translational Research Centre, Rigshospitalet, Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev Hospital, Herlev, Denmark
| | - Henrik T Sørensen
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
- Department of Epidemiology, Boston University, Boston, MA, USA
| | - Allan Vaag
- Steno Diabetes Center Copenhagen, Herlev Hospital, Herlev, Denmark
- Lund University Diabetes Care, Lund University, Malmö, Sweden
- Department of Endocrinology, Skåne University Hospital, Malmö, Sweden
| | - Jens S Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Allan Linneberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical 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.
| |
Collapse
|
5
|
Lima ACDS, Cruvinel NT, da Silva NR, Mendes MM, Duarte ACS, Coelho ASG, Vimaleswaran KS, Horst MA. Interaction Between Dietary Fiber Intake and MTNR1B rs10830963 Polymorphism on Glycemic Profiles in Young Brazilian Adults. Genes (Basel) 2025; 16:497. [PMID: 40428319 PMCID: PMC12110926 DOI: 10.3390/genes16050497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVE The single-nucleotide polymorphism (SNP) rs10830963 in the melatonin receptor 1B (MTNR1B) gene influences insulin secretion and glucose metabolism and has been associated with an increased risk of type-2 diabetes. This study aimed to explore the interaction between dietary intake and the MTNR1B rs10830963 polymorphism on glycemic profiles in young Brazilian adults. METHODS This cross-sectional study assessed 200 healthy young adults (19-24 years), evaluating the MTNR1B rs10830963 genotype, anthropometric parameters, glycemic markers (fasting insulin, glucose, HOMA-IR, and HOMA-β), and dietary intake via three 24 h dietary recalls. Genotype-diet interactions were tested using multivariate linear regression models adjusted for confounders. RESULTS The carriers of the G allele exhibited a positive association with fasting insulin levels (p = 0.003), insulin/glucose ratio (p = 0.004), HOMA-IR (p = 0.003), and HOMA-β (p = 0.018). Energy-adjusted fiber intake showed a significant genotype-specific interaction only in carriers of the G allele, where higher dietary fiber intake was significantly associated with lower fasting insulin (pinteraction = 0.034) and HOMA-IR (pinteraction = 0.028). CONCLUSION Our findings indicate that the MTNR1B rs10830963 polymorphism is associated with glycemic markers, and dietary fiber intake may attenuate the adverse effects of the MTNR1B rs10830963 G allele on glycemic profiles in young Brazilian adults. This highlights the potential role of fiber in improving health outcomes for individuals carrying this risk allele. To validate these results and assess the broader implications for the Brazilian population, further intervention studies and larger-scale research are essential.
Collapse
Affiliation(s)
- Ana Carolina da Silva Lima
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Nathália Teixeira Cruvinel
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Nara Rubia da Silva
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Marcela Moraes Mendes
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Amélia Cristina Stival Duarte
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
- Health Research Coordination, Organization: State Department of Health from Goiás (SES-GO), Goiânia 74853-070, GO, Brazil
| | | | - Karani S. Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK
- The Institute for Food, Nutrition, and Health, University of Reading, Reading RG6 6AH, UK
| | - Maria Aderuza Horst
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| |
Collapse
|
6
|
Škrlec I, Biloglav Z, Lešić D, Talapko J, Žabić I, Katalinić D. Association of MTNR1B Gene Polymorphisms with Body Mass Index in Hashimoto's Thyroiditis. Int J Mol Sci 2025; 26:3667. [PMID: 40332199 PMCID: PMC12027080 DOI: 10.3390/ijms26083667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Hashimoto's thyroiditis (HT) is an autoimmune disorder of the thyroid gland characterized by chronic inflammation, which in most cases results in hypothyroidism. The melatonin receptor MTNR1B is sporadically expressed in the thyroid gland. It modulates immune responses, and alterations in the melatonin-MTNR1B receptor signaling pathway may play a role in developing autoimmune diseases. Obesity worsens the severity and progression of some autoimmune diseases and reduces treatment efficacy. This study aimed to investigate the association of MTNR1B gene polymorphisms (rs10830963, rs1387153, and rs4753426) with HT with regards to the body mass index (BMI). Patients with HT were categorized into normal weight BMI ≤ 25 kg/m2 and overweight/obese BMI > 25 kg/m2 groups. This study included 115 patients with a clinical-, ultrasound-, and laboratory-confirmed diagnosis of HT (64 normal-weight patients and 51 overweight/obese patients) with a mean age of 43 ± 12 years. The results showed that specific MTNR1B polymorphisms are associated with obesity in HT patients. BMI was found to be associated with the rs10830963 polymorphism, and the G allele and GG genotype of the rs10830963 polymorphism were more common in overweight/obese HT patients. Furthermore, the results suggest that genetic factors associated with BMI play a role in developing HT and open new possibilities for personalized treatment approaches.
Collapse
Affiliation(s)
- Ivana Škrlec
- Faculty of Dental Medicine and Health, University J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Zrinka Biloglav
- Department of Medical Statistics, Epidemiology and Medical Informatics, School of Public Health Andrija Štampar, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Jasminka Talapko
- Faculty of Dental Medicine and Health, University J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Igor Žabić
- County Hospital Koprivnica, 48000 Koprivnica, Croatia
| | - Darko Katalinić
- Faculty of Dental Medicine and Health, University J. J. Strossmayer Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
7
|
Tripathy S, Bhattamisra SK. Cellular signalling of melatonin and its role in metabolic disorders. Mol Biol Rep 2025; 52:193. [PMID: 39903334 DOI: 10.1007/s11033-025-10306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Melatonin released from the pineal gland plays an important role in maintaining the light/dark cycle. Melatonin exerts its effects on various organs through receptor and nonreceptor pathways. Recently, the role of melatonin in various metabolic disorders has been investigated. This review focuses on the molecular pathways associated with melatonin and its role in metabolic disorders. In humans, melatonin acts through two G protein-coupled receptors (MT1 and MT2). Melatonin modulates insulin release, such as elevated insulin levels in the evening compared to morning hours, exerts cardioprotective effects through the cGMP pathway and nitric oxide production in endothelial cells, and controls oxidative stress and apoptosis in myocardial tissue. Melatonin through MT2 receptors increases lipolysis and thermogenesis, which have a positive effect on weight reduction in obese individuals. Currently, most drugs that target melatonin receptors are primarily used to treat neurological disorders. A detailed investigation to explore the role of melatonin and its signalling pathway in peripheral organs is essential to develop therapeutic molecules for managing metabolic disorders.
Collapse
Affiliation(s)
- Snehasis Tripathy
- IMT Pharmacy College, Sai Bihar, Gopalpur, Puri, Odisha, 752004, India
| | - Subrat Kumar Bhattamisra
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya, Bihar, 824236, India.
| |
Collapse
|
8
|
Lee J, Lee NK, Moon JH. Gestational Diabetes Mellitus: Mechanisms Underlying Maternal and Fetal Complications. Endocrinol Metab (Seoul) 2025; 40:10-25. [PMID: 39844628 PMCID: PMC11898322 DOI: 10.3803/enm.2024.2264] [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: 12/03/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Gestational diabetes mellitus (GDM) affects over 10% of all pregnancies, both in Korea and worldwide. GDM not only increases the risk of adverse pregnancy outcomes such as preeclampsia, preterm birth, macrosomia, neonatal hypoglycemia, and shoulder dystocia, but it also significantly increases the risk of developing postpartum type 2 diabetes mellitus and cardiovascular disease in the mother. Additionally, GDM is linked to a higher risk of childhood obesity and diabetes in offspring, as well as neurodevelopmental disorders, including autistic spectrum disorder. This review offers a comprehensive summary of clinical epidemiological studies concerning maternal and fetal complications and explores mechanistic investigations that reveal the underlying pathophysiology.
Collapse
Affiliation(s)
- Jooyeop Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Armed Forces Yangju Hospital, Yangju, Korea
| | - Na Keum Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Joon Ho Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
9
|
Pourbarkhordar V, Rahmani S, Roohbakhsh A, Hayes AW, Karimi G. Melatonin effect on breast and ovarian cancers by targeting the PI3K/Akt/mTOR pathway. IUBMB Life 2024; 76:1035-1049. [PMID: 39212097 DOI: 10.1002/iub.2900] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/29/2024] [Indexed: 09/04/2024]
Abstract
Melatonin, the hormone of the pineal gland, possesses a range of physiological functions, and recently, its anticancer effect has become more apparent. A more thorough understanding of molecular alterations in the components of several signaling pathways as new targets for cancer therapy is needed because of current innate restrictions such as drug toxicity, side effects, and acquired or de novo resistance. The PI3K/Akt/mTOR pathway is overactivated in many solid tumors, such as breast and ovarian cancers. This pathway in normal cells is essential for growth, proliferation, and survival. However, it is an undesirable characteristic in malignant cells. We have reviewed multiple studies about the effect of melatonin on breast and ovarian cancer, focusing on the PI3K/Akt/mTOR pathway. Melatonin exerts its inhibitory effects via several mechanisms. A: Downregulation of downstream or upstream components of the signaling pathway such as phosphatase and tensin homolog (PTEN), phosphatidylinositol (3,4,5)-trisphosphate kinase (PI3K), p-PI3K, Akt, p-Akt, mammalian target of rapamycin (mTOR), and mTOR complex1 (mTORC1). B: Apoptosis induction by decreasing MDM2 expression, a downstream target of Akt, and mTOR, which leads to Bad activation in addition to Bcl-XL and p53 inhibition. C: Induction of autophagy in cancer cells via activating ULK1 after mTOR inhibition, resulting in Beclin-1 phosphorylation. Beclin-1 with AMBRA1 and VPS34 promotes PI3K complex I activity and autophagy in cancer cells. The PI3K/Akt/mTOR pathway overlaps with other intracellular signaling pathways and components such as AMP-activated protein kinase (AMPK), Wnt/β-catenin, mitogen-activated protein kinase (MAPK), and other similar pathways. Cancer therapy can benefit from understanding how these pathways interact and how melatonin affects these pathways.
