1
|
Zhang M, Ward J, Strawbridge RJ, Anderson JJ, Celis-Morales C, Pell JP, Ho FK, Lyall DM. Genetic predisposition to adiposity, and type 2 diabetes: the role of lifestyle and phenotypic adiposity. Eur J Endocrinol 2025; 192:549-557. [PMID: 40315335 PMCID: PMC12056655 DOI: 10.1093/ejendo/lvaf084] [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: 01/21/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
AIMS Genetic predisposition to adiposity is associated with type 2 diabetes (T2D), even in the absence of phenotypic adiposity (obesity and central obesity). We aimed to quantify the overall contribution of obesity and modifiable lifestyle factors to the association between genetic predisposition to adiposity and the development of T2D. METHODS This prospective cohort study involved 220 703 White British participants from the UK Biobank. It examined the associations between genetic predisposition to adiposity [body mass index polygenic risk (BMI-PRS) and waist-hip ratio polygenic risk (WHR-PRS)] and incident T2D, as well as interactions and mediation via lifestyle factors (diet quality, physical activity levels, total energy intake, sleep duration, and smoking and alcohol intake) and phenotypic adiposity. RESULTS People with high phenotypic adiposity and high adiposity PRS values (>1 SD above the mean) had the highest risk of incident T2D (versus non-obese/central obese and non-high PRS). This was the case for BMI-PRS [hazard ratio (HR) = 3.72] and WHR-PRS (HR = 4.17). Lifestyle factors explained 30.5% of the BMI-PRS/T2D association (2.0% mediation; 28.5% effect modification), and lifestyle and obesity together explained 92.1% (78.8% mediation; 13.3% effect modification). Lifestyle factors explained 20.4% of the WHR-PRS/T2D association (3.4% mediation; 17.0% effect modification), and lifestyle and central obesity together explained 72.8% (41.1% mediation; 31.7% effect modification). CONCLUSIONS Whilst phenotypic adiposity explains a large proportion of the association between BMI-PRS/WHR-PRS and T2D, modifiable lifestyle factors also make contributions. Promoting healthy lifestyles among people prone to adiposity is important in reducing the global burden of T2D.
Collapse
Affiliation(s)
- Mengrong Zhang
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
- Department of Medicine Solna, Karolinska Institute, Stockholm 17177, Sweden
| | - Jana J Anderson
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Carlos Celis-Morales
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, United Kingdom
- Human Performance Lab, Education, Physical Activity, and Health Research Unit, Universidad Católica del Maule, Talca 115 3745, Chile
- Centro de Investigación en Medicina de Altura (CEIMA), Universidad Arturo Prat, Iquique 1100012, Chile
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Frederick K Ho
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Donald M Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| |
Collapse
|
2
|
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
|
3
|
He J, Liu F, Xu P, Xu T, Yu H, Wu B, Wang H, Chen J, Zhang K, Zhang J, Meng K, Yan X, Yang Q, Zhang X, Sun D, Chen X. Aerobic Exercise Improves the Overall Outcome of Type 2 Diabetes Mellitus Among People With Mental Disorders. Depress Anxiety 2024; 2024:6651804. [PMID: 40226688 PMCID: PMC11918971 DOI: 10.1155/da/6651804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/09/2024] [Indexed: 04/15/2025] Open
Abstract
The escalating global prevalence of type 2 diabetes mellitus (T2DM) and mental disorder (MD) including schizophrenia, bipolar disorder, major depressive disorder, and anxiety highlights the urgency for comprehensive therapeutic strategies. Aerobic exercise (AE) is a viable adjunct therapy, providing significant benefits for individuals dealing with both T2DM and MD. This review consolidates evidence on AE's role in alleviating the physiological and psychological effects of these comorbid conditions. It delves into the pathophysiological connections between T2DM and various MD, including depression, schizophrenia, anxiety, and bipolar disorder-emphasizing their reciprocal exacerbation. Key neurophysiological mechanisms through which AE confers benefits are explored, including neuroinflammation modulation, brain structure and neuroplasticity enhancement, growth factor expression regulation, and hypothalamic-pituitary-adrenal (HPA)/microbiota-gut-brain (MGB) axis normalization. Clinical results indicate that AE significantly improves both metabolic and psychological parameters in patients with T2DM and MD, providing a substantial argument for integrating AE into comprehensive treatment plans. Future research should aim to establish detailed, personalized exercise prescriptions and explore the long-term benefits of AE in this population. This review underscores the potential of AE to complement existing therapeutic modalities and enhance the management of patients with T2DM and MD.
Collapse
Affiliation(s)
- Jiaxuan He
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Fan Liu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Peiye Xu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Ting Xu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Baihui Wu
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Jia Chen
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611100, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Junbei Zhang
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Kaikai Meng
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Xiaoqing Yan
- The Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Da Sun
- Institute of Life Sciences and Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Xia Chen
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| |
Collapse
|
4
|
Yu L, Liu W, Zhang Y, Tan Q, Song J, Fan L, You X, Zhou M, Wang B, Chen W. Styrene and ethylbenzene exposure and type 2 diabetes mellitus: A longitudinal gene-environment interaction study. ECO-ENVIRONMENT & HEALTH 2024; 3:452-457. [PMID: 39559189 PMCID: PMC11570399 DOI: 10.1016/j.eehl.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/12/2024] [Accepted: 07/21/2024] [Indexed: 11/20/2024]
Abstract
Styrene and ethylbenzene (S/EB) are identified as hazardous air contaminants that raise significant concerns. The association between S/EB exposure and the incidence of type 2 diabetes mellitus (T2DM), and the interaction between genes and environment, remains poorly understood. Our study consisted of 2219 Chinese adults who were part of the Wuhan-Zhuhai cohort. A follow-up assessment was conducted after six years. Exposure to S/EB was quantified by determining the concentrations of urinary biomarkers of exposure to S/EB (UBE-S/EB; urinary phenylglyoxylic acid level plus urinary mandelic acid level). Logistic regression models were constructed to investigate the relations of UBE-S/EB and genetic risk score (GRS) with T2DM prevalence and incidence. The interaction effects of UBE-S/EB and GRS on T2DM were investigated on multiplicative and additive scales. UBE-S/EB was dose-dependently and positively related to T2DM prevalence and incidence. Participants with high levels of UBE-S/EB [relative risk (RR) = 1.930, 95% confidence interval (CI): 1.157-3.309] or GRS (1.943, 1.110-3.462) demonstrated the highest risk of incident T2DM, in comparison to those with low levels of UBE-S/EB or GRS. Significant additive interaction between UBE-S/EB and GRS on T2DM incidence was discovered with relative excess risk due to interaction (95% CI) of 0.178 (0.065-0.292). The RR (95% CI) of T2DM incidence was 2.602 (1.238-6.140) for individuals with high UBE-S/EB and high GRS, compared to those with low UBE-S/EB and low GRS. This study presented the initial evidence that S/EB exposure was significantly related to increased risk of T2DM incidence, and the relationship was interactively aggravated by genetic predisposition.
Collapse
Affiliation(s)
- Linling Yu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Public Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongfang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiyou Tan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiahao Song
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lieyang Fan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaojie You
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
5
|
Sokolowski EK, Kursawe R, Selvam V, Bhuiyan RM, Thibodeau A, Zhao C, Spracklen CN, Ucar D, Stitzel ML. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab 2024; 36:2468-2488.e7. [PMID: 39383866 PMCID: PMC11798411 DOI: 10.1016/j.cmet.2024.09.006] [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: 12/22/2023] [Revised: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses and whether T2D-associated genetic variants modulate them is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress responsive, modulated largely in an ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked the rs6917676-T T2D risk allele to increased islet ER-stress-responsive CRE accessibility and allele-specific β cell nuclear factor binding. MAP3K5, the ER-stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β cell abundance and was elevated in T2D β cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.