Collapse
Affiliation(s)
- Vahid Pourbarkhordar
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sohrab Rahmani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Goyal V, Tosini G. Disruption of Melatonin Signaling Leads to Lipids Accumulation in the Liver of Melatonin Proficient Mice. J Pineal Res 2024; 76:e70007. [PMID: 39539075 DOI: 10.1111/jpi.70007] [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: 03/05/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Melatonin signaling via melatonin receptor type 1 (MT1) and type 2 (MT2) plays an important role in the regulation of several physiological functions. Studies in rodents and humans have demonstrated that disruption of melatonin signaling may affect glucose metabolism, insulin sensitivity, and leptin levels. Accumulating experimental evidence also indicates that in rodents the administration of exogenous melatonin has a beneficial effect on the blood lipid levels. However, the molecular mechanism by which melatonin signaling may regulate lipids is still unclear. In addition, most of the studies with mice have been performed in melatonin-deficient mice by administering exogenous melatonin at supraphysiological doses. Hence the results of these studies may be greatly affected by these two factors. In this study, we report the effects of melatonin signaling removal on the liver biology and transcriptome using melatonin-proficient mice (C3H-f+/f+) in which MT1 or MT2 have been genetically ablated. Our data indicate that the absence of MT1 or MT2 signaling leads to disruption of the blood lipids profile and an increase in lipids deposition in the liver. These effects were more pronounced in the mice lacking MT1 than MT2. The gene expression profiles obtained with RNA-seq from the livers of the three genotypes revealed that removal of MT1 affected the transcription of 4255 genes (i.e., 40.6%). Conversely, the removal of MT2 affected the transcription of 1864 transcripts (i.e., 17.2%). Finally, we identified a group of 13 genes involved in lipids biology that may play a key role in the accumulation of lipids in the liver when melatonin signaling is disrupted. In conclusion, our study indicates that melatonin signaling is an important modulator of liver physiology and metabolism. Our study also indicated that the removal of MT1 signaling is more deleterious than MT2 removal.
Collapse
Affiliation(s)
- Varunika Goyal
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Neuroscience Institute, Atlanta, Georgia, USA
| | - Gianluca Tosini
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Neuroscience Institute, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Nath A, Ghosh S, Bandyopadhyay D. Role of melatonin in mitigation of insulin resistance and ensuing diabetic cardiomyopathy. Life Sci 2024; 355:122993. [PMID: 39154810 DOI: 10.1016/j.lfs.2024.122993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Addressing insulin resistance or hyperinsulinemia might offer a viable treatment approach to stop the onset of diabetic cardiomyopathy, as these conditions independently predispose to the development of the disease, which is initially characterized by diastolic abnormalities. The development of diabetic cardiomyopathy appears to be driven mainly by insulin resistance or impaired insulin signalling and/or hyperinsulinemia. Oxidative stress, hypertrophy, fibrosis, cardiac diastolic dysfunction, and, ultimately, systolic heart failure are the outcomes of these pathophysiological alterations. Melatonin is a ubiquitous indoleamine, a widely distributed compound secreted mainly by the pineal gland, and serves a variety of purposes in almost every living creature. Melatonin is found to play a leading role by improving myocardial cell metabolism, decreasing vascular endothelial cell death, reversing micro-circulation disorders, reducing myocardial fibrosis, decreasing oxidative and endoplasmic reticulum stress, regulating cell autophagy and apoptosis, and enhancing mitochondrial function. This review highlights a relationship between insulin resistance and associated cardiomyopathy. It explores the potential therapeutic strategies offered by the neurohormone melatonin, an important antioxidant that plays a leading role in maintaining glucose homeostasis by influencing the glucose transporters independently and through its receptors. The vast distribution of melatonin receptors in the body, including beta cells of pancreatic islets, asserts the role of this indole molecule in maintaining glucose homeostasis. Melatonin controls the production of GLUT4 and/or the phosphorylation process of the receptor for insulin and its intracellular substrates, activating the insulin-signalling pathway through its G-protein-coupled membrane receptors.
Collapse
Affiliation(s)
- Anupama Nath
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India
| | - Songita Ghosh
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India
| | - Debasish Bandyopadhyay
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India.
| |
Collapse
|
12
|
Li FXZ, Xu F, Li CC, Lei LM, Shan SK, Zheng MH, Lin X, Guo B, Tang KX, Duan JY, Wu YY, Cao YC, Liu JJ, Yuan LQ. Cold Exposure Alleviates T2DM Through Plasma-Derived Extracellular Vesicles. Int J Nanomedicine 2024; 19:10077-10095. [PMID: 39371478 PMCID: PMC11456273 DOI: 10.2147/ijn.s441847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/14/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Anecdotal reports have praised the benefits of cold exposure, exemplified by activities like winter swimming and cold water immersion. Cold exposure has garnered acclaim for its potential to confer benefits and potentially alleviate diabetes. We posited that systemic cold temperature (CT, 4-8°C) likely influences the organism's blood components through ambient temperature, prompting our investigation into the effects of chronic cold exposure on type 2 diabetic (T2DM) mice and our initial exploration of how cold exposure mitigates the incidence of T2DM. Methods The effects of CT (4-8°C) or room temperature (RT, 22-25°C) on T2DM mice were investigated. Mice blood and organ specimens were collected for fully automated biochemical testing, ELISA, HE staining, immunohistochemistry, and immunofluorescence. Glucose uptake was assessed using flow cytometry with 2-NBDG. Changes in potential signaling pathways such as protein kinase B (AKT), phosphorylated AKT (p-AKT), insulin receptor substrates 1 (IRS1), and phosphorylated IRS1 (p-IRS1) were evaluated by Western blot. Results CT or CT mice plasma-derived extracellular vesicles (CT-EVs) remarkably reduced blood glucose levels and improved insulin sensitivity in T2DM mice. This treatment enhanced glucose metabolism, systemic insulin sensitivity, and insulin secretion function while promoting glycogen accumulation in the liver and muscle. Additionally, CT-EVs treatment protected against the streptozocin (STZ)-induced destruction of islets in T2DM mice by inhibiting β-cell apoptosis. CT-EVs also shielded islets from destruction and increased the expression of p-IRS1 and p-AKT in adipocytes and hepatocytes. In vitro experiments further confirmed its pro-insulin sensitivity effect. Conclusion Our data indicate that cold exposure may have a potentially beneficial effect on the development of T2DM, mainly through the anti-diabetic effect of plasma-derived EVs released during cold stimulation. This phenomenon could significantly contribute to understanding the lower prevalence of diabetes in colder regions.
Collapse
Affiliation(s)
- Fu-Xing-Zi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Chang-Chun Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Ke-Xin Tang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Jia-Yue Duan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Yun-Yun Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Ye-Chi Cao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| | - Jun-Jie Liu
- Department of Periodontal Division, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People’s Republic of China
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Dalle S, Abderrahmani A. Receptors and Signaling Pathways Controlling Beta-Cell Function and Survival as Targets for Anti-Diabetic Therapeutic Strategies. Cells 2024; 13:1244. [PMID: 39120275 PMCID: PMC11311556 DOI: 10.3390/cells13151244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/10/2024] Open
Abstract
Preserving the function and survival of pancreatic beta-cells, in order to achieve long-term glycemic control and prevent complications, is an essential feature for an innovative drug to have clinical value in the treatment of diabetes. Innovative research is developing therapeutic strategies to prevent pathogenic mechanisms and protect beta-cells from the deleterious effects of inflammation and/or chronic hyperglycemia over time. A better understanding of receptors and signaling pathways, and of how they interact with each other in beta-cells, remains crucial and is a prerequisite for any strategy to develop therapeutic tools aimed at modulating beta-cell function and/or mass. Here, we present a comprehensive review of our knowledge on membrane and intracellular receptors and signaling pathways as targets of interest to protect beta-cells from dysfunction and apoptotic death, which opens or could open the way to the development of innovative therapies for diabetes.
Collapse
Affiliation(s)
- Stéphane Dalle
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 34094 Montpellier, France
| | - Amar Abderrahmani
- Université Lille, Centre National de la Recherche Scientifique (CNRS), Centrale Lille, Université Polytechnique Hauts-de-France, UMR 8520, IEMN, F59000 Lille, France
| |
Collapse
|
16
|
Sinnott-Armstrong N, Strausz S, Urpa L, Abner E, Valliere J, Palta P, Dashti HS, Daly M, Pritchard JK, Saxena R, Jones SE, Ollila HM. Genetic variants affect diurnal glucose levels throughout the day. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604631. [PMID: 39091879 PMCID: PMC11291026 DOI: 10.1101/2024.07.22.604631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Circadian rhythms not only coordinate the timing of wake and sleep but also regulate homeostasis within the body, including glucose metabolism. However, the genetic variants that contribute to temporal control of glucose levels have not been previously examined. Using data from 420,000 individuals from the UK Biobank and replicating our findings in 100,000 individuals from the Estonian Biobank, we show that diurnal serum glucose is under genetic control. We discover a robust temporal association of glucose levels at the Melatonin receptor 1B ( MTNR1B) (rs10830963, P = 1e-22) and a canonical circadian pacemaker gene Cryptochrome 2 ( CRY2) loci (rs12419690, P = 1e-16). Furthermore, we show that sleep modulates serum glucose levels and the genetic variants have a separate mechanism of diurnal control. Finally, we show that these variants independently modulate risk of type 2 diabetes. Our findings, together with earlier genetic and epidemiological evidence, show a clear connection between sleep and metabolism and highlight variation at MTNR1B and CRY2 as temporal regulators for glucose levels.
Collapse
|
17
|
Vella M, Mohan S, Christie H, Bailey KR, Cobelli C, Dalla Man C, Matveyenko A, Egan AM, Vella A. Diabetes-associated Genetic Variation in MTNR1B and Its Effect on Islet Function. J Endocr Soc 2024; 8:bvae130. [PMID: 39011323 PMCID: PMC11249077 DOI: 10.1210/jendso/bvae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Indexed: 07/17/2024] Open
Abstract
Context Multiple common genetic variants have been associated with type 2 diabetes, but the mechanism by which they predispose to diabetes is incompletely understood. One such example is variation in MTNR1B, which implicates melatonin and its receptor in the pathogenesis of type 2 diabetes. Objective To characterize the effect of diabetes-associated genetic variation at rs10830963 in the MTNR1B locus on islet function in people without type 2 diabetes. Design The association of genetic variation at rs10830963 with glucose, insulin, C-peptide, glucagon, and indices of insulin secretion and action were tested in a cohort of 294 individuals who had previously undergone an oral glucose tolerance test (OGTT). Insulin sensitivity, β-cell responsivity to glucose, and Disposition Indices were measured using the oral minimal model. Setting The Clinical Research and Translation Unit at Mayo Clinic, Rochester, MN. Participants Two cohorts were utilized for this analysis: 1 cohort was recruited on the basis of prior participation in a population-based study in Olmsted County. The other cohort was recruited on the basis of TCF7L2 genotype at rs7903146 from the Mayo Biobank. Intervention Two-hour, 7-sample OGTT. Main Outcome Measures Fasting, nadir, and integrated glucagon concentrations. Results One or 2 copies of the G-allele at rs10830963 were associated with increased postchallenge glucose and glucagon concentrations compared to subjects with the CC genotype. Conclusion The effects of rs10830963 on glucose homeostasis and predisposition to type 2 diabetes are likely to be partially mediated through changes in α-cell function.
Collapse
Affiliation(s)
- Max Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Sneha Mohan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Hannah Christie
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Claudio Cobelli
- Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, 35128 Padova, Italy
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Aoife M Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
18
|
Zhao H, Zhang T, Zhang H, Wang Y, Cheng L. Exercise-with-melatonin therapy improves sleep disorder and motor dysfunction in a rat model of ischemic stroke. Neural Regen Res 2024; 19:1336-1343. [PMID: 37905883 PMCID: PMC11467917 DOI: 10.4103/1673-5374.385844] [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: 02/19/2023] [Revised: 05/15/2023] [Accepted: 08/15/2023] [Indexed: 11/02/2023] Open
Abstract
Exercise-with-melatonin therapy has complementary and synergistic effects on spinal cord injury and Alzheimer’s disease, but its effect on stroke is still poorly understood. In this study, we established a rat model of ischemic stroke by occluding the middle cerebral artery for 60 minutes. We treated the rats with exercise and melatonin therapy for 7 consecutive days. Results showed that exercise-with-melatonin therapy significantly prolonged sleep duration in the model rats, increased delta power values, and regularized delta power rhythm. Additionally, exercise-with-melatonin therapy improved coordination, endurance, and grip strength, as well as learning and memory abilities. At the same time, it led to higher hippocampal CA1 neuron activity and postsynaptic density thickness and lower expression of glutamate receptor 2 than did exercise or melatonin therapy alone. These findings suggest that exercise-with-melatonin therapy can alleviate sleep disorder and motor dysfunction by increasing glutamate receptor 2 protein expression and regulating hippocampal CA1 synaptic plasticity.
Collapse
Affiliation(s)
- Haitao Zhao
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Tong Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Haojie Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Yunlei Wang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Lingna Cheng
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| |
Collapse
|
19
|
Wang B, Hou J, Mao Z, Chen C, Wang C, Yu S. Association between dinner-bedtime interval and type 2 diabetes mellitus: a large-scale cross-sectional study. J Diabetes Metab Disord 2024; 23:1039-1045. [PMID: 38932796 PMCID: PMC11196427 DOI: 10.1007/s40200-023-01382-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/21/2023] [Indexed: 06/28/2024]
Abstract
Objective Glucose metabolism is impacted by circadian disruption. Dinner-bedtime interval (DBI) was an accessible indicator to reflect the alignment between dinner time and circadian clock. We aimed to investigate the association of DBI with type 2 diabetes mellitus (T2DM). Methods 7676 adult subjects from the Henan Rural Cohort were included. Their demographic information including dinner time and bedtime was collected. Fasting venous blood samples were collected for biochemical determinations. Generalized linear regression model was used to analyze the factors influencing DBI. Furthermore, logistic regression incorporated with restricted cubic spline model was applied to evaluate the association between DBI and T2DM. Results The results of multiple linear regression model showed that age (β: -0.018, 95% CI: -0.021, -0.015) was negatively correlated with DBI. Female (β: 0.311, 95% CI: 0.229, 0.393), junior high school education (β: 0.246, 95% CI: 0.187, 0.306), high school education or above (β: 0.346, 95% CI: 0.259, 0.433), average monthly income with 1000-1999 CNY(0.102, 95% CI: 0.032, 0.171), average monthly income ≥ 2000 CNY (β: 0.164, 95% CI: 0.076, 0.251), moderate physical activity (β: 0.134, 95% CI: 0.071, 0.197), current smokers (β: 0.214, 95% CI: 0.118, 0.309), current drinkers (β: 0.099, 95% CI: 0.008, 0.190) were positively correlated with DBI. Furthermore, DBI was significantly associated with T2DM (adjusted OR: 0.910, 95%CI: 0.845-0.979, P = 0.012). DBI longer than 3 h was associated with decreased risk of T2DM (adjusted OR: 0.773, 95%CI: 0.648-0.921, P = 0.004). Conclusions DBI larger than 3 h is beneficial to T2DM prevention. Further investigation is required to verify the association.
Collapse
Affiliation(s)
- Bingqian Wang
- College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou City, 450001 People’s Republic of China
| | - Jian Hou
- College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou City, 450001 People’s Republic of China
| | - Zhenxing Mao
- College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou City, 450001 People’s Republic of China
| | - Changying Chen
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052 China
- Institute for Hospital Management of Henan Province, Zhengzhou, 450052 China
| | - Chongjian Wang
- College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou City, 450001 People’s Republic of China
| | - Songcheng Yu
- College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou City, 450001 People’s Republic of China
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052 China
- Institute for Hospital Management of Henan Province, Zhengzhou, 450052 China
| |
Collapse
|
20
|
Kurgan N, Kjærgaard Larsen J, Deshmukh AS. Harnessing the power of proteomics in precision diabetes medicine. Diabetologia 2024; 67:783-797. [PMID: 38345659 DOI: 10.1007/s00125-024-06097-5] [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: 11/14/2023] [Accepted: 12/20/2023] [Indexed: 03/21/2024]
Abstract
Precision diabetes medicine (PDM) aims to reduce errors in prevention programmes, diagnosis thresholds, prognosis prediction and treatment strategies. However, its advancement and implementation are difficult due to the heterogeneity of complex molecular processes and environmental exposures that influence an individual's disease trajectory. To address this challenge, it is imperative to develop robust screening methods for all areas of PDM. Innovative proteomic technologies, alongside genomics, have proven effective in precision cancer medicine and are showing promise in diabetes research for potential translation. This narrative review highlights how proteomics is well-positioned to help improve PDM. Specifically, a critical assessment of widely adopted affinity-based proteomic technologies in large-scale clinical studies and evidence of the benefits and feasibility of using MS-based plasma proteomics is presented. We also present a case for the use of proteomics to identify predictive protein panels for type 2 diabetes subtyping and the development of clinical prediction models for prevention, diagnosis, prognosis and treatment strategies. Lastly, we discuss the importance of plasma and tissue proteomics and its integration with genomics (proteogenomics) for identifying unique type 2 diabetes intra- and inter-subtype aetiology. We conclude with a call for action formed on advancing proteomics technologies, benchmarking their performance and standardisation across sites, with an emphasis on data sharing and the inclusion of diverse ancestries in large cohort studies. These efforts should foster collaboration with key stakeholders and align with ongoing academic programmes such as the Precision Medicine in Diabetes Initiative consortium.
Collapse
Affiliation(s)
- Nigel Kurgan
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe Kjærgaard Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Speksnijder EM, Bisschop PH, Siegelaar SE, Stenvers DJ, Kalsbeek A. Circadian desynchrony and glucose metabolism. J Pineal Res 2024; 76:e12956. [PMID: 38695262 DOI: 10.1111/jpi.12956] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Esther M Speksnijder
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Challet E, Pévet P. Melatonin in energy control: Circadian time-giver and homeostatic monitor. J Pineal Res 2024; 76:e12961. [PMID: 38751172 DOI: 10.1111/jpi.12961] [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: 12/05/2023] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Melatonin is a neurohormone synthesized from dietary tryptophan in various organs, including the pineal gland and the retina. In the pineal gland, melatonin is produced at night under the control of the master clock located in the suprachiasmatic nuclei of the hypothalamus. Under physiological conditions, the pineal gland seems to constitute the unique source of circulating melatonin. Melatonin is involved in cellular metabolism in different ways. First, the circadian rhythm of melatonin helps the maintenance of proper internal timing, the disruption of which has deleterious effects on metabolic health. Second, melatonin modulates lipid metabolism, notably through diminished lipogenesis, and it has an antidiabetic effect, at least in several animal models. Third, pharmacological doses of melatonin have antioxidative, free radical-scavenging, and anti-inflammatory properties in various in vitro cellular models. As a result, melatonin can be considered both a circadian time-giver and a homeostatic monitor of cellular metabolism, via multiple mechanisms of action that are not all fully characterized. Aging, circadian disruption, and artificial light at night are conditions combining increased metabolic risks with diminished circulating levels of melatonin. Accordingly, melatonin supplementation could be of potential therapeutic value in the treatment or prevention of metabolic disorders. More clinical trials in controlled conditions are needed, notably taking greater account of circadian rhythmicity.