Collapse
Affiliation(s)
- Eishani K Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
6
|
Wuni R, Ventura EF, Curi-Quinto K, Murray C, Nunes R, Lovegrove JA, Penny M, Favara M, Sanchez A, Vimaleswaran KS. Interactions between genetic and lifestyle factors on cardiometabolic disease-related outcomes in Latin American and Caribbean populations: A systematic review. Front Nutr 2023; 10:1067033. [PMID: 36776603 PMCID: PMC9909204 DOI: 10.3389/fnut.2023.1067033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction The prevalence of cardiometabolic diseases has increased in Latin American and the Caribbean populations (LACP). To identify gene-lifestyle interactions that modify the risk of cardiometabolic diseases in LACP, a systematic search using 11 search engines was conducted up to May 2022. Methods Eligible studies were observational and interventional studies in either English, Spanish, or Portuguese. A total of 26,171 publications were screened for title and abstract; of these, 101 potential studies were evaluated for eligibility, and 74 articles were included in this study following full-text screening and risk of bias assessment. The Appraisal tool for Cross-Sectional Studies (AXIS) and the Risk Of Bias In Non-Randomized Studies-of Interventions (ROBINS-I) assessment tool were used to assess the methodological quality and risk of bias of the included studies. Results We identified 122 significant interactions between genetic and lifestyle factors on cardiometabolic traits and the vast majority of studies come from Brazil (29), Mexico (15) and Costa Rica (12) with FTO, APOE, and TCF7L2 being the most studied genes. The results of the gene-lifestyle interactions suggest effects which are population-, gender-, and ethnic-specific. Most of the gene-lifestyle interactions were conducted once, necessitating replication to reinforce these results. Discussion The findings of this review indicate that 27 out of 33 LACP have not conducted gene-lifestyle interaction studies and only five studies have been undertaken in low-socioeconomic settings. Most of the studies were cross-sectional, indicating a need for longitudinal/prospective studies. Future gene-lifestyle interaction studies will need to replicate primary research of already studied genetic variants to enable comparison, and to explore the interactions between genetic and other lifestyle factors such as those conditioned by socioeconomic factors and the built environment. The protocol has been registered on PROSPERO, number CRD42022308488. Systematic review registration https://clinicaltrials.gov, identifier CRD420223 08488.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | - Eduard F. Ventura
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | | | - Claudia Murray
- Department of Real Estate and Planning, University of Reading, Reading, United Kingdom
| | - Richard Nunes
- Department of Real Estate and Planning, University of Reading, Reading, United Kingdom
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | - Mary Penny
- Instituto de Investigación Nutricional, Lima, Peru
| | - Marta Favara
- Oxford Department of International Development, University of Oxford, Oxford, United Kingdom
| | - Alan Sanchez
- Grupo de Análisis para el Desarrollo (GRADE), Lima, Peru
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading, United Kingdom
| |
Collapse
|
7
|
Hosseinpour-Niazi S, Mirmiran P, Hosseini S, Hadaegh F, Ainy E, Daneshpour MS, Azizi F. Effect of TCF7L2 on the relationship between lifestyle factors and glycemic parameters: a systematic review. Nutr J 2022; 21:59. [PMID: 36155628 PMCID: PMC9511734 DOI: 10.1186/s12937-022-00813-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/14/2022] [Indexed: 11/12/2022] Open
Abstract
Background Among candidate genes related to type 2 diabetes (T2DM), one of the strongest genes is Transcription factor 7 like 2 (TCF7L2), regarding the Genome-Wide Association Studies. We aimed to conduct a systematic review of the literature on the modification effect of TCF7L2 on the relation between glycemic parameters and lifestyle factors. Methods A systematic literature search was done for relevant publications using electronic databases, including PubMed, EMBASE, Scopus, and Web of Science, from January 1, 2000, to November 2, 2021. Results Thirty-eight studies (16 observational studies, six meal test trials, and 16 randomized controlled trials (RCTs)) were included. Most observational studies had been conducted on participants with non-diabetes showing that TCF7L2 modified the association between diet (fatty acids and fiber) and insulin resistance. In addition, findings from meal test trials showed that, compared to non-risk-allele carriers, consumption of meals with different percentages of total dietary fat in healthy risk-allele carriers increased glucose concentrations and impaired insulin sensitivity. However, ten RCTs, with intervention periods of less than ten weeks and more than one year, showed that TCF7L2 did not modify glycemic parameters in response to a dietary intervention involving different macronutrients. However, two weight loss dietary RCTs with more than 1-year duration showed that serum glucose and insulin levels decreased and insulin resistance improved in non-risk allele subjects with overweight/obesity. Regarding artichoke extract supplementation (ALE), two RCTs observed that ALE supplementation significantly decreased insulin concentration and improved insulin resistance in the TT genotype of the rs7903146 variant of TCF7L2. In addition, four studies suggested that physical activity levels and smoking status modified the association between TCF7L2 and glycemic parameters. However, three studies observed no effect of TCF7L2 on glycemic parameters in participants with different levels of physical activity and smoking status. Conclusion The modification effects of TCF7L2 on the relation between the lifestyle factors (diet, physical activity, and smoking status) and glycemic parameters were contradictory. PROSPERO registration number CRD42020196327 Supplementary Information The online version contains supplementary material available at 10.1186/s12937-022-00813-w.
Collapse
Affiliation(s)
- Somayeh Hosseinpour-Niazi
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shabnam Hosseini
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elaheh Ainy
- Department of Vice Chancellor Research Affairs, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Vu THT, Beck T, Bennett DA, Schneider JA, Hayden KM, Shadyab AH, Rajan KB, Morris MC, Cornelis MC. Adherence to MIND Diet, Genetic Susceptibility, and Incident Dementia in Three US Cohorts. Nutrients 2022; 14:2759. [PMID: 35807939 PMCID: PMC9268772 DOI: 10.3390/nu14132759] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Adherence to Mediterranean-DASH Diet Intervention for Neurodegenerative Delay (MIND) may lower the risk of dementia by impacting immunity and cholesterol, which are pathways also implicated by genome-wide association studies of Alzheimer’s Dementia (AD). We examined whether adherence to the MIND diet could modify the association of genetic risk for AD with incident dementia. We used three ongoing US cohorts: Chicago Health and Aging Project (CHAP, n = 2449), Rush Memory and Aging Project (MAP, n = 725), and Women’s Health Initiative Memory Study (WHIMS, n = 5308). Diagnosis of dementia was based on clinical neurological examination and standardized criteria. Repeated measures of global cognitive function were available in MAP and CHAP. Self-reported adherence to MIND was estimated using food-frequency questionnaires. Global and pathway-specific genetic scores (GS) for AD were derived. Cox proportional hazard, logistic regression, and mixed models were used to examine associations of MIND, GS, and GS-MIND interactions with incident dementia and cognitive decline. Higher adherence to MIND and lower GS were associated with a lower risk of dementia in MAP and WHIMS and a slower rate of cognitive decline in MAP (p < 0.05). MIND or GS were not associated with incident dementia or cognitive decline in CHAP. No gene−diet interaction was replicated across cohorts. Genetic risk and MIND adherence are independently associated with dementia among older US men and women.
Collapse
Affiliation(s)
- Thanh Huyen T. Vu
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Todd Beck
- Rush Institute for Healthy Aging, Chicago, IL 60612, USA; (T.B.); (K.B.R.)
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Chicago, IL 60612, USA; (D.A.B.); (J.A.S.)
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Chicago, IL 60612, USA; (D.A.B.); (J.A.S.)
| | - Kathleen M. Hayden
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA;
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, USA;
| | - Kumar B. Rajan
- Rush Institute for Healthy Aging, Chicago, IL 60612, USA; (T.B.); (K.B.R.)
| | | | - Marilyn C. Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| |
Collapse
|
9
|
Chen HH, Petty LE, North KE, McCormick JB, Fisher-Hoch SP, Gamazon ER, Below JE. OUP accepted manuscript. Hum Mol Genet 2022; 31:3191-3205. [PMID: 35157052 PMCID: PMC9476627 DOI: 10.1093/hmg/ddac039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Type 2 diabetes is a complex, systemic disease affected by both genetic and environmental factors. Previous research has identified genetic variants associated with type 2 diabetes risk; however, gene regulatory changes underlying progression to metabolic dysfunction are still largely unknown. We investigated RNA expression changes that occur during diabetes progression using a two-stage approach. In our discovery stage, we compared changes in gene expression using two longitudinally collected blood samples from subjects whose fasting blood glucose transitioned to a level consistent with type 2 diabetes diagnosis between the time points against those who did not with a novel analytical network approach. Our network methodology identified 17 networks, one of which was significantly associated with transition status. This 822-gene network harbors many genes novel to the type 2 diabetes literature but is also significantly enriched for genes previously associated with type 2 diabetes. In the validation stage, we queried associations of genetically determined expression with diabetes-related traits in a large biobank with linked electronic health records. We observed a significant enrichment of genes in our identified network whose genetically determined expression is associated with type 2 diabetes and other metabolic traits and validated 31 genes that are not near previously reported type 2 diabetes loci. Finally, we provide additional functional support, which suggests that the genes in this network are regulated by enhancers that operate in human pancreatic islet cells. We present an innovative and systematic approach that identified and validated key gene expression changes associated with type 2 diabetes transition status and demonstrated their translational relevance in a large clinical resource.
Collapse
Affiliation(s)
- Hung-Hsin Chen
- Vanderbilt Genetics Institute and Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren E Petty
- Vanderbilt Genetics Institute and Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joseph B McCormick
- The University of Texas Health Science Center at Houston (UTHealth) School of Public Health, Brownsville, TX 78520, USA
| | - Susan P Fisher-Hoch
- The University of Texas Health Science Center at Houston (UTHealth) School of Public Health, Brownsville, TX 78520, USA
| | - Eric R Gamazon
- Vanderbilt Genetics Institute and Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Clare Hall, University of Cambridge, Cambridgeshire, UK
| | - Jennifer E Below
- To whom correspondence should be addressed. Tel: +1-615-343-1655;
| |
Collapse
|
10
|
Lu X, Fan K, Ren J, Wu C. Identifying Gene-Environment Interactions With Robust Marginal Bayesian Variable Selection. Front Genet 2021; 12:667074. [PMID: 34956304 PMCID: PMC8693717 DOI: 10.3389/fgene.2021.667074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/13/2021] [Indexed: 01/02/2023] Open
Abstract
In high-throughput genetics studies, an important aim is to identify gene–environment interactions associated with the clinical outcomes. Recently, multiple marginal penalization methods have been developed and shown to be effective in G×E studies. However, within the Bayesian framework, marginal variable selection has not received much attention. In this study, we propose a novel marginal Bayesian variable selection method for G×E studies. In particular, our marginal Bayesian method is robust to data contamination and outliers in the outcome variables. With the incorporation of spike-and-slab priors, we have implemented the Gibbs sampler based on Markov Chain Monte Carlo (MCMC). The proposed method outperforms a number of alternatives in extensive simulation studies. The utility of the marginal robust Bayesian variable selection method has been further demonstrated in the case studies using data from the Nurse Health Study (NHS). Some of the identified main and interaction effects from the real data analysis have important biological implications.
Collapse
Affiliation(s)
- Xi Lu
- Department of Statistics, Kansas State University, Manhattan, KS, United States
| | - Kun Fan
- Department of Statistics, Kansas State University, Manhattan, KS, United States
| | - Jie Ren
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Cen Wu
- Department of Statistics, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
11
|
Paternal Exercise Improves the Metabolic Health of Offspring via Epigenetic Modulation of the Germline. Int J Mol Sci 2021; 23:ijms23010001. [PMID: 35008427 PMCID: PMC8744992 DOI: 10.3390/ijms23010001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND/AIMS Epigenetic regulation is considered the main molecular mechanism underlying the developmental origin of health and disease's (DOHAD) hypothesis. Previous studies that have investigated the role of paternal exercise on the metabolic health of the offspring did not control for the amount and intensity of the training or possible effects of adaptation to exercise and produced conflicting results regarding the benefits of parental exercise to the next generation. We employed a precisely regulated exercise regimen to study the transgenerational inheritance of improved metabolic health. METHODS We subjected male mice to a well-controlled exercise -training program to investigate the effects of paternal exercise on glucose tolerance and insulin sensitivity in their adult progeny. To investigate the molecular mechanisms of epigenetic inheritance, we determined chromatin markers in the skeletal muscle of the offspring and the paternal sperm. RESULTS Offspring of trained male mice exhibited improved glucose homeostasis and insulin sensitivity. Paternal exercise modulated the DNA methylation profile of PI3Kca and the imprinted H19/Igf2 locus at specific differentially methylated regions (DMRs) in the skeletal muscle of the offspring, which affected their gene expression. Remarkably, a similar DNA methylation profile at the PI3Kca, H19, and Igf2 genes was present in the progenitor sperm indicating that exercise-induced epigenetic changes that occurred during germ cell development contributed to transgenerational transmission. CONCLUSION Paternal exercise might be considered as a strategy that could promote metabolic health in the offspring as the benefits can be inherited transgenerationally.