Collapse
Affiliation(s)
- Etienne Challet
- Centre National de la Recherche Scientifique (CNRS), Institute of Cellular and Integrative Neurosciences, University of Strasbourg, Strasbourg, France
| | - Paul Pévet
- Centre National de la Recherche Scientifique (CNRS), Institute of Cellular and Integrative Neurosciences, University of Strasbourg, Strasbourg, France
| |
Collapse
|
23
|
Odriozola A, González A, Álvarez-Herms J, Corbi F. Host genetics and nutrition. ADVANCES IN GENETICS 2024; 111:199-235. [PMID: 38908900 DOI: 10.1016/bs.adgen.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Optimal nutrition is essential for health and physiological performance. Nutrition-related diseases such as obesity and diabetes are major causes of death and reduced quality of life in modern Western societies. Thanks to combining nutrigenetics and nutrigenomics, genomic nutrition allows the study of the interaction between nutrition, genetics and physiology. Currently, interrelated multi-genetic and multifactorial phenotypes are studied from a multiethnic and multi-omics approach, step by step identifying the important role of pathways, in addition to those directly related to metabolism. It allows the progressive identification of genetic profiles associated with specific susceptibilities to diet-related phenotypes, which may facilitate individualised dietary recommendations to improve health and quality of life.
Collapse
Affiliation(s)
- Adrián Odriozola
- Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Adriana González
- Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jesús Álvarez-Herms
- Phymo® Lab, Physiology, and Molecular Laboratory, Collado Hermoso, Segovia, Spain
| | - Francesc Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), Centre de Lleida, Universitat de Lleida (UdL), Lleida, Spain
| |
Collapse
|
24
|
Heianza Y, Zhou T, Wang X, Furtado JD, Appel LJ, Sacks FM, Qi L. MTNR1B genotype and effects of carbohydrate quantity and dietary glycaemic index on glycaemic response to an oral glucose load: the OmniCarb trial. Diabetologia 2024; 67:506-515. [PMID: 38052941 DOI: 10.1007/s00125-023-06056-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023]
Abstract
AIMS/HYPOTHESIS A type 2 diabetes-risk-increasing variant, MTNR1B (melatonin receptor 1B) rs10830963, regulates the circadian function and may influence the variability in metabolic responses to dietary carbohydrates. We investigated whether the effects of carbohydrate quantity and dietary glycaemic index (GI) on glycaemic response during OGTTs varied by the risk G allele of MTNR1B-rs10830963. METHODS This study included participants (n=150) of a randomised crossover-controlled feeding trial of four diets with high/low GI levels and high/low carbohydrate content for 5 weeks. The MTNR1B-rs10830963 (C/G) variant was genotyped. Glucose response during 2 h OGTT was measured at baseline and the end of each diet intervention. RESULTS Among the four study diets, carrying the risk G allele (CG/GG vs CC genotype) of MTNR1B-rs10830963 was associated with the largest AUC of glucose during 2 h OGTT after consuming a high-carbohydrate/high-GI diet (β 134.32 [SE 45.69] mmol/l × min; p=0.004). The risk G-allele carriers showed greater increment of glucose during 0-60 min (β 1.26 [0.47] mmol/l; p=0.008) or 0-90 min (β 1.10 [0.50] mmol/l; p=0.028) after the high-carbohydrate/high-GI diet intervention, but not after consuming the other three diets. At high carbohydrate content, reducing GI levels decreased 60 min post-OGTT glucose (mean -0.67 [95% CI: -1.18, -0.17] mmol/l) and the increment of glucose during 0-60 min (mean -1.00 [95% CI: -1.67, -0.33] mmol/l) and 0-90 min, particularly in the risk G-allele carriers (pinteraction <0.05 for all). CONCLUSIONS/INTERPRETATION Our study shows that carrying the risk G allele of MTNR1B-rs10830963 is associated with greater glycaemic responses after consuming a diet with high carbohydrates and high GI levels. Reducing GI in a high-carbohydrate diet may decrease post-OGTT glucose concentrations among the risk G-allele carriers.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
| | - Tao Zhou
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Epidemiology, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Xuan Wang
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Jeremy D Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Biogen Epidemiology, Cambridge, MA, USA
| | - Lawrence J Appel
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
25
|
Zhang Y, Zhao K, Jin L, Zhou Y, Shang X, Wang X, Yu H. MTNR1B gene variations and high pre-pregnancy BMI increase gestational diabetes mellitus risk in Chinese women. Gene 2024; 894:148023. [PMID: 38007162 DOI: 10.1016/j.gene.2023.148023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 11/27/2023]
Abstract
AIM To investigate the association of melatonin receptor 1B (MTNR1B) gene variations and pre-pregnancy body mass index (BMI) with gestational diabetes mellitus (GDM). MATERIALS AND METHOD In this study, 1566 Chinese Han pregnant women were enrolled and multiple genetic models were used to evaluate the association between MTNR1B gene polymorphisms and the risk of GDM. The clinical value of pre-pregnancy BMI in predicting GDM was analyzed and evaluated using receiver operating characteristic (ROC) curves. Several methods of analysis were used to examine the impact of gene-gene and gene-BMI interactions on the incidence of GDM influence. RESULTS For the MTNR1B gene, rs1387153 (C > T), rs10830962 (C > G), rs4753426 (T > C), and rs10830963 (C > G) are all risk mutations associated with the susceptibility of GDM. The ROC curve analysis indicated that the BMI demonstrated an area under the curve (AUC) of 0.595. Alongside, the sensitivity and specificity stood at 0.676 and 0.474 respectively. The maximum Joden index was found to be 0.150, with a corresponding critical BMI value of 20.5691 kg/m2. Interaction analysis revealed that gene-gene and gene-BMI interactions had no significant effect on GDM occurrence. CONCLUSION MTNR1B genetic variations confers the risk to GDM in Chinese women. Furthermore, the high pre-pregnancy BMI (≥20.5691 kg/m2) significantly increases the risk of GDM in Chinese women.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Jin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
| | - Yuanzhong Zhou
- School of Public health, Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Guizhou, China
| | - Xuejun Shang
- Department of Urology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Xin Wang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China.
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China.
| |
Collapse
|
26
|
Roberts FL, Cataldo LR, Fex M. Monoamines' role in islet cell function and type 2 diabetes risk. Trends Mol Med 2023; 29:1045-1058. [PMID: 37722934 DOI: 10.1016/j.molmed.2023.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/20/2023]
Abstract
The two monoamines serotonin and melatonin have recently been highlighted as potent regulators of islet hormone secretion and overall glucose homeostasis in the body. In fact, dysregulated signaling of both amines are implicated in β-cell dysfunction and development of type 2 diabetes mellitus (T2DM). Serotonin is a key player in β-cell physiology and plays a role in expansion of β-cell mass. Melatonin regulates circadian rhythm and nutrient metabolism and reduces insulin release in human and rodent islets in vitro. Herein, we focus on the role of serotonin and melatonin in islet physiology and the pathophysiology of T2DM. This includes effects on hormone secretion, receptor expression, genetic variants influencing β-cell function, melatonin treatment, and compounds that alter serotonin availability and signaling.
Collapse
Affiliation(s)
- Fiona Louise Roberts
- Lund University Diabetes Centre, Department of Clinical Sciences, Unit for Molecular Metabolism, SE-21428 Malmö, Sweden
| | - Luis Rodrigo Cataldo
- Lund University Diabetes Centre, Department of Clinical Sciences, Unit for Molecular Metabolism, SE-21428 Malmö, Sweden; The Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Malin Fex
- Lund University Diabetes Centre, Department of Clinical Sciences, Unit for Molecular Metabolism, SE-21428 Malmö, Sweden.
| |
Collapse
|
27
|
Ruiz-Pablos M, Paiva B, Zabaleta A. Epstein-Barr virus-acquired immunodeficiency in myalgic encephalomyelitis-Is it present in long COVID? J Transl Med 2023; 21:633. [PMID: 37718435 PMCID: PMC10506247 DOI: 10.1186/s12967-023-04515-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023] Open
Abstract
Both myalgic encephalomyelitis or chronic fatigue syndrome (ME/CFS) and long COVID (LC) are characterized by similar immunological alterations, persistence of chronic viral infection, autoimmunity, chronic inflammatory state, viral reactivation, hypocortisolism, and microclot formation. They also present with similar symptoms such as asthenia, exercise intolerance, sleep disorders, cognitive dysfunction, and neurological and gastrointestinal complaints. In addition, both pathologies present Epstein-Barr virus (EBV) reactivation, indicating the possibility of this virus being the link between both pathologies. Therefore, we propose that latency and recurrent EBV reactivation could generate an acquired immunodeficiency syndrome in three steps: first, an acquired EBV immunodeficiency develops in individuals with "weak" EBV HLA-II haplotypes, which prevents the control of latency I cells. Second, ectopic lymphoid structures with EBV latency form in different tissues (including the CNS), promoting inflammatory responses and further impairment of cell-mediated immunity. Finally, immune exhaustion occurs due to chronic exposure to viral antigens, with consolidation of the disease. In the case of LC, prior to the first step, there is the possibility of previous SARS-CoV-2 infection in individuals with "weak" HLA-II haplotypes against this virus and/or EBV.
Collapse
Affiliation(s)
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IdiSNA, Instituto de Investigación Sanitaria de Navarra, Av. Pío XII 55, 31008, Pamplona, Spain
| | - Aintzane Zabaleta
- Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IdiSNA, Instituto de Investigación Sanitaria de Navarra, Av. Pío XII 55, 31008, Pamplona, Spain.
| |
Collapse
|
28
|
Ouyang S, Su Y, Ding N, Su Y, He L. The association between bedtime at night and diabetes in US adults: Data from National Health and Nutrition Examination Survey (NHANES) 2015-March -2020 pre-pandemic. PLoS One 2023; 18:e0287090. [PMID: 37310940 DOI: 10.1371/journal.pone.0287090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 05/30/2023] [Indexed: 06/15/2023] Open
Abstract
OBJECTIVE The purpose of this study was to investigate the relationship between bedtime at night and the risk of diabetes in adults. METHODS We extracted data from 14,821 target subjects from the NHANES database for a cross-sectional study. The data on bedtime came from the question in the sleep questionnaire: "What time do you usually fall asleep on weekdays or workdays?". Diabetes was defined as fasting blood sugar ≥ 126mg/dL, or glycohemoglobin ≥ 6.5%, or 2-hour Oral Glucose Tolerance Test blood sugar ≥ 200mg/dL, or taking hypoglycemic agent and insulin, or self-reported diabetes mellitus. A weighted multivariate logistic regression analysis was conducted to explore the relationship between bedtime at night and diabetes in adults. RESULTS From 19:00 to 23:00, a significantly negative association can be found between bedtime and diabetes(OR, 0.91 [95%CI, 0.83, 0.99]). From 23:00 to 02:00, The relationship between the two was positive(OR, 1.07 [95%CI, 0.94, 1.22]), nevertheless, the P values was not statistically significant(p = 0.3524). In subgroup analysis, from 19:00-23:00, the relationship was negative across genders, and in males, the P-values were still statistically significant(p = 0.0414). From 23:00-02:00, the relationship was positive across genders. CONCLUSION Earlier bedtime (before 23:00) increased the risk of developing diabetes. And this effect was not significantly different between male and female. For bedtime between 23:00-2:00, there was a trend of increasing the risk of diabetes as the bedtime was delayed.
Collapse
Affiliation(s)
- Shayuan Ouyang
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital Central South University, Changsha, Hunan, China
- Department of Cardiovascular Surgery, The Second Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yinghua Su
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital Central South University, Changsha, Hunan, China
- Department of Cardiovascular Surgery, The Second Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Ning Ding
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Yingjie Su
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Liudang He
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
29
|
Bijnens S, Depoortere I. Controlled light exposure and intermittent fasting as treatment strategies for metabolic syndrome and gut microbiome dysregulation in night shift workers. Physiol Behav 2023; 263:114103. [PMID: 36731762 DOI: 10.1016/j.physbeh.2023.114103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/09/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023]
Abstract
The mammalian circadian clocks are entrained by environmental time cues, such as the light-dark cycle and the feeding-fasting cycle. In modern society, circadian misalignment is increasingly more common under the guise of shift work. Shift workers, accounting for roughly 20% of the workforce population, are more susceptible to metabolic disease. Exposure to artificial light at night and eating at inappropriate times of the day uncouples the central and peripheral circadian clocks. This internal circadian desynchrony is believed to be one of the culprits leading to metabolic disease. In this review, we discuss how alterations in the rhythm of gut microbiota and their metabolites during chronodisruption send conflicting signals to the host, which may ultimately contribute to disturbed metabolic processes. We propose two behavioral interventions to improve health in shift workers. Firstly, by carefully timing the moments of exposure to blue light, and hence shifting the melatonin peak, to improve sleep quality of daytime sleeping episodes. Secondly, by timing the daily time window of caloric intake to the biological morning, to properly align the feeding-fasting cycle with the light-dark cycle and to reduce the risk of metabolic disease. These interventions can be a first step in reducing the worldwide burden of health problems associated with shift work.
Collapse
Affiliation(s)
- Sofie Bijnens
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium.
| |
Collapse
|
30
|
Qin X, Fu Y, Fan J, Liu B, Liu P, Zhang Y, Jiang T, Zheng Q. Melatonin increases susceptibility to atrial fibrillation in obesity via Akt signaling impairment in response to lipid overload. J Pineal Res 2023; 74:e12851. [PMID: 36639364 DOI: 10.1111/jpi.12851] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Melatonin has been proven to have antiarrhythmic potential; however, several studies have recently challenged this view. Herein, using a mouse model of obesity-induced atrial fibrillation (AF), we tentatively explored whether exogenous melatonin supplementation could increase AF susceptibility in the context of obesity. We observed that an 8-week drinking administration of melatonin (60 µg/ml in water) induced a greater susceptibility to AF in obese mice, although obesity-induced structural remodeling was alleviated. An investigation of systemic insulin sensitivity showed that melatonin treatment improved insulin sensitivity in obese mice, whereas it inhibited glucose-stimulated insulin secretion. Notably, melatonin treatment inhibited protein kinase B (Akt) signaling in the atria of obese mice and palmitate-treated neonatal rat cardiomyocytes, thereby providing an AF substrate. Melatonin increased lipid stress in obesity, as evidenced by elevated lipid accumulation and lipolysis-related gene expression, thus contributing to the impairment in atrial Akt signaling. Taken together, our results demonstrated that melatonin could increase AF susceptibility in obesity, probably due to increased lipid stress and resultant impairment of atrial Akt signaling. Our findings suggest that special precautions should be taken when administering melatonin to obese subjects.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Beilin District, Xi'an, Shaanxi, China
| | - Yuping Fu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xincheng District, Xi'an, Shaanxi, China
| | - Jiali Fan
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xincheng District, Xi'an, Shaanxi, China
| | - Binghua Liu
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Beilin District, Xi'an, Shaanxi, China
| | - Peng Liu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xincheng District, Xi'an, Shaanxi, China
| | - Yudi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xincheng District, Xi'an, Shaanxi, China
| | - Tiannan Jiang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xincheng District, Xi'an, Shaanxi, China
| |
Collapse
|
31
|
Chen F, Fei X, Li M, Zhang Z, Zhu W, Zhang M, Chen X, Xu J, Zhang M, Shen Y, Du J. Associations of the MTNR1B rs10830963 and PPARG rs1801282 variants with gestational diabetes mellitus: A meta-analysis. Int J Diabetes Dev Ctries 2023. [DOI: 10.1007/s13410-023-01188-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
|
32
|
Lodato M, Plaisance V, Pawlowski V, Kwapich M, Barras A, Buissart E, Dalle S, Szunerits S, Vicogne J, Boukherroub R, Abderrahmani A. Venom Peptides, Polyphenols and Alkaloids: Are They the Next Antidiabetics That Will Preserve β-Cell Mass and Function in Type 2 Diabetes? Cells 2023; 12:cells12060940. [PMID: 36980281 PMCID: PMC10047094 DOI: 10.3390/cells12060940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Improvement of insulin secretion by pancreatic β-cells and preservation of their mass are the current challenges that future antidiabetic drugs should meet for achieving efficient and long-term glycemic control in patients with type 2 diabetes (T2D). The successful development of glucagon-like peptide 1 (GLP-1) analogues, derived from the saliva of a lizard from the Helodermatidae family, has provided the proof of concept that antidiabetic drugs directly targeting pancreatic β-cells can emerge from venomous animals. The literature reporting on the antidiabetic effects of medicinal plants suggests that they contain some promising active substances such as polyphenols and alkaloids, which could be active as insulin secretagogues and β-cell protectors. In this review, we discuss the potential of several polyphenols, alkaloids and venom peptides from snake, frogs, scorpions and cone snails. These molecules could contribute to the development of new efficient antidiabetic medicines targeting β-cells, which would tackle the progression of the disease.
Collapse
Affiliation(s)
- Michele Lodato
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valérie Plaisance
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valérie Pawlowski
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Maxime Kwapich
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
- Service de Diabétologie et d’Endocrinologie, CH Dunkerque, 59385 Dunkirk, France
| | - Alexandre Barras
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Emeline Buissart
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Stéphane Dalle
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Sabine Szunerits
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Jérôme Vicogne
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Rabah Boukherroub
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Amar Abderrahmani
- University Lille, CNRS, Centrale Lille, University Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
- Correspondence: ; Tel.: +33-362531704
| |
Collapse
|
33
|
Xia AY, Zhu H, Zhao ZJ, Liu HY, Wang PH, Ji LD, Xu J. Molecular Mechanisms of the Melatonin Receptor Pathway Linking Circadian Rhythm to Type 2 Diabetes Mellitus. Nutrients 2023; 15:nu15061406. [PMID: 36986139 PMCID: PMC10052080 DOI: 10.3390/nu15061406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/04/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Night-shift work and sleep disorders are associated with type 2 diabetes (T2DM), and circadian rhythm disruption is intrinsically involved. Studies have identified several signaling pathways that separately link two melatonin receptors (MT1 and MT2) to insulin secretion and T2DM occurrence, but a comprehensive explanation of the molecular mechanism to elucidate the association between these receptors to T2DM, reasonably and precisely, has been lacking. This review thoroughly explicates the signaling system, which consists of four important pathways, linking melatonin receptors MT1 or MT2 to insulin secretion. Then, the association of the circadian rhythm with MTNR1B transcription is extensively expounded. Finally, a concrete molecular and evolutionary mechanism underlying the macroscopic association between the circadian rhythm and T2DM is established. This review provides new insights into the pathology, treatment, and prevention of T2DM.