Collapse
|
12
|
Chen Y, Qie X, Quan W, Zeng M, Qin F, Chen J, Adhikari B, He Z. Omnifarious fruit polyphenols: an omnipotent strategy to prevent and intervene diabetes and related complication? Crit Rev Food Sci Nutr 2021:1-37. [PMID: 34792409 DOI: 10.1080/10408398.2021.2000932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a metabolic syndrome which cannot be cured. Recently, considerable interest has been focused on food ingredients to prevent and intervene in complications of diabetes. Polyphenolic compounds are one of the bioactive phytochemical constituents with various biological activities, which have drawn increasing interest in human health. Fruits are part of the polyphenol sources in daily food consumption. Fruit-derived polyphenols possess the anti-diabetic activity that has already been proved either from in vitro studies or in vivo studies. The mechanisms of fruit polyphenols in treating diabetes and related complications are under discussion. This is a comprehensive review on polyphenols from the edible parts of fruits, including those from citrus, berries, apples, cherries, mangoes, mangosteens, pomegranates, and other fruits regarding their potential benefits in preventing and treating diabetes mellitus. The signal pathways of characteristic polyphenols derived from fruits in reducing high blood glucose and intervening hyperglycemia-induced diabetic complications were summarized.
Collapse
Affiliation(s)
- Yao Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Xuejiao Qie
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Quan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Fang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Jie Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Benu Adhikari
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Zhiyong He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
13
|
Zhuang P, Liu X, Li Y, Wan X, Wu Y, Wu F, Zhang Y, Jiao J. Effect of Diet Quality and Genetic Predisposition on Hemoglobin A 1c and Type 2 Diabetes Risk: Gene-Diet Interaction Analysis of 357,419 Individuals. Diabetes Care 2021; 44:2470-2479. [PMID: 34433621 DOI: 10.2337/dc21-1051] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/29/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the interactions between diet quality and genetic predisposition to incident type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS Between 2006 and 2010, 357,419 participants with genetic and complete dietary data from the UK Biobank were enrolled and prospectively followed up to 2017. The genetic risk score (GRS) was calculated on the basis of 424 variants associated with T2D risk, and a higher GRS indicates a higher genetic predisposition to T2D. The adherence to a healthy diet was assessed by a diet quality score comprising 10 important dietary components, with a higher score representing a higher overall diet quality. RESULTS There were 5,663 incident T2D cases documented during an average of 8.1 years of follow-up. A significant negative interaction was observed between the GRS and the diet quality score. After adjusting for major risk factors, per SD increment in the GRS and the diet quality score was associated with a 54% higher and a 9% lower risk of T2D, respectively. A simultaneous increment of 1 SD in both the diet quality score and GRS was additionally associated with a 3% lower T2D risk due to the antagonistic interaction. In categorical analyses, a sharp reduction of 23% in T2D risk associated with a 1-SD increment in the diet quality score was detected among participants in the extremely high GRS group (GRS >95%). We also observed a strong negative interaction between the GRS and the diet quality score on the blood HbA1c level at baseline (P < 0.001). CONCLUSIONS The adherence to a healthy diet was associated with more reductions in blood HbA1c levels and subsequent T2D risk among individuals with a higher genetic risk. Our findings support tailoring dietary recommendations to an individual's genetic makeup for T2D prevention.
Collapse
Affiliation(s)
- Pan Zhuang
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohui Liu
- Department of Nutrition, School of Public Health, and Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yin Li
- Department of Nutrition, School of Public Health, and Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuzhi Wan
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuqi Wu
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fei Wu
- Department of Nutrition, School of Public Health, and Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, and Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Wang J, Ning J, Shete S. Mediation model with a categorical exposure and a censored mediator with application to a genetic study. PLoS One 2021; 16:e0257628. [PMID: 34637449 PMCID: PMC8509986 DOI: 10.1371/journal.pone.0257628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Mediation analysis is a statistical method for evaluating the direct and indirect effects of an exposure on an outcome in the presence of a mediator. Mediation models have been widely used to determine direct and indirect contributions of genetic variants in clinical phenotypes. In genetic studies, the additive genetic model is the most commonly used model because it can detect effects from either recessive or dominant models (or any model in between). However, the existing approaches for mediation model cannot be directly applied when the genetic model is additive (e.g. the most commonly used model for SNPs) or categorical (e.g. polymorphic loci), and thus modification to measures of indirect and direct effects is warranted. In this study, we proposed overall measures of indirect, direct, and total effects for a mediation model with a categorical exposure and a censored mediator, which accounts for the frequency of different values of the categorical exposure. The proposed approach provides the overall contribution of the categorical exposure to the outcome variable. We assessed the empirical performance of the proposed overall measures via simulation studies and applied the measures to evaluate the mediating effect of a women's age at menopause on the association between genetic variants and type 2 diabetes.
Collapse
Affiliation(s)
- Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
15
|
Amador C, Zeng Y, Barber M, Walker RM, Campbell A, McIntosh AM, Evans KL, Porteous DJ, Hayward C, Wilson JF, Navarro P, Haley CS. Genome-wide methylation data improves dissection of the effect of smoking on body mass index. PLoS Genet 2021; 17:e1009750. [PMID: 34499657 PMCID: PMC8428545 DOI: 10.1371/journal.pgen.1009750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/28/2021] [Indexed: 11/18/2022] Open
Abstract
Variation in obesity-related traits has a genetic basis with heritabilities between 40 and 70%. While the global obesity pandemic is usually associated with environmental changes related to lifestyle and socioeconomic changes, most genetic studies do not include all relevant environmental covariates, so the genetic contribution to variation in obesity-related traits cannot be accurately assessed. Some studies have described interactions between a few individual genes linked to obesity and environmental variables but there is no agreement on their total contribution to differences between individuals. Here we compared self-reported smoking data and a methylation-based proxy to explore the effect of smoking and genome-by-smoking interactions on obesity related traits from a genome-wide perspective to estimate the amount of variance they explain. Our results indicate that exploiting omic measures can improve models for complex traits such as obesity and can be used as a substitute for, or jointly with, environmental records to better understand causes of disease.
Collapse
Affiliation(s)
- Carmen Amador
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Yanni Zeng
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, China
| | - Michael Barber
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Rosie M. Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, Chancellor’s Building, 49 Little France Crescent, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew M. McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris S. Haley
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Qie R, Han M, Huang S, Li Q, Liu L, Zhang D, Cheng C, Zhao Y, Liu D, Qin P, Guo C, Zhou Q, Tian G, Zhang Y, Wu X, Wu Y, Li Y, Yang X, Zhao Y, Feng Y, Hu F, Zhang M, Hu D, Lu J. Association of TCF7L2 gene polymorphisms, methylation, and gene-environment interaction with type 2 diabetes mellitus risk: A nested case-control study in the Rural Chinese Cohort Study. J Diabetes Complications 2021; 35:107829. [PMID: 33419631 DOI: 10.1016/j.jdiacomp.2020.107829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/08/2020] [Accepted: 12/01/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND To assess the associations of single-nucleotide polymorphisms (SNPs) and methylation of transcription factor 7-like 2 (TCF7L2) gene with type 2 diabetes mellitus (T2DM) risk and further explore the interactions among SNPs, methylation, and environmental factors involved in T2DM risk. METHODS We conducted a nested case-control study with 290 pairs of T2DM cases and matched controls. We genotyped 3 SNPs of TCF7L2 in all included participants and tested 14 CpG loci of TCF7L2 in 76 pairs of cases and controls. Conditional logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (95% CIs) for T2DM risk according to SNPs and methylation of TCF7L2. Multifactor dimensionality reduction (MDR) analysis was used to explore the potential TCF7L2 gene-environment interactions in T2DM risk. RESULTS We found no statistically significant association between the TCF7L2 polymorphisms and T2DM risk. We observed significant positive associations of methylation at CpG5 and CpG7_8 with T2DM risk. For each 1% increase in DNA methylation at CpG5 and CpG7_8, T2DM risk increased 12% (OR 1.12, 95% CI 1.01-1.25) and 32% (OR 1.32, 95% CI 1.07-1.63), respectively. Additionally, MDR analyses identified significant SNP-environment interactions among rs290487, alcohol drinking, and hypertension and methylation-environment interactions among CpG5, CpG7_8 and hypertension (P <0.05). CONCLUSIONS TCF7L2 polymorphisms were not independently associated with T2DM risk. However, TCF7L2 methylation were positively associated with T2DM risk in rural Chinese adults. Interactions among TCF7L2 polymorphisms, TCF7L2 methylation and environmental factors also suggest a possible etiologic pattern for T2DM.
Collapse
Affiliation(s)
- Ranran Qie
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Minghui Han
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shengbing Huang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Quanman Li
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Leilei Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Dongdong Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Cheng Cheng
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Dechen Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Pei Qin
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Chunmei Guo
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qionggui Zhou
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Gang Tian
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yanyan Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xiaoyan Wu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yuying Wu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yang Li
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Xingjin Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yifei Feng
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Dongsheng Hu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| | - Jie Lu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
17
|
Goyal S, Sanghera DK. Genetic and Non-genetic Determinants of Cardiovascular Disease in South Asians. Curr Diabetes Rev 2021; 17:e011721190373. [PMID: 33461471 PMCID: PMC10370262 DOI: 10.2174/1573399817666210118103022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 01/09/2023]
Abstract
South Asians (SAs), people from the Indian subcontinent (e.g., India, Pakistan, Bangladesh, Sri Lanka, and Nepal) have a higher prevalence of cardiovascular disease (CVD) and suffer from a greater risk of CVD-associated mortality compared to other global populations. These problems are compounded by the alterations in lifestyles due to urbanization and changing cultural, social, economic, and political environments. Current methods of CV risk prediction are based on white populations that under-estimate the CVD risk in SAs. Prospective studies are required to obtain actual CVD morbidity/mortality rates so that comparisons between predicted CVD risk can be made with actual events. Overwhelming data support a strong influence of genetic factors. Genome-Wide Association Studies (GWAS) serve as a starting point for future genetic and functional studies since the mechanisms of action by which these associated loci influence CVD is still unclear. It is difficult to predict the potential implication of these findings in clinical settings. This review provides a systematic assessment of the risk factors, genetics, and environmental causes of CV health disparity in SAs, and highlights progress made in clinical and genomics discoveries in the rapidly evolving field, which has the potential to show clinical relevance in the near future.