Collapse
Affiliation(s)
- An-Yu Xia
- Department of Clinical Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hui Zhu
- Department of Internal Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Zhi-Jia Zhao
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Hong-Yi Liu
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Peng-Hao Wang
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Lin-Dan Ji
- Department of Biochemistry, School of Medicine, Ningbo University, Ningbo 315211, China
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
- Correspondence: (L.-D.J.); (J.X.)
| | - Jin Xu
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, China
- Correspondence: (L.-D.J.); (J.X.)
| |
Collapse
|
34
|
Xu W, Cui Y, Guo D, Wang W, Xu H, Qiao S, Yu H, Ji E, Liu Y, Li Q. UPLC-MS/MS simultaneous quantification of urinary circadian rhythm hormones and related metabolites: Application to air traffic controllers. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1222:123664. [PMID: 37040674 DOI: 10.1016/j.jchromb.2023.123664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/31/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Civil aviation flight crew and civil aviation air traffic controllers are prone to circadian rhythm abnormalities, which can lead to a slew of other maladies. It could endanger people's health and provide a serious threat to the safety of civil aviation flights if it is not appropriately evaluated and addressed. Early detection of rhythm irregularities and prompt treatment for particular populations that are vulnerable to rhythm disorders are crucial for enhancing civil aviation safety. In general, monitoring of the classical circadian rhythm biomarkers (melatonin or cortisol) in plasma or saliva is an effective way to evaluate the rhythm status. Due to the challenging sample procedure and the trauma of plasma, urine sample testing has received an increasing amount of attention. While, urine circadian rhythm biomarkers have seldom been examined, and the relationship between urinary steroid hormones and melatonin is still poorly understood. In most cases, hormones are determined by immunoassays respectively, mainly enzyme-linked immunosorbent assay (ELISA) or radioimmunoassay (RIA). There are also reports describing the liquid chromatography with tandem mass spectrometry (LC-MS/MS) technique as a method of melatonin or few steroid hormones quantification, however, the simultaneous detection of multiple rhythmic hormones in human urine is rarely reported. For the quantification of the rhythmic hormones in human urine, an accurate approach using ultra performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was devised in this work. Nine endogenous hormones (melatonin, 6-hydroxymelatonin, 6-sulfatoxymelatonin, cortisol, corticosterone, cortisone, testosterone, epitestosterone and androsterone), in human overnight urine, were quantified after solid phase extraction (SPE). A reverse phase HSS C18 column was used for chromatographic separation with a 9-minute gradient elution and deuterated analogues of each analyte were applied as internal standards. This method was successfully applied to the analysis of 596 overnight urine samples (23:00-9:00) collected from 84 air traffic controllers in the Beijing area during shift work. This study's findings showed a clear correlation not only between melatonin and its metabolites; cortisol-related metabolites, but also between melatonin metabolites and endogenous metabolites upstream and downstream of cortisol, implying that these two categories of hormones can be used as potential biological rhythm indicators to provide circadian rhythm data support for future studies on circadian rhythm disorders.
Collapse
Affiliation(s)
- Weizhe Xu
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Yujing Cui
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Danming Guo
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Wei Wang
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Haishan Xu
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Shi Qiao
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Hongyan Yu
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Enhui Ji
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Yongsuo Liu
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| | - Qingyan Li
- Civil Aviation Medical Center, Civil Aviation Administration of China, Beijing 100123, China
| |
Collapse
|
35
|
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
|
36
|
Duan D, Kim LJ, Jun JC, Polotsky VY. Connecting insufficient sleep and insomnia with metabolic dysfunction. Ann N Y Acad Sci 2023; 1519:94-117. [PMID: 36373239 PMCID: PMC9839511 DOI: 10.1111/nyas.14926] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The global epidemic of obesity and type 2 diabetes parallels the rampant state of sleep deprivation in our society. Epidemiological studies consistently show an association between insufficient sleep and metabolic dysfunction. Mechanistically, sleep and circadian rhythm exert considerable influences on hormones involved in appetite regulation and energy metabolism. As such, data from experimental sleep deprivation in humans demonstrate that insufficient sleep induces a positive energy balance with resultant weight gain, due to increased energy intake that far exceeds the additional energy expenditure of nocturnal wakefulness, and adversely impacts glucose metabolism. Conversely, animal models have found that sleep loss-induced energy expenditure exceeds caloric intake resulting in net weight loss. However, animal models have significant limitations, which may diminish the clinical relevance of their metabolic findings. Clinically, insomnia disorder and insomnia symptoms are associated with adverse glucose outcomes, though it remains challenging to isolate the effects of insomnia on metabolic outcomes independent of comorbidities and insufficient sleep durations. Furthermore, both pharmacological and behavioral interventions for insomnia may have direct metabolic effects. The goal of this review is to establish an updated framework for the causal links between insufficient sleep and insomnia and risks for type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Daisy Duan
- Division of Endocrinology, Diabetes, and Metabolism; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lenise J. Kim
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan C. Jun
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Lane JM, Qian J, Mignot E, Redline S, Scheer FAJL, Saxena R. Genetics of circadian rhythms and sleep in human health and disease. Nat Rev Genet 2023; 24:4-20. [PMID: 36028773 PMCID: PMC10947799 DOI: 10.1038/s41576-022-00519-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 12/13/2022]
Abstract
Circadian rhythms and sleep are fundamental biological processes integral to human health. Their disruption is associated with detrimental physiological consequences, including cognitive, metabolic, cardiovascular and immunological dysfunctions. Yet many of the molecular underpinnings of sleep regulation in health and disease have remained elusive. Given the moderate heritability of circadian and sleep traits, genetics offers an opportunity that complements insights from model organism studies to advance our fundamental molecular understanding of human circadian and sleep physiology and linked chronic disease biology. Here, we review recent discoveries of the genetics of circadian and sleep physiology and disorders with a focus on those that reveal causal contributions to complex diseases.
Collapse
Affiliation(s)
- Jacqueline M Lane
- Center for Genomic Medicine and Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jingyi Qian
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Emmanuel Mignot
- Center for Narcolepsy, Stanford University, Palo Alto, California, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
38
|
Ocak Ö, Silan F, Şahin EM. Melatonin receptor gene polymorphisms as a risk factor in patients with diabetic peripheral neuropathy. Diabetes Metab Res Rev 2022; 38:e3573. [PMID: 36018079 DOI: 10.1002/dmrr.3573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/24/2022] [Indexed: 11/08/2022]
Abstract
AIMS Oxidative stress plays an important role in the pathogenesis of diabetic peripheral neuropathy (DPN). Melatonin is one of the most powerful endogenous antioxidants and has anti-inflammatory properties. We investigated how the gene polymorphism of melatonin differs in patients with DPN compared to an healthy control group. MATERIALS AND METHODS A total of 54 diabetic peripheral neuropathy patients who applied to the Neurology outpatient clinic between 2020 and 2021, and 53 healthy controls comparable with the patient group in terms of age and gender were included in the study. Electromyography was performed and the melatonin gene polymorphism was analysed using the pyrosequencing method. RESULTS Melatonin gene variants rs2119882, rs13140012, and rs10830963 were analysed in patients and controls. The rs2119882 (G allele) has a protective role, and rs13140012 polymorphism has a related 5-fold higher risk of DPN in the recessive model. CONCLUSIONS Melatonin gene polymorphisms have been shown to be associated with DPN. This is the first and only study investigating the relationship between melatonin gene polymorphisms and DPN. Ethnicity is very important in genetic studies, and it will give us more information on the role of melatonin gene variants in larger study groups of diabetic patients of other ethnic origin.
Collapse
Affiliation(s)
- Özgül Ocak
- Department of Neurology, Çanakkale Onsekiz Mart University Faculty of Medicine, Çanakkale, Turkey
| | - Fatma Silan
- Department of Medical Genetics, Çanakkale Onsekiz Mart University Faculty of Medicine, Çanakkale, Turkey
| | - Erkan Melih Şahin
- Department of Family Medicine, Çanakkale Onsekiz Mart University Faculty of Medicine, Çanakkale, Turkey
| |
Collapse
|
39
|
Abstract
The pineal gland is a interface between light-dark cycle and shows neuro-endocrine functions. Melatonin is the primary hormone of pineal gland, secreted at night. The night-time melatonin peak regulates the physiological functions at dark. Melatonin has several unique features as it synchronises internal rhythm with daily and seasonal variations, regulates circadian rhythm and sleep-wake cycle. Physiologically melatonin involves in detoxification of free radicals, immune functions, neuro-protection, oncostatic effects, cardiovascular functions, reproduction, and foetal development. The precise functions of melatonin are exhibited by specific receptors. In relation to pathophysiology, impaired melatonin secretion promotes sleep disorder, cancer progression, type-2 diabetes, and neurodegenerative diseases. Several reports have highlighted the therapeutic benefits of melatonin specially related to cancer protection, sleep disorder, psychiatric disorders, and jet lag problems. This review will touch the most of the area of melatonin-oriented health impacts and its therapeutic aspects.
Collapse
|
40
|
Wu J, Tan X. The role of MTNR1B polymorphism on circadian rhythm-related cancer: A UK Biobank cohort study. Int J Cancer 2022; 151:888-896. [PMID: 35467761 PMCID: PMC9545001 DOI: 10.1002/ijc.34047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022]
Abstract
A common G risk allele in the melatonin receptor 1B (MTNR1B, rs10830963) gene has been associated with altered melatonin signaling and secretion. Given that melatonin possesses anticancerogenic properties, we hypothesized that breast and prostate cancer risks vary by rs10830963 genotype. A total of 216 702 participants from the UK Biobank without cancer at baseline (aged 56.4 ± 8.0 years, 50.79% female) were included. Multivariable Cox regression adjusting for known risk factors for breast or prostate cancer was used to estimate the independent effects of the rs10830963 SNP and chronotype on cancer risk. Over a median follow-up of 8 years, 2367 (2.15% of women) incidences of breast cancer and 2866 (2.69% of men) incidences of prostate cancer were documented in females and males, respectively. rs10830963 genotype is not associated with cancer risk independently (female Ptrend = .103, male Ptrend = .281). A late chronotype is associated with breast cancer risk in females (Ptrend = .014), but not prostate cancer risk in males (Ptrend = .915). Further stratification analysis revealed that the rs10830963 genotype is associated with a breast cancer risk in females with moderate evening chronotype (Ptrend = .001) and late chronotype is associated with breast cancer risk in females who carry rs10830963 G risk allele (Ptrend = .015). Our study suggests that having a late chronotype might increase the risk of breast cancer among females, while the effect of MTNR1B rs10830963 genotype on breast cancer risk is mediated by chronotype.