Collapse
Affiliation(s)
- Shiwali Goyal
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Dharambir K Sanghera
- Department of Pediatrics, Section of Genetics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
18
|
Zhou F, Ren J, Lu X, Ma S, Wu C. Gene-Environment Interaction: A Variable Selection Perspective. Methods Mol Biol 2021; 2212:191-223. [PMID: 33733358 DOI: 10.1007/978-1-0716-0947-7_13] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gene-environment interactions have important implications for elucidating the genetic basis of complex diseases beyond the joint function of multiple genetic factors and their interactions (or epistasis). In the past, G × E interactions have been mainly conducted within the framework of genetic association studies. The high dimensionality of G × E interactions, due to the complicated form of environmental effects and the presence of a large number of genetic factors including gene expressions and SNPs, has motivated the recent development of penalized variable selection methods for dissecting G × E interactions, which has been ignored in the majority of published reviews on genetic interaction studies. In this article, we first survey existing studies on both gene-environment and gene-gene interactions. Then, after a brief introduction to the variable selection methods, we review penalization and relevant variable selection methods in marginal and joint paradigms, respectively, under a variety of conceptual models. Discussions on strengths and limitations, as well as computational aspects of the variable selection methods tailored for G × E studies, have also been provided.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Statistics, Kansas State University, Manhattan, KS, USA
| | - Jie Ren
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xi Lu
- Department of Statistics, Kansas State University, Manhattan, KS, USA
| | - Shuangge Ma
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT, USA
| | - Cen Wu
- Department of Statistics, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
19
|
Sharma A, Mittal S, Aggarwal R, Chauhan MK. Diabetes and cardiovascular disease: inter-relation of risk factors and treatment. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00151-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abstract
Background
The diabetes mellitus prevalence is still advancing and increasingly becoming one of the globally most severe and expensive chronic illnesses. The strong correlation between diabetes as well as the most prominent reason for diabetes and death in diabetic patients is cardiovascular disorders. Health conditions like dyslipidemia, hypertension, obesity, and other factors of risk like the risk of cardiovascular are frequent in diabetic persons and raise the likelihood of heart attacks.
Main text
In particular, several researchers have found diabetes mellitus-related biochemical pathways that raise the likelihood of cardiovascular disorder in people with diabetes individually. This review describes diabetes-cardiovascular disorder relationships, explores potential therapeutic mechanisms, addresses existing treatment, care, and describes the directions for the future for study.
Conclusion
Thus, in individuals with diabetes, it is important to concentrate on cardiovascular threat variables to reduce the illness’s lasting cardiovascular complications. Further work to enhance knowledge of the disease state and its impact on cardiovascular function is required to boost medical treatment and cardiovascular disorders result in people with diabetes.
Collapse
|
20
|
Borse SP, Chhipa AS, Sharma V, Singh DP, Nivsarkar M. Management of Type 2 Diabetes: Current Strategies, Unfocussed Aspects, Challenges, and Alternatives. Med Princ Pract 2020; 30:109-121. [PMID: 32818934 PMCID: PMC8114074 DOI: 10.1159/000511002] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) accounts for >90% of the cases of diabetes in adults. Resistance to insulin action is the major cause that leads to chronic hyperglycemia in diabetic patients. T2DM is the consequence of activation of multiple pathways and factors involved in insulin resistance and β-cell dysfunction. Also, the etiology of T2DM involves the complex interplay between genetics and environmental factors. This interplay can be governed efficiently by lifestyle modifications to achieve better management of diabetes. The present review aims at discussing the major factors involved in the development of T2DM that remain unfocussed during the anti-diabetic therapy. The review also focuses on lifestyle modifications that are warranted for the successful management of T2DM. In addition, it attempts to explain flaws in current strategies to combat diabetes. The employability of phytoconstituents as multitargeting molecules and their potential use as effective therapeutic adjuvants to first line hypoglycemic agents to prevent side effects caused by the synthetic drugs are also discussed.
Collapse
Affiliation(s)
- Swapnil P Borse
- AYUSH-Center of Excellence, Center for Complementary and Integrative Health (CCIH), Interdisciplinary School of Health Sciences, Savitribai Phule Pune University (SPPU), Pune, India
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, India
| | - Abu Sufiyan Chhipa
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, India
- Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Vipin Sharma
- Translational Health Science and Technology Institute, Faridabad, India
| | | | - Manish Nivsarkar
- Department of Pharmacology and Toxicology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, India,
| |
Collapse
|
21
|
Coffee Consumption, Genetic Polymorphisms, and the Risk of Type 2 Diabetes Mellitus: A Pooled Analysis of Four Prospective Cohort Studies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17155379. [PMID: 32722593 PMCID: PMC7432682 DOI: 10.3390/ijerph17155379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022]
Abstract
The association between coffee consumption and the risk of type 2 diabetes may vary by genetic variants. Our study addresses the question of whether the incidence of type 2 diabetes is related to the consumption of coffee and whether this relationship is modified by polymorphisms related to type 2 diabetes. We performed a pooled analysis of four Korean prospective studies that included 71,527 participants; median follow-up periods ranged between 2 and 13 years. All participants had completed a validated food-frequency questionnaire (FFQ) at baseline. The odds ratios (ORs) and 95% confidence intervals (CIs) for type 2 diabetes were calculated using logistic regression models. The ORs were combined using a fixed or random effects model depending on the heterogeneity across the studies. Compared with 0 to <0.5 cups/day of coffee consumption, the OR for type 2 diabetes was 0.89 (95% CI: 0.80-0.98, p for trend = 0.01) for ≥3 cups/day of coffee consumption. We did not observe significant interactions by five single nucleotide polymorphisms (SNPs) related to type 2 diabetes (CDKAL1 rs7756992, CDKN2A/B rs10811661, KCNJ11 rs5215, KCNQ1 rs163184, and PEPD rs3786897) in the association between coffee and the risk of type 2 diabetes. We found that coffee consumption was inversely associated with the risk of type 2 diabetes.
Collapse
|
22
|
van den Burg EL, Schoonakker MP, van Peet PG, van den Akker-van Marle ME, Willems van Dijk K, Longo VD, Lamb HJ, Numans ME, Pijl H. Fasting in diabetes treatment (FIT) trial: study protocol for a randomised, controlled, assessor-blinded intervention trial on the effects of intermittent use of a fasting-mimicking diet in patients with type 2 diabetes. BMC Endocr Disord 2020; 20:94. [PMID: 32580710 PMCID: PMC7315472 DOI: 10.1186/s12902-020-00576-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/15/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Caloric restriction is an effective way to treat Type 2 diabetes (T2D). However, chronic and severe restriction of food intake is difficult to sustain and is known to promote slower metabolism. Intermittent and frequent fasting can exert similar metabolic effects, but may be even more challenging for most patients. A fasting-mimicking diet (FMD) is low in calories, sugars and proteins, but includes relatively high levels of plant based complex carbohydrates and healthy fats. The metabolic effects of such a diet mimic the benefits of water-only fasting. The effects of a FMD applied periodically in T2D patients are still unknown. The Fasting In diabetes Treatment (FIT) trial was designed to determine the effect of intermittent use (5 consecutive days a month during a year) of a FMD in T2D patients on metabolic parameters and T2D medication use compared to usual care. METHODS One hundred T2D patients from general practices in the Netherlands with a BMI ≥ 27 kg/m2, treated with lifestyle advice only or lifestyle advice plus metformin, will be randomised to receive the FMD plus usual care or usual care only. Primary outcomes are HbA1c and T2D medication dosage. Secondary outcomes are anthropometrics, blood pressure, plasma lipid profiles, quality of life, treatment satisfaction, metabolomics, microbiome composition, MRI data including cardiac function, fat distribution and ectopic fat storage, cost-effectiveness, and feasibility in clinical practice. DISCUSSION This study will establish whether monthly 5-day cycles of a FMD during a year improve metabolic parameters and/or reduce the need for medication in T2D. Furthermore, additional health benefits and the feasibility in clinical practice will be measured and a cost-effectiveness evaluation will be performed. TRIAL REGISTRATION The trial was registered on ClinicalTrials.gov. Identifier: NCT03811587. Registered 21th of January, 2019; retrospectively registered.
Collapse
Affiliation(s)
- Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Center (LUMC), Postzone V0-P, Postbus 9600, 2300 RC, Leiden, The Netherlands.
| | - Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Center (LUMC), Postzone V0-P, Postbus 9600, 2300 RC, Leiden, The Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Center (LUMC), Postzone V0-P, Postbus 9600, 2300 RC, Leiden, The Netherlands
| | | | - Ko Willems van Dijk
- Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Valter D Longo
- FIRC Institute of Molecular Oncology, Milan, Italy
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, USA
| | - Hildo J Lamb
- Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Center (LUMC), Postzone V0-P, Postbus 9600, 2300 RC, Leiden, The Netherlands
| | - Hanno Pijl
- Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
23
|
Popa-Wagner A, Dumitrascu DI, Capitanescu B, Petcu EB, Surugiu R, Fang WH, Dumbrava DA. Dietary habits, lifestyle factors and neurodegenerative diseases. Neural Regen Res 2020; 15:394-400. [PMID: 31571647 PMCID: PMC6921346 DOI: 10.4103/1673-5374.266045] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/20/2019] [Indexed: 12/12/2022] Open
Abstract
Worldwide stroke is increasing in parallel with modernization, changes in lifestyle, and the growing elderly population. Our review is focused on the link between diet, as part of 'modern lifestyle', and health in the context of genetic predisposition of individuals to 'unhealthy' metabolic pathway activity. It is concluded that lifestyle including high sugar diets, alcohol and tobacco addiction or high fat diets as well as ageing, brain injury, oxidative stress and neuroinflammation, negatively influence the onset, severity and duration of neurodegenerative diseases. Fortunately, there are several healthy dietary components such as polyunsaturated fatty acids and the anti-oxidants curcumin, resveratrol, blueberry polyphenols, sulphoraphane, salvionic acid as well as caloric restriction and physical activity, which may counteract ageing and associated neurodegenerative diseases via increased autophagy or increased neurogenesis in the adult brain.
Collapse
Affiliation(s)
- Aurel Popa-Wagner
- Griffith University School of Medicine, Gold Coast Campus, QLD, Australia
| | | | - Bogdan Capitanescu
- Department of Human Anatomy, Faculty of Medicine, University of Medicine and Pharmacy, Craiova, Romania
| | - Eugen Bogdan Petcu
- Griffith University School of Medicine, Gold Coast Campus, QLD, Australia
| | - Roxana Surugiu
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, Craiova, Romania
| | - Wen-Hui Fang
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Danut-Adrian Dumbrava
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, Craiova, Romania
| |
Collapse
|
24
|
Liang F, Quan Y, Wu A, Chen Y, Xu R, Zhu Y, Xiong J. Insulin-resistance and depression cohort data mining to identify nutraceutical related DNA methylation biomarker for type 2 diabetes. Genes Dis 2020; 8:669-676. [PMID: 34291138 PMCID: PMC8278533 DOI: 10.1016/j.gendis.2020.01.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 11/29/2022] Open
Abstract
Insulin-resistance (IR) is one of the most important precursors of type 2 diabetes (T2D). Recent evidence suggests an association of depression with the onset of T2D. Accumulating evidence shows that depression and T2D share common biological origins, and DNA methylation examination might reveal the link between lifestyle, disease risk, and potential therapeutic targets for T2D. Here we hypothesize that integrative mining of IR and depression cohort data will facilitate predictive biomarkers identification for T2D. We utilized a newly proposed method to extract gene-level information from probe level data on genome-wide DNA methylation array. We identified a set of genes associated with IR and depression in clinical cohorts. By overlapping the IR-related nutraceutical-gene network with depression networks, we identified a common subnetwork centered with Vitamin D Receptor (VDR) gene. Preliminary clinical validation of gene methylation set in a small cohort of T2D patients and controls was established using the Sequenome matrix-assisted laser desorption ionization-time flight mass spectrometry. A set of sites in the promoter regions of VDR showed a significant difference between T2D patients and controls. Using a logistic regression model, the optimal prediction performance of these sites was AUC = 0.902,and an odds ratio = 19.76. Thus, monitoring the methylation status of specific VDR promoter region might help stratify the high-risk individuals who could potentially benefit from vitamin D dietary supplementation. Our results highlight the link between IR and depression, and the DNA methylation analysis might facilitate the search for their shared mechanisms in the etiology of T2D.
Collapse
Affiliation(s)
- Fengji Liang
- Lab of Epigenetics and Advanced Health Technology, SPACEnter Space Science and Technology Institute, Shenzhen, Guangdong Province, 518117, PR China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, PR China
| | - Yuan Quan
- Lab of Epigenetics and Advanced Health Technology, SPACEnter Space Science and Technology Institute, Shenzhen, Guangdong Province, 518117, PR China.,School of Computer Science and Technology, Harbin Institute of Technology Shenzhen Graduate School, Shenzhen, Guangdong Province, 518055, PR China
| | - Andong Wu
- Lab of Epigenetics and Advanced Health Technology, SPACEnter Space Science and Technology Institute, Shenzhen, Guangdong Province, 518117, PR China
| | - Ying Chen
- Lab of Epigenetics and Advanced Health Technology, SPACEnter Space Science and Technology Institute, Shenzhen, Guangdong Province, 518117, PR China
| | - Ruifeng Xu
- School of Computer Science and Technology, Harbin Institute of Technology Shenzhen Graduate School, Shenzhen, Guangdong Province, 518055, PR China
| | - Yuexing Zhu
- Lab of Epigenetics and Advanced Health Technology, SPACEnter Space Science and Technology Institute, Shenzhen, Guangdong Province, 518117, PR China
| | - Jianghui Xiong
- Lab of Epigenetics and Advanced Health Technology, SPACEnter Space Science and Technology Institute, Shenzhen, Guangdong Province, 518117, PR China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, PR China
| |
Collapse
|
25
|
Abstract
Approximately 12% of U.S. adults have type 2 diabetes (T2D). Diagnosed T2D is caused by a combination of genetic and environmental factors including age and lifestyle. In adults 45 years and older, the Discordant Twin (DISCOTWIN) consortium of twin registries from Europe and Australia showed a moderate-to-high contribution of genetic factors of T2D with a pooled heritability of 72%. The purpose of this study was to investigate the contributions of genetic and environmental factors of T2D in twins 45 years and older in a U.S. twin cohort (Washington State Twin Registry, WSTR) and compare the estimates to the DISCOTWIN consortium. We also compared these estimates with twins under the age of 45. Data were obtained from 2692 monozygotic (MZ) and same-sex dizygotic (DZ) twin pairs over 45 and 4217 twin pairs under 45 who responded to the question 'Has a doctor ever diagnosed you with (type 2) diabetes?' Twin similarity was analyzed using both tetrachoric correlations and structural equation modeling. Overall, 9.4% of MZ and 14.7% of DZ twins over the age of 45 were discordant for T2D in the WSTR, compared to 5.1% of MZ and 8% of DZ twins in the DISCOTWIN consortium. Unlike the DISCOTWIN consortium in which heritability was 72%, heritability was only 52% in the WSTR. In twins under the age of 45, heritability did not contribute to the variance in T2D. In a U.S. sample of adult twins, environmental factors appear to be increasingly important in the development of T2D.
Collapse
|
26
|
Yang T, Li X, Montazeri Z, Little J, Farrington SM, Ioannidis JP, Dunlop MG, Campbell H, Timofeeva M, Theodoratou E. Gene-environment interactions and colorectal cancer risk: An umbrella review of systematic reviews and meta-analyses of observational studies. Int J Cancer 2019; 145:2315-2329. [PMID: 30536881 PMCID: PMC6767750 DOI: 10.1002/ijc.32057] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Abstract
The cause of colorectal cancer (CRC) is multifactorial, involving both genetic variants and environmental risk factors. We systematically searched the MEDLINE, EMBASE, China National Knowledge Infrastructure (CNKI) and Wanfang databases from inception to December 2016, to identify systematic reviews and meta-analyses of observational studies that investigated gene-environment (G×E) interactions in CRC risk. Then, we critically evaluated the cumulative evidence for the G×E interactions using an extension of the Human Genome Epidemiology Network's Venice criteria. Overall, 15 articles reporting systematic reviews of observational studies on 89 G×E interactions, 20 articles reporting meta-analyses of candidate gene- or single-nucleotide polymorphism-based studies on 521 G×E interactions, and 8 articles reporting 33 genome-wide G×E interaction analyses were identified. On the basis of prior and observed scores, only the interaction between rs6983267 (8q24) and aspirin use was found to have a moderate overall credibility score as well as main genetic and environmental effects. Though 5 other interactions were also found to have moderate evidence, these interaction effects were tenuous due to the lack of main genetic effects and/or environmental effects. We did not find highly convincing evidence for any interactions, but several associations were found to have moderate strength of evidence. Our conclusions are based on application of the Venice criteria which were designed to provide a conservative assessment of G×E interactions and thus do not include an evaluation of biological plausibility of an observed joint effect.
Collapse
Affiliation(s)
- Tian Yang
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
| | - Xue Li
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
| | - Zahra Montazeri
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Julian Little
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Susan M. Farrington
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| | - John P.A. Ioannidis
- Stanford Prevention Research Center, Departments of Medicine, of Health Research and Policy, and of Biomedical Data Science, Stanford University School of Medicine, and Department of StatisticsStanford University School of Humanities and SciencesStanfordCaliforniaUSA
- Meta‐Research Innovation Center at Stanford (METRICS)Stanford UniversityStanfordCaliforniaUSA
| | - Malcolm G. Dunlop
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
| | - Maria Timofeeva
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| | - Evropi Theodoratou
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
27
|
Dietrich S, Jacobs S, Zheng JS, Meidtner K, Schwingshackl L, Schulze MB. Gene-lifestyle interaction on risk of type 2 diabetes: A systematic review. Obes Rev 2019; 20:1557-1571. [PMID: 31478326 PMCID: PMC8650574 DOI: 10.1111/obr.12921] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
The pathophysiological influence of gene-lifestyle interactions on the risk to develop type 2 diabetes (T2D) is currently under intensive research. This systematic review summarizes the evidence for gene-lifestyle interactions regarding T2D incidence. MEDLINE, EMBASE, and Web of Science were systematically searched until 31 January 2019 to identify publication with (a) prospective study design; (b) T2D incidence; (c) gene-diet, gene-physical activity, and gene-weight loss intervention interaction; and (d) population who are healthy or prediabetic. Of 66 eligible publications, 28 reported significant interactions. A variety of different genetic variants and dietary factors were studied. Variants at TCF7L2 were most frequently investigated and showed interactions with fiber and whole grain on T2D incidence. Further gene-diet interactions were reported for, eg, a western dietary pattern with a T2D-GRS, fat and carbohydrate with IRS1 rs2943641, and heme iron with variants of HFE. Physical activity showed interaction with HNF1B, IRS1, PPARγ, ADRA2B, SLC2A2, and ABCC8 variants and weight loss interventions with ENPP1, PPARγ, ADIPOR2, ADRA2B, TNFα, and LIPC variants. However, most findings represent single study findings obtained in European ethnicities. Although some interactions have been reported, their conclusiveness is still low, as most findings were not yet replicated across multiple study populations.
Collapse
Affiliation(s)
- Stefan Dietrich
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam Rehbruecke, Nuthetal, Germany
| | - Simone Jacobs
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam Rehbruecke, Nuthetal, Germany
| | - Ju-Sheng Zheng
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK.,School of Life Sciences, Westlake University, Hangzhou, China
| | - Karina Meidtner
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam Rehbruecke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Lukas Schwingshackl
- Institute for Evidence in Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam Rehbruecke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Sciences, Nuthetal, Germany
| |
Collapse
|
28
|
Wang J, Ning J, Shete S. Mediation analysis in a case-control study when the mediator is a censored variable. Stat Med 2019; 38:1213-1229. [PMID: 30421436 DOI: 10.1002/sim.8028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 11/10/2022]
Abstract
Mediation analysis is an approach for assessing the direct and indirect effects of an initial variable on an outcome through a mediator. In practice, mediation models can involve a censored mediator (eg, a woman's age at menopause). The current research for mediation analysis with a censored mediator focuses on scenarios where outcomes are continuous. However, the outcomes can be binary (eg, type 2 diabetes). Another challenge when analyzing such a mediation model is to use data from a case-control study, which results in biased estimations for the initial variable-mediator association if a standard approach is directly applied. In this study, we propose an approach (denoted as MAC-CC) to analyze the mediation model with a censored mediator given data from a case-control study, based on the semiparametric accelerated failure time model along with a pseudo-likelihood function. We adapted the measures for assessing the indirect and direct effects using counterfactual definitions. We conducted simulation studies to investigate the performance of MAC-CC and compared it to those of the naïve approach and the complete-case approach. MAC-CC accurately estimates the coefficients of different paths, the indirect effects, and the proportions of the total effects mediated. We applied the proposed and existing approaches to the mediation study of genetic variants, a woman's age at menopause, and type 2 diabetes based on a case-control study of type 2 diabetes. Our results indicate that there is no mediating effect from the age at menopause on the association between the genetic variants and type 2 diabetes.