Collapse
Affiliation(s)
- Jiafei Wu
- Department of Surgical Sciences (Sleep Science Lab), Uppsala University, Uppsala, Sweden
| | - Xiao Tan
- Department of Surgical Sciences (Sleep Science Lab), Uppsala University, Uppsala, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
41
|
Hu S, Hu C, Luo L, Zhang H, Zhao S, Liu Z, Zeng L. Pu-erh tea increases the metabolite Cinnabarinic acid to improve circadian rhythm disorder-induced obesity. Food Chem 2022; 394:133500. [PMID: 35749873 DOI: 10.1016/j.foodchem.2022.133500] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/04/2022]
Abstract
Obesity is one of the circadian rhythm disorders (CRD)-mediated metabolic disorder syndromes. Pu-erh tea is a viable dietary intervention for CRD, however its effect on CRD-induced obesity is unclear. Here, we found that Pu-erh tea improved obesity in CRD-induced mice, which stemmed from the production of Cinnabarinic acid (CA). CA promoted adipose tissue lipolysis and thermogenic response (HSL, ATGL, Pparα, CKB, UCP1) and increased adipocyte sensitivity to hormones and neurotransmitters by targeting the expression of adipose tissue receptor proteins (Q6KAT8, P51655, A2AKQ0, M0QWX7, Q6ZQ33, and mGluR4). This improved mitochondrial activity and facilitated adipose tissue metabolic processes, thereby accelerating glucolipid metabolism. Also, CA-induced alterations in gut microbes and short-chain fatty acids further improved CRD-mediated lipid accumulation. These results suggest that the increase of CA caused by Pu-erh tea, targeted to adipose tissue via the metabolite-blood circulation-adipose tissue axis, maybe a key mechanism for reducing the development of CRD-induced obesity.
Collapse
Affiliation(s)
- Shanshan Hu
- College of Food Science, Southwest University, Beibei, Chongqing 400715, China
| | - Changhua Hu
- College of Pharmaceutical Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Liyong Luo
- College of Food Science, Southwest University, Beibei, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Beibei, Chongqing 400715, China
| | - Haotian Zhang
- College of Pharmaceutical Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Sibo Zhao
- College of Food Science, Southwest University, Beibei, Chongqing 400715, China
| | - Zhonghua Liu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China.
| | - Liang Zeng
- College of Food Science, Southwest University, Beibei, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
42
|
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
|
43
|
Molekulargenetische Diagnostik des Diabetes mellitus. DIABETOLOGE 2022. [DOI: 10.1007/s11428-022-00876-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Harmsen JF, Wefers J, Doligkeit D, Schlangen L, Dautzenberg B, Rense P, van Moorsel D, Hoeks J, Moonen-Kornips E, Gordijn MCM, van Marken Lichtenbelt WD, Schrauwen P. The influence of bright and dim light on substrate metabolism, energy expenditure and thermoregulation in insulin-resistant individuals depends on time of day. Diabetologia 2022; 65:721-732. [PMID: 35106618 PMCID: PMC8894310 DOI: 10.1007/s00125-021-05643-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/22/2021] [Indexed: 11/04/2022]
Abstract
AIMS/HYPOTHESIS In our modern society, artificial light is available around the clock and most people expose themselves to electrical light and light-emissive screens during the dark period of the natural light/dark cycle. Such suboptimal lighting conditions have been associated with adverse metabolic effects, and redesigning indoor lighting conditions to mimic the natural light/dark cycle more closely holds promise to improve metabolic health. Our objective was to compare metabolic responses to lighting conditions that resemble the natural light/dark cycle in contrast to suboptimal lighting in individuals at risk of developing metabolic diseases. METHODS Therefore, we here performed a non-blinded, randomised, controlled, crossover trial in which overweight insulin-resistant volunteers (n = 14) were exposed to two 40 h laboratory sessions with different 24 h lighting protocols while staying in a metabolic chamber under real-life conditions. In the Bright day-Dim evening condition, volunteers were exposed to electric bright light (~1250 lx) during the daytime (08:00-18:00 h) and to dim light (~5 lx) during the evening (18:00-23:00 h). Vice versa, in the Dim day-Bright evening condition, volunteers were exposed to dim light during the daytime and bright light during the evening. Randomisation and allocation to light conditions were carried out by sequential numbering. During both lighting protocols, we performed 24 h indirect calorimetry, and continuous core body and skin temperature measurements, and took frequent blood samples. The primary outcome was plasma glucose focusing on the pre- and postprandial periods of the intervention. RESULTS Spending the day in bright light resulted in a greater increase in postprandial triacylglycerol levels following breakfast, but lower glucose levels preceding the dinner meal at 18:00 h, compared with dim light (5.0 ± 0.2 vs 5.2 ± 0.2 mmol/l, n = 13, p=0.02). Dim day-Bright evening reduced the increase in postprandial glucose after dinner compared with Bright day-Dim evening (incremental AUC: 307 ± 55 vs 394 ± 66 mmol/l × min, n = 13, p=0.009). After the Bright day-Dim evening condition the sleeping metabolic rate was identical compared with the baseline night, whereas it dropped after Dim day-Bright evening. Melatonin secretion in the evening was strongly suppressed for Dim day-Bright evening but not for Bright day-Dim evening. Distal skin temperature for Bright day-Dim evening was lower at 18:00 h (28.8 ± 0.3°C vs 29.9 ± 0.4°C, n = 13, p=0.039) and higher at 23:00 h compared with Dim day-Bright evening (30.1 ± 0.3°C vs 28.8 ± 0.3°C, n = 13, p=0.006). Fasting and postprandial plasma insulin levels and the respiratory exchange ratio were not different between the two lighting protocols at any time. CONCLUSIONS/INTERPRETATION Together, these findings suggest that the indoor light environment modulates postprandial substrate handling, energy expenditure and thermoregulation of insulin-resistant volunteers in a time-of-day-dependent manner. TRIAL REGISTRATION ClinicalTrials.gov NCT03829982. FUNDING We acknowledge the financial support from the Netherlands Cardiovascular Research Initiative: an initiative with support from the Dutch Heart Foundation (CVON2014-02 ENERGISE).
Collapse
Affiliation(s)
- Jan-Frieder Harmsen
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jakob Wefers
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Daniel Doligkeit
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Luc Schlangen
- Human-Technology Interaction Group and Intelligent Lighting Institute, Department of Industrial Engineering and Innovation Sciences, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Bas Dautzenberg
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Pascal Rense
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Dirk van Moorsel
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Esther Moonen-Kornips
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marijke C M Gordijn
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
- Chrono@Work, Groningen, the Netherlands
| | - Wouter D van Marken Lichtenbelt
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|
45
|
Mason IC, Grimaldi D, Reid KJ, Warlick CD, Malkani RG, Abbott SM, Zee PC. Light exposure during sleep impairs cardiometabolic function. Proc Natl Acad Sci U S A 2022; 119:e2113290119. [PMID: 35286195 PMCID: PMC8944904 DOI: 10.1073/pnas.2113290119] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/30/2022] [Indexed: 12/11/2022] Open
Abstract
SignificanceAmbient nighttime light exposure is implicated as a risk factor for adverse health outcomes, including cardiometabolic disease. However, the effects of nighttime light exposure during sleep on cardiometabolic outcomes and the related mechanisms are unclear. This laboratory study shows that, in healthy adults, one night of moderate (100 lx) light exposure during sleep increases nighttime heart rate, decreases heart rate variability (higher sympathovagal balance), and increases next-morning insulin resistance when compared to sleep in a dimly lit (<3 lx) environment. Moreover, a positive relationship between higher sympathovagal balance and insulin levels suggests that sympathetic activation may play a role in the observed light-induced changes in insulin sensitivity.
Collapse
Affiliation(s)
- Ivy C. Mason
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA 02115
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115
| | - Daniela Grimaldi
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Kathryn J. Reid
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Chloe D. Warlick
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Roneil G. Malkani
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Sabra M. Abbott
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Phyllis C. Zee
- Center for Circadian and Sleep Medicine, Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
46
|
Gkouskou KK, Grammatikopoulou MG, Lazou E, Sanoudou D, Goulis DG, Eliopoulos AG. Genetically-Guided Medical Nutrition Therapy in Type 2 Diabetes Mellitus and Pre-diabetes: A Series of n-of-1 Superiority Trials. Front Nutr 2022; 9:772243. [PMID: 35265654 PMCID: PMC8899711 DOI: 10.3389/fnut.2022.772243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a heterogeneous metabolic disorder of multifactorial etiology that includes genetic and dietary influences. By addressing the latter, medical nutrition therapy (MNT) contributes to the management of T2DM or pre-diabetes toward achieving glycaemic control and improved insulin sensitivity. However, the clinical outcomes of MNT vary and may further benefit from personalized nutritional plans that take into consideration genetic variations associated with individual responses to macronutrients. The aim of the present series of n-of-1 trials was to assess the effects of genetically-guided vs. conventional MNT on patients with pre-diabetes or T2DM. A quasi-experimental, cross-over design was adopted in three Caucasian adult men with either diagnosis. Complete diet, bioclinical and anthropometric assessment was performed and a conventional MNT, based on the clinical practice guidelines was applied for 8 weeks. After a week of “wash-out,” a precision MNT was prescribed for an additional 8-week period, based on the genetic characteristics of each patient. Outcomes of interest included changes in body weight (BW), fasting plasma glucose (FPG), and blood pressure (BP). Collectively, the trials indicated improvements in BW, FPG, BP, and glycosylated hemoglobin (HbA1c) following the genetically-guided precision MNT intervention. Moreover, both patients with pre-diabetes experienced remission of the condition. We conclude that improved BW loss and glycemic control can be achieved in patients with pre-diabetes/T2DM, by coupling MNT to their genetic makeup, guiding optimal diet, macronutrient composition, exercise and oral nutrient supplementation in a personalized manner.