Collapse
Affiliation(s)
- Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
29
|
Interactions between plasma copper concentrations and SOD1 gene polymorphism for impaired glucose regulation and type 2 diabetes. Redox Biol 2019; 24:101172. [PMID: 30909159 PMCID: PMC6434161 DOI: 10.1016/j.redox.2019.101172] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 01/11/2023] Open
Abstract
Aims To examine the associations of plasma copper concentrations and superoxide dismutase 1 (SOD1) polymorphisms as well as their gene-environment interaction with newly diagnosed impaired glucose regulation (IGR) and type 2 diabetes (T2D). Methods We performed a large case-control study in 2520 Chinese Han subjects: 1004 newly diagnosed T2D patients, 512 newly diagnosed IGR patients and 1004 individuals with normal glucose tolerance. Results After multivariable adjustment, the ORs (95% CIs) of T2D across tertiles of plasma copper were 1.00 (reference), 1.85 (95% CI: 1.39, 2.45), and 4.21 (95% CI: 3.20, 5.55) (P-trend < 0.001). Each SD increment of ln-transformed plasma copper was associated with 104% higher odds (OR 2.04, 95%CI 1.82–2.28) increment in ORs of T2D. Meanwhile, compared with the GG genotype of rs2070424, the OR of T2D associated with AG and AA genotypes were 1.44 (95% CI 1.15–1.81) and 1.74 (95% CI 1.33–2.28), respectively. In addition, the positive association between plasma copper and T2D was modified by rs2070424 genotypes. The adjusted ORs and 95% CIs of T2D per SD increment of ln-transformed plasma copper were 2.40 (1.93–2.99), 1.85 (1.59–2.16) and 1.76 (1.44–2.15) in rs2070424 GG, AG and GG carriers respectively (P for interaction < 0.05). Similar interactions were also found for IGR and IGR&T2D. When the joint effects were examined, individuals with rs2070424 AA genotype and the highest tertile of plasma copper concentration had a much higher risk of IGR&T2D (OR 5.34, 95% CI 3.48–8.21) than those with rs2070424 GG genotype and the lowest tertile of plasma copper concentrations. Conclusions Plasma copper concentrations are positively and significantly associated with IGR as well as T2D, and these associations may be modified by SOD1 polymorphism. Further studies are warranted to elucidate the potential mechanisms. Plasma copper concentrations are positively and significantly associated with IGR as well as T2D. Compared with the GG genotype of rs2070424, the risk of T2D associated with AG and AA genotypes were higher. The associations between copper and T2D as well as IGR may be modified by SOD1 rs2070424 polymorphism. Evaluating the interaction of copper and gene polymorphisms may shed etiologic insight into the copper-diabetes relation.
Collapse
|
30
|
Abstract
Diabetes develops due to deficient functional β cell mass, insulin resistance, or both. Yet, various challenges in understanding the mechanisms underlying diabetes development in vivo remain to be overcome owing to the lack of appropriate intravital imaging technologies. To meet these challenges, we have exploited the anterior chamber of the eye (ACE) as a novel imaging site to understand diabetes basics and clinics in vivo. We have developed a technology platform transplanting pancreatic islets into the ACE where they later on can be imaged non-invasively for long time. It turns out that the ACE serves as an optimal imaging site and provides implanted islets with an oxygen-rich milieu and an immune-privileged niche where they undergo optimal engraftment, rich vascularization and dense innervation, preserve organotypic features and live with satisfactory viability and functionality. The ACE technology has led to a series of significant observations. It enables in vivo microscopy of islet cytoarchitecture, function and viability in the physiological context and intravital imaging of a variety of pathological events such as autoimmune insulitis, defects in β cell function and mass and insulin resistance during diabetes development in a real-time manner. Furthermore, application of the ACE technology in humanized mice and non-human primates verifies translational and clinical values of the technology. In this article, we describe the ACE technology in detail, review accumulated knowledge gained by means of the ACE technology and delineate prospective avenues for the ACE technology.
Collapse
|
31
|
Ahmad S, Fatima SS, Rukh G, Smith CE. Gene Lifestyle Interactions With Relation to Obesity, Cardiometabolic, and Cardiovascular Traits Among South Asians. Front Endocrinol (Lausanne) 2019; 10:221. [PMID: 31024458 PMCID: PMC6465946 DOI: 10.3389/fendo.2019.00221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 03/20/2019] [Indexed: 01/05/2023] Open
Abstract
The rapid rise of obesity, type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD) during the last few decades among South Asians has been largely attributed to a major shift in lifestyles including physical inactivity, unhealthy dietary patterns, and an overall pattern of sedentary lifestyle. Genetic predisposition to these cardiometabolic risk factors may have interacted with these obesogenic environments in determining the higher cardiometabolic disease prevalence. Based on the premise that gene-environment interactions cause obesity and cardiometabolic diseases, we systematically searched the literature and considered the knowledge gaps that future studies might fulfill. We identified only seven published studies that focused specifically on gene-environment interactions for cardiometabolic traits in South Asians, most of which were limited by relatively small sample and lack of replication. Some studies reported that the differences in metabolic response to higher physical activity and low caloric diet might be modified by genetic risk related to these cardiometabolic traits. Although studies on gene lifestyle interactions in cardiometabolic traits report significant interactions, future studies must focus on more precise assessment of lifestyle factors, investigation of a larger set of genetic variants and the application of powerful statistical methods to facilitate translatable approaches. Future studies should also be integrated with findings both using mechanistic studies through laboratory settings and randomized clinical trials for clinical outcomes.
Collapse
Affiliation(s)
- Shafqat Ahmad
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
- Preventive Medicine Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, United States
- *Correspondence: Shafqat Ahmad
| | - Syeda Sadia Fatima
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Gull Rukh
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Caren E. Smith
- Nutrition and Genomics Laboratory, Jean Mayer U. S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| |
Collapse
|
32
|
Witka BZ, Oktaviani DJ, Marcellino M, Barliana MI, Abdulah R. Type 2 Diabetes-Associated Genetic Polymorphisms as Potential Disease Predictors. Diabetes Metab Syndr Obes 2019; 12:2689-2706. [PMID: 31908510 PMCID: PMC6927489 DOI: 10.2147/dmso.s230061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetes is a major cause of mortality worldwide. There are several types of diabetes, with type 2 diabetes mellitus (T2DM) being the most common. Many factors, including environmental and genetic factors, are involved in the etiology of the disease. Numerous studies have reported the role of genetic polymorphisms in the initiation and development of T2DM. While genome-wide association studies have identified around more than 200 susceptibility loci, it remains unclear whether these loci are correlated with the pathophysiology of the disease. The present review aimed to elucidate the potential genetic mechanisms underlying T2DM. We found that some genetic polymorphisms were related to T2DM, either in the form of single-nucleotide polymorphisms or direct amino acid changes in proteins. These polymorphisms are potential predictors for the management of T2DM.
Collapse
Affiliation(s)
- Beska Z Witka
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Dede J Oktaviani
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Marcellino Marcellino
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Melisa I Barliana
- Departement of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
- Correspondence: Melisa I Barliana Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM. 21, Jatinangor45363, Indonesia Email
| | - Rizky Abdulah
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
33
|
Zhou T, Kim TW, Chong CN, Tan L, Amin S, Sadat Badieyan Z, Mukherjee S, Ghazizadeh Z, Zeng H, Guo M, Crespo M, Zhang T, Kenyon R, Robinson CL, Apostolou E, Wang H, Xiang JZ, Evans T, Studer L, Chen S. A hPSC-based platform to discover gene-environment interactions that impact human β-cell and dopamine neuron survival. Nat Commun 2018; 9:4815. [PMID: 30446643 PMCID: PMC6240096 DOI: 10.1038/s41467-018-07201-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 10/23/2018] [Indexed: 01/12/2023] Open
Abstract
Common disorders, including diabetes and Parkinson’s disease, are caused by a combination of environmental factors and genetic susceptibility. However, defining the mechanisms underlying gene-environment interactions has been challenging due to the lack of a suitable experimental platform. Using pancreatic β-like cells derived from human pluripotent stem cells (hPSCs), we discovered that a commonly used pesticide, propargite, induces pancreatic β-cell death, a pathological hallmark of diabetes. Screening a panel of diverse hPSC-derived cell types we extended this observation to a similar susceptibility in midbrain dopamine neurons, a cell type affected in Parkinson’s disease. We assessed gene-environment interactions using isogenic hPSC lines for genetic variants associated with diabetes and Parkinson’s disease. We found GSTT1−/− pancreatic β-like cells and dopamine neurons were both hypersensitive to propargite-induced cell death. Our study identifies an environmental chemical that contributes to human β-cell and dopamine neuron loss and validates a novel hPSC-based platform for determining gene-environment interactions. Diseases such as diabetes and Parkinson's manifest based on interactions between genes and environment. Here, the authors find among a panel of cell types that propargite, a common pesticide, induces pancreatic β-cell and dopamine neuron death and that loss of the gene GSTT1 confers hypersensitivity.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Tae Wan Kim
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, 10065, USA.,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, 10065, USA
| | - Chi Nok Chong
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Lei Tan
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA.,School of Public health, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200000, China
| | - Sadaf Amin
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Zohreh Sadat Badieyan
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Suranjit Mukherjee
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Zaniar Ghazizadeh
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Hui Zeng
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Min Guo
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Miguel Crespo
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Reyn Kenyon
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Christopher L Robinson
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Effie Apostolou
- Department of Medicine, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Hui Wang
- School of Public health, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200000, China
| | - Jenny Zhaoying Xiang
- Genomic Resource Core Facility, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, 10065, USA. .,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, 10065, USA.
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA. .,Department of Biochemistry, Weill Cornell Medical College, 1300 York Ave, New York, 10065, NY, USA.
| |
Collapse
|
34
|
Abstract
Molecular clocks are important for the circadian regulation of ß-cell function. DBP/E4BP4 plays central roles among clock-related genes in the metabolic regulation.
Collapse
Affiliation(s)
- Akihiko Taguchi
- Department of Endocrinology, Metabolism, Hematological Science and TherapeuticsGraduate School of MedicineYamaguchi UniversityUbeYamaguchiJapan
| | - Yasuharu Ohta
- Department of Endocrinology, Metabolism, Hematological Science and TherapeuticsGraduate School of MedicineYamaguchi UniversityUbeYamaguchiJapan
- Department of Diabetes ResearchSchool of MedicineYamaguchi UniversityUbeYamaguchiJapan
| | - Yukio Tanizawa
- Department of Endocrinology, Metabolism, Hematological Science and TherapeuticsGraduate School of MedicineYamaguchi UniversityUbeYamaguchiJapan
- The Research Institute for Time Studies (RITS)Yamaguchi UniversityYamaguchiJapan
| |
Collapse
|
35
|
Pan A, Lin X, Hemler E, Hu FB. Diet and Cardiovascular Disease: Advances and Challenges in Population-Based Studies. Cell Metab 2018; 27:489-496. [PMID: 29514062 PMCID: PMC5844273 DOI: 10.1016/j.cmet.2018.02.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/16/2018] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
In this Minireview, we provide an epidemiologist's perspective on the debate and recent advances in determining the relationship between diet and cardiovascular health. We conclude that, in order to reduce the global burden of cardiovascular disease, there should be a greater emphasis on improving overall diet quality and food sources of macronutrients, such as dietary fats and carbohydrates. In addition, building a strong evidence base through high-quality intervention and observational studies is crucial for effective policy changes, which can greatly improve the food environment and population health.
Collapse
Affiliation(s)
- An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China.
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Elena Hemler
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
36
|
He L, Zhbannikov I, Arbeev KG, Yashin AI, Kulminski AM. A genetic stochastic process model for genome-wide joint analysis of biomarker dynamics and disease susceptibility with longitudinal data. Genet Epidemiol 2017; 41:620-635. [PMID: 28636232 PMCID: PMC5643257 DOI: 10.1002/gepi.22058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/06/2017] [Accepted: 05/17/2017] [Indexed: 12/31/2022]
Abstract
Unraveling the underlying biological mechanisms or pathways behind the effects of genetic variations on complex diseases remains one of the major challenges in the post-GWAS (where GWAS is genome-wide association study) era. To further explore the relationship between genetic variations, biomarkers, and diseases for elucidating underlying pathological mechanism, a huge effort has been placed on examining pleiotropic and gene-environmental interaction effects. We propose a novel genetic stochastic process model (GSPM) that can be applied to GWAS and jointly investigate the genetic effects on longitudinally measured biomarkers and risks of diseases. This model is characterized by more profound biological interpretation and takes into account the dynamics of biomarkers during follow-up when investigating the hazards of a disease. We illustrate the rationale and evaluate the performance of the proposed model through two GWAS. One is to detect single nucleotide polymorphisms (SNPs) having interaction effects on type 2 diabetes (T2D) with body mass index (BMI) and the other is to detect SNPs affecting the optimal BMI level for protecting from T2D. We identified multiple SNPs that showed interaction effects with BMI on T2D, including a novel SNP rs11757677 in the CDKAL1 gene (P = 5.77 × 10-7 ). We also found a SNP rs1551133 located on 2q14.2 that reversed the effect of BMI on T2D (P = 6.70 × 10-7 ). In conclusion, the proposed GSPM provides a promising and useful tool in GWAS of longitudinal data for interrogating pleiotropic and interaction effects to gain more insights into the relationship between genes, quantitative biomarkers, and risks of complex diseases.
Collapse
Affiliation(s)
- Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708
| | - Ilya Zhbannikov
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708
| | - Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708
| |
Collapse
|
37
|
Affiliation(s)
- Lina Radzeviciene
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rytas Ostrauskas
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
38
|
Li SX, Imamura F, Ye Z, Schulze MB, Zheng J, Ardanaz E, Arriola L, Boeing H, Dow C, Fagherazzi G, Franks PW, Agudo A, Grioni S, Kaaks R, Katzke VA, Key TJ, Khaw KT, Mancini FR, Navarro C, Nilsson PM, Onland-Moret NC, Overvad K, Palli D, Panico S, Quirós JR, Rolandsson O, Sacerdote C, Sánchez MJ, Slimani N, Sluijs I, Spijkerman AM, Tjonneland A, Tumino R, Sharp SJ, Riboli E, Langenberg C, Scott RA, Forouhi NG, Wareham NJ. Interaction between genes and macronutrient intake on the risk of developing type 2 diabetes: systematic review and findings from European Prospective Investigation into Cancer (EPIC)-InterAct. Am J Clin Nutr 2017; 106:263-275. [PMID: 28592605 PMCID: PMC5486199 DOI: 10.3945/ajcn.116.150094] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 04/26/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Gene-diet interactions have been reported to contribute to the development of type 2 diabetes (T2D). However, to our knowledge, few examples have been consistently replicated to date.Objective: We aimed to identify existing evidence for gene-macronutrient interactions and T2D and to examine the reported interactions in a large-scale study.Design: We systematically reviewed studies reporting gene-macronutrient interactions and T2D. We searched the MEDLINE, Human Genome Epidemiology Network, and WHO International Clinical Trials Registry Platform electronic databases to identify studies published up to October 2015. Eligibility criteria included assessment of macronutrient quantity (e.g., total carbohydrate) or indicators of quality (e.g., dietary fiber) by use of self-report or objective biomarkers of intake. Interactions identified in the review were subsequently examined in the EPIC (European Prospective Investigation into Cancer)-InterAct case-cohort study (n = 21,148, with 9403 T2D cases; 8 European countries). Prentice-weighted Cox regression was used to estimate country-specific HRs, 95% CIs, and P-interaction values, which were then pooled by random-effects meta-analysis. A primary model was fitted by using the same covariates as reported in the published studies, and a second model adjusted for additional covariates and estimated the effects of isocaloric macronutrient substitution.Results: Thirteen observational studies met the eligibility criteria (n < 1700 cases). Eight unique interactions were reported to be significant between macronutrients [carbohydrate, fat, saturated fat, dietary fiber, and glycemic load derived from self-report of dietary intake and circulating n-3 (ω-3) polyunsaturated fatty acids] and genetic variants in or near transcription factor 7-like 2 (TCF7L2), gastric inhibitory polypeptide receptor (GIPR), caveolin 2 (CAV2), and peptidase D (PEPD) (P-interaction < 0.05). We found no evidence of interaction when we tried to replicate previously reported interactions. In addition, no interactions were detected in models with additional covariates.Conclusions: Eight gene-macronutrient interactions were identified for the risk of T2D from the literature. These interactions were not replicated in the EPIC-InterAct study, which mirrored the analyses undertaken in the original reports. Our findings highlight the importance of independent replication of reported interactions.
Collapse
Affiliation(s)
- Sherly X Li
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Fumiaki Imamura
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Zheng Ye
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Jusheng Zheng
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Eva Ardanaz
- Navarre Public Health Institute (ISPN), Pamplona, Spain
- Center for Biomedical Research in Network Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Larraitz Arriola
- Center for Biomedical Research in Network Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Public Health Division of Gipuzkoa, San Sebastian, Spain
- Bio-Donostia Institute, Basque Government, San Sebastian, Spain
| | - Heiner Boeing
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Courtney Dow
- French National Institute of Health and Medical Research (INSERM) U1018, Institut Gustave Roussy, Center for Research in Epidemiology and Population Health (CESP), Villejuif, France
- University Paris-Saclay, University Paris-Sud, Villejuif, France
| | - Guy Fagherazzi
- French National Institute of Health and Medical Research (INSERM) U1018, Institut Gustave Roussy, Center for Research in Epidemiology and Population Health (CESP), Villejuif, France
- University Paris-Saclay, University Paris-Sud, Villejuif, France
| | - Paul W Franks
- Lund University, Malmö, Sweden
- Umeå University, Umeå, Sweden
| | - Antonio Agudo
- Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Sara Grioni
- Epidemiology and Prevention Unit, Milan, Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Kay Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Francesca R Mancini
- French National Institute of Health and Medical Research (INSERM) U1018, Institut Gustave Roussy, Center for Research in Epidemiology and Population Health (CESP), Villejuif, France
- University Paris-Saclay, University Paris-Sud, Villejuif, France
| | - Carmen Navarro
- Center for Biomedical Research in Network Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, Biomedical Research Institute of Murcia (IMIB)-Arrixaca, Murcia, Spain
- Unit of Preventive Medicine and Public Health, School of Medicine, University of Murcia, Murcia, Spain
| | | | | | - Kim Overvad
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
- Aalborg University Hospital, Aalborg, Denmark
| | - Domenico Palli
- Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Salvatore Panico
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | | | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, City of Health and Science Hospital, University of Turin, Torino, Italy
- Center for Cancer Prevention (CPO), Torino, Italy
- Human Genetics Foundation (HuGeF), Torino, Italy
| | - María-José Sánchez
- Center for Biomedical Research in Network Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Andalusian School of Public Health, Granada, Spain
- Biosanitary Research Institute of Granada (Granada.ibs), Granada, Spain
| | - Nadia Slimani
- International Agency for Research on Cancer, Lyon, France
| | - Ivonne Sluijs
- University Medical Center Utrecht, Utrecht, Netherlands
| | | | | | - Rosario Tumino
- Provincial Healthcare Company (ASP) Ragusa, Vittoria, Italy; and
| | - Stephen J Sharp
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Elio Riboli
- School of Public Health, Imperial College London, London, United Kingdom
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Robert A Scott
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nita G Forouhi
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom;
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
39
|
Gene-Diet Interactions in Type 2 Diabetes: The Chicken and Egg Debate. Int J Mol Sci 2017; 18:ijms18061188. [PMID: 28574454 PMCID: PMC5486011 DOI: 10.3390/ijms18061188] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/23/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Consistent evidence from both experimental and human studies indicates that Type 2 diabetes mellitus (T2DM) is a complex disease resulting from the interaction of genetic, epigenetic, environmental, and lifestyle factors. Nutrients and dietary patterns are important environmental factors to consider in the prevention, development and treatment of this disease. Nutritional genomics focuses on the interaction between bioactive food components and the genome and includes studies of nutrigenetics, nutrigenomics and epigenetic modifications caused by nutrients. There is evidence supporting the existence of nutrient-gene and T2DM interactions coming from animal studies and family-based intervention studies. Moreover, many case-control, cohort, cross-sectional cohort studies and clinical trials have identified relationships between individual genetic load, diet and T2DM. Some of these studies were on a large scale. In addition, studies with animal models and human observational studies, in different countries over periods of time, support a causative relationship between adverse nutritional conditions during in utero development, persistent epigenetic changes and T2DM. This review provides comprehensive information on the current state of nutrient-gene interactions and their role in T2DM pathogenesis, the relationship between individual genetic load and diet, and the importance of epigenetic factors in influencing gene expression and defining the individual risk of T2DM.