Collapse
Affiliation(s)
- Kalliopi K Gkouskou
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Embiodiagnostics Biology Research Company, Heraklion, Greece
| | - Maria G Grammatikopoulou
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Faculty of Health Sciences, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Nutritional Sciences and Dietetics, Faculty of Health Sciences, International Hellenic University, Thessaloniki, Greece
| | - Evgenia Lazou
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, Fourth Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Faculty of Health Sciences, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aristides G Eliopoulos
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
47
|
Manoogian ENC, Chow LS, Taub PR, Laferrère B, Panda S. Time-restricted Eating for the Prevention and Management of Metabolic Diseases. Endocr Rev 2022; 43:405-436. [PMID: 34550357 PMCID: PMC8905332 DOI: 10.1210/endrev/bnab027] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Indexed: 02/08/2023]
Abstract
Time-restricted feeding (TRF, animal-based studies) and time-restricted eating (TRE, humans) are an emerging behavioral intervention approach based on the understanding of the role of circadian rhythms in physiology and metabolism. In this approach, all calorie intake is restricted within a consistent interval of less than 12 hours without overtly attempting to reduce calories. This article will summarize the origin of TRF/TRE starting with concept of circadian rhythms and the role of chronic circadian rhythm disruption in increasing the risk for chronic metabolic diseases. Circadian rhythms are usually perceived as the sleep-wake cycle and dependent rhythms arising from the central nervous system. However, the recent discovery of circadian rhythms in peripheral organs and the plasticity of these rhythms in response to changes in nutrition availability raised the possibility that adopting a consistent daily short window of feeding can sustain robust circadian rhythm. Preclinical animal studies have demonstrated proof of concept and identified potential mechanisms driving TRF-related benefits. Pilot human intervention studies have reported promising results in reducing the risk for obesity, diabetes, and cardiovascular diseases. Epidemiological studies have indicated that maintaining a consistent long overnight fast, which is similar to TRE, can significantly reduce risks for chronic diseases. Despite these early successes, more clinical and mechanistic studies are needed to implement TRE alone or as adjuvant lifestyle intervention for the prevention and management of chronic metabolic diseases.
Collapse
Affiliation(s)
| | - Lisa S Chow
- University of Minnesota, Division of Diabetes, Endocrinology and Metabolism, Minneapolis, Minnesota 55455, USA
| | - Pam R Taub
- University of California, San Diego, Division of Cardiovascular Diseases, Department of Medicine, 9434 Medical Center Drive, La Jolla, California 92037, USA
| | - Blandine Laferrère
- New York Nutrition Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | | |
Collapse
|
48
|
Garaulet M, Lopez-Minguez J, Dashti HS, Vetter C, Hernández-Martínez AM, Pérez-Ayala M, Baraza JC, Wang W, Florez JC, Scheer FAJL, Saxena R. Interplay of Dinner Timing and MTNR1B Type 2 Diabetes Risk Variant on Glucose Tolerance and Insulin Secretion: A Randomized Crossover Trial. Diabetes Care 2022; 45:512-519. [PMID: 35015083 PMCID: PMC8918262 DOI: 10.2337/dc21-1314] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We tested whether the concurrence of food intake and elevated concentrations of endogenous melatonin, as occurs with late eating, results in impaired glucose control, in particular in carriers of the type 2 diabetes-associated G allele in the melatonin receptor-1B gene (MTNR1B). RESEARCH DESIGN AND METHODS In a Spanish natural late-eating population, a randomized, crossover study was performed. Each participant (n = 845) underwent two evening 2-h 75-g oral glucose tolerance tests following an 8-h fast: an early condition scheduled 4 h prior to habitual bedtime ("early dinner timing") and a late condition scheduled 1 h prior to habitual bedtime ("late dinner timing"), simulating an early and a late dinner timing, respectively. Differences in postprandial glucose and insulin responses between early and late dinner timing were determined using incremental area under the curve (AUC) calculated by the trapezoidal method. RESULTS Melatonin serum levels were 3.5-fold higher in the late versus early condition, with late dinner timing resulting in 6.7% lower insulin AUC and 8.3% higher glucose AUC. The effect of late eating impairing glucose tolerance was stronger in the MTNR1B G-allele carriers than in noncarriers. Genotype differences in glucose tolerance were attributed to reductions in β-cell function (P for interaction, Pint glucose area under the curve = 0.009, Pint corrected insulin response = 0.022, and Pint disposition index = 0.018). CONCLUSIONS Concurrently high endogenous melatonin and carbohydrate intake, as typical for late eating, impairs glucose tolerance, especially in MTNR1B G-risk allele carriers, attributable to insulin secretion defects.
Collapse
Affiliation(s)
- Marta Garaulet
- Department of Physiology, University of Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, Murcia, Spain.,Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| | - Jesus Lopez-Minguez
- Department of Physiology, University of Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, Murcia, Spain
| | - Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA.,Broad Institute, Cambridge, MA.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Céline Vetter
- Broad Institute, Cambridge, MA.,Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | | | - Millán Pérez-Ayala
- Department of Clinical Analysis, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Juan Carlos Baraza
- Department of Physiology, University of Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, Murcia, Spain
| | - Wei Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Jose C Florez
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA.,Broad Institute, Cambridge, MA.,Department of Medicine, Harvard Medical School, Boston, MA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA.,Broad Institute, Cambridge, MA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA.,Broad Institute, Cambridge, MA.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
49
|
Xue P, Tan X, Wu J, Tang X, Benedict C. No association between a common type 2 diabetes risk gene variant in the melatonin receptor gene (MTNR1B) and mortality among type 2 diabetes patients. J Pineal Res 2022; 72:e12785. [PMID: 34967052 DOI: 10.1111/jpi.12785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023]
Abstract
The minor G risk allele in the common melatonin receptor gene (MTNR1B, rs10830963) has been associated with an increased risk of myocardial infarction among patients with type 2 diabetes (T2D). Furthermore, activating the melatonin receptor 1B through melatonin has been shown to promote cell proliferation, which could be hypothesized to increase cancer risk. Cardiovascular disease (CVD) and cancer are common causes of death among patients with T2D. Using data from 14 736 patients with T2D who participated in the UK Biobank investigation, we hypothesized an additive effect of the G risk allele on all-cause mortality, CVD mortality, and cancer mortality. As shown by Cox regression adjusted for confounders such as age, glucose-lowering medication, and socioeconomic status, no significant trend between the number of G risk alleles and mortality outcomes was found during the follow-up period of 11.1 years. Our negative findings do not speak against the role of this gene variant in the development of T2D, as repeatedly shown by previous large-scale studies. Instead, they may suggest that rs10830963 is less relevant for mortality risk in patients with T2D.
Collapse
Affiliation(s)
- Pei Xue
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao Tan
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
| | - Jiafei Wu
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Christian Benedict
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
| |
Collapse
|
50
|
van Poppel MNM, Corcoy R, Hill D, Simmons D, Mendizabal L, Zulueta M, Simon L, Desoye G. Interaction between rs10830962 polymorphism in MTNR1B and lifestyle intervention on maternal and neonatal outcomes: secondary analyses of the DALI lifestyle randomized controlled trial. Am J Clin Nutr 2022; 115:388-396. [PMID: 34669935 DOI: 10.1093/ajcn/nqab347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Interactions between polymorphisms of the melatonin receptor 1B (MTNR1B) gene and lifestyle intervention for gestational diabetes have been described. Whether these are specific for physical activity or the healthy eating intervention is unknown. OBJECTIVES The aim was to assess the interaction between MTNR1B rs10830962 and rs10830963 polymorphisms and lifestyle interventions during pregnancy. METHODS Women with a BMI (in kg/m2) of ≥29 (n = 436) received counseling on healthy eating (HE), physical activity (PA), or both. The control group received usual care. This secondary analysis had a factorial design with comparison of HE compared with no HE and PA compared with no PA. Maternal outcomes at 24-28 wk were gestational weight gain (GWG), maternal fasting glucose, insulin, insulin resistance (HOMA-IR), disposition index, and development of GDM. Neonatal outcomes were cord blood leptin and C-peptide and estimated neonatal fat percentage. The interaction between receiving either the HE or PA intervention and genotypes of both rs10830962 and rs10830963 was assessed using multilevel regression analysis. RESULTS GDM risk was increased in women homozygous for the G allele of rs10830962 (OR: 2.60; 95% CI: 1.34, 5.06) or rs10830963 (OR: 2.83; 95% CI: 1.24, 6.47). Significant interactions between rs10830962 and interventions were found: in women homozygous for the G allele but not in the other genotypes, the PA intervention reduced maternal fasting insulin (β: -0.16; 95% CI: -0.33, 0.02; P = 0.08) and HOMA-IR (β: -0.17; 95% CI: -0.35, 0.01; P = 0.06), and reduced cord blood leptin (β: -0.84; 95% CI: -1.42, -0.25; P = 0.01) and C-peptide (β: -0.62; 95% CI: -1.07, -0.17; P = 0.01). In heterozygous women, the HE intervention had no effect, whereas in women homozygous for the C allele, HE intervention reduced GWG (β: -1.6 kg; 95% CI: -2.4, -0.8 kg). No interactions were found. CONCLUSIONS In women homozygous for the risk allele of MTNR1B rs10830962, GDM risk was increased and PA intervention might be more beneficial than HE intervention for reducing maternal insulin resistance, cord blood C-peptide, and cord blood leptin.
Collapse
Affiliation(s)
| | - Rosa Corcoy
- Institut de Recerca de l´Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBER Bioengineering, Biomaterials and Nanotechnology, Instituto de Salud Carlos III, Madrid, Spain
| | - David Hill
- Recherche en Santé Lawson SA, Bronschhofen, Switzerland.,Lawson Health Research Institute, London, Ontario, Canada
| | - David Simmons
- Western Sydney University, Campbelltown, New South Wales, Australia.,Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | | | - Laureano Simon
- Department of Obstetrics and Gynecology, Medical University Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University Graz, Graz, Austria
| | | |
Collapse
|