Collapse
|
40
|
Zheng JS, Li K, Huang T, Chen Y, Xie H, Xu D, Sun J, Li D. Genetic Risk Score of Nine Type 2 Diabetes Risk Variants that Interact with Erythrocyte Phospholipid Alpha-Linolenic Acid for Type 2 Diabetes in Chinese Hans: A Case-Control Study. Nutrients 2017; 9:nu9040376. [PMID: 28398239 PMCID: PMC5409715 DOI: 10.3390/nu9040376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/10/2017] [Accepted: 03/28/2017] [Indexed: 11/16/2022] Open
Abstract
Modulation of n-3 fatty acids on genetic susceptibility to type 2 diabetes (T2D) is still not clear. In a case-control study of 622 Chinese T2D patients and 293 healthy controls, a genetic risk score (GRS) was created based on nine T2D genetic variants. Logistic regression was used to examine the interaction of the GRS with erythrocyte phospholipid n-3 fatty acids for T2D risk. Every 1-unit (corresponding to 1 risk allele) increase in GRS was associated with 12% (Odds ratio (OR): 1.12; 95% confidence intervals (CI): 1.04–1.20) higher risk of T2D. Compared with the lowest quartile, participants had lower T2D risk in the 2nd (OR: 0.55; 95% CI: 0.36–0.84), 3rd (OR: 0.58; 95% CI: 0.38–0.88) and 4th (OR: 0.67; 95% CI: 0.44–1.03) quartile of alpha-linolenic acid (ALA) levels. Significant interaction (p-interaction = 0.029) of GRS with ALA for T2D risk was observed. Higher ALA levels were associated with lower T2D risk only among participants within the lowest GRS tertile, with ORs 0.51 (95% CI: 0.26–1.03), 0.44 (95% CI: 0.22–0.89) and 0.49 (95% CI: 0.25–0.96) for the 2nd, 3rd and 4th ALA quartile, compared with the 1st. This study suggests that higher erythrocyte ALA levels are inversely associated with T2D risk only among participants with low T2D genetic risk, with high genetic risk abolishing the ALA-T2D association.
Collapse
Affiliation(s)
- Ju-Sheng Zheng
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, China.
| | - Kelei Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
| | - Tao Huang
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore.
| | - Yanqiu Chen
- Clinical Nutrition Center, Huadong Hospital, Fudan University, Shanghai 200040, China.
| | - Hua Xie
- Clinical Nutrition Center, Huadong Hospital, Fudan University, Shanghai 200040, China.
| | - Danfeng Xu
- Clinical Nutrition Center, Huadong Hospital, Fudan University, Shanghai 200040, China.
| | - Jianqin Sun
- Clinical Nutrition Center, Huadong Hospital, Fudan University, Shanghai 200040, China.
| | - Duo Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
41
|
Clock Gene Dysregulation Induced by Chronic ER Stress Disrupts β-cell Function. EBioMedicine 2017; 18:146-156. [PMID: 28389215 PMCID: PMC5405175 DOI: 10.1016/j.ebiom.2017.03.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/09/2017] [Accepted: 03/27/2017] [Indexed: 12/26/2022] Open
Abstract
In Wfs1-/-Ay/a islets, in association with endoplasmic reticulum (ER) stress, D-site-binding protein (Dbp) expression decreased and Nuclear Factor IL-3 (Nfil3)/E4 Promoter-binding protein 4 (E4bp4) expression increased, leading to reduced DBP transcriptional activity. Similar alterations were observed with chemically-induced ER stress. Transgenic mice expressing E4BP4 under the control of the mouse insulin I gene promoter (MIP), in which E4BP4 in β-cells is expected to compete with DBP for D-box, displayed remarkable glucose intolerance with severely impaired insulin secretion. Basal ATP/ADP ratios in MIP-E4BP4 islets were elevated without the circadian oscillations observed in wild-type islets. Neither elevation of the ATP/ADP ratio nor an intracellular Ca2+ response was observed after glucose stimulation. RNA expressions of genes involved in insulin secretion gradually increase in wild-type islets early in the feeding period. In MIP-E4BP4 islets, however, these increases were not observed. Thus, molecular clock output DBP transcriptional activity, susceptible to ER stress, plays pivotal roles in β-cell priming for insulin release by regulating β-cell metabolism and gene expressions. Because ER stress is also involved in the β-cell failure in more common Type-2 diabetes, understanding the currently identified ER stress-associated mechanisms warrants novel therapeutic and preventive strategies for both rare form and common diabetes.
Collapse
|
42
|
Gong L, Li R, Ren W, Wang Z, Wang Z, Yang M, Zhang S. The FOXO1 Gene-Obesity Interaction Increases the Risk of Type 2 Diabetes Mellitus in a Chinese Han Population. J Korean Med Sci 2017; 32:264-271. [PMID: 28049237 PMCID: PMC5219992 DOI: 10.3346/jkms.2017.32.2.264] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Here, we aimed to study the effect of the forkhead box O1-insulin receptor substrate 2 (FOXO1-IRS2) gene interaction and the FOXO1 and IRS2 genes-environment interaction for the risk of type 2 diabetes mellitus (T2DM) in a Chinese Han population. We genotyped 7 polymorphism sites of FOXO1 gene and IRS2 gene in 780 unrelated Chinese Han people (474 cases of T2DM, 306 cases of healthy control). The risk of T2DM in individuals with AA genotype for rs7986407 and CC genotype for rs4581585 in FOXO1 gene was 2.092 and 2.57 times higher than that with GG genotype (odds ratio [OR] = 2.092; 95% confidence interval [CI] = 1.178-3.731; P = 0.011) and TT genotype (OR = 2.571; 95% CI = 1.404-4.695; P = 0.002), respectively. The risk of T2DM in individuals with GG genotype for Gly1057Asp in IRS2 gene was 1.42 times higher than that with AA genotype (OR = 1.422; 95% CI = 1.037-1.949; P = 0.029). The other 4 single nucleotide polymorphisms (SNPs) had no significant association with T2DM (P > 0.05). Multifactor dimensionality reduction (MDR) analysis showed that the interaction between SNPs rs7986407 and rs4325426 in FOXO1 gene and waist was the best model confirmed by interaction analysis, closely associating with T2DM. There was an increased risk for T2DM in the case of non-obesity with genotype combined AA/CC, AA/AC or AG/AA for rs7986407 and rs4325426, and obesity with genotype AA for rs7986407 or AA for rs4325426 (OR = 3.976; 95% CI = 1.156-13.675; P value from sign test [P(sign)] = 0.025; P value from permutation test [P(perm)] = 0.000-0.001). Together, this study indicates an association of FOXO1 and IRS2 gene polymorphisms with T2DM in Chinese Han population, supporting FOXO1-obesity interaction as a key factor for the risk of T2DM.
Collapse
Affiliation(s)
- Lilin Gong
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Rong Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Ren
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zengchan Wang
- Laboratory for Disease and Gene, Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, Department of Public Health, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihong Wang
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Maosheng Yang
- Laboratory for Disease and Gene, Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, Department of Public Health, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory of Disorders Genes and Department of Pharmacology, Jishou University, Jishou, China
| | - Suhua Zhang
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
43
|
Wang J, Shete S. Estimation of indirect effect when the mediator is a censored variable. Stat Methods Med Res 2017; 27:3010-3025. [PMID: 28132585 DOI: 10.1177/0962280217690414] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A mediation model explores the direct and indirect effects of an initial variable ( X) on an outcome variable ( Y) by including a mediator ( M). In many realistic scenarios, investigators observe censored data instead of the complete data. Current research in mediation analysis for censored data focuses mainly on censored outcomes, but not censored mediators. In this study, we proposed a strategy based on the accelerated failure time model and a multiple imputation approach. We adapted a measure of the indirect effect for the mediation model with a censored mediator, which can assess the indirect effect at both the group and individual levels. Based on simulation, we established the bias in the estimations of different paths (i.e. the effects of X on M [ a], of M on Y [ b] and of X on Y given mediator M [ c']) and indirect effects when analyzing the data using the existing approaches, including a naïve approach implemented in software such as Mplus, complete-case analysis, and the Tobit mediation model. We conducted simulation studies to investigate the performance of the proposed strategy compared to that of the existing approaches. The proposed strategy accurately estimates the coefficients of different paths, indirect effects and percentages of the total effects mediated. We applied these mediation approaches to the study of SNPs, age at menopause and fasting glucose levels. Our results indicate that there is no indirect effect of association between SNPs and fasting glucose level that is mediated through the age at menopause.
Collapse
Affiliation(s)
- Jian Wang
- 1 Department of Biostatistics, The University of Texas MD Anderson Cancer Center, USA
| | - Sanjay Shete
- 1 Department of Biostatistics, The University of Texas MD Anderson Cancer Center, USA.,2 Department of Epidemiology, The University of Texas MD Anderson Cancer Center, USA
| |
Collapse
|
44
|
Farooq R, Amin S, Hayat Bhat M, Malik R, Wani HA, Majid S. Type 2 diabetes and metabolic syndrome - adipokine levels and effect of drugs. Gynecol Endocrinol 2017; 33:75-78. [PMID: 27705028 DOI: 10.1080/09513590.2016.1207165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a consequence of complex interactions among multiple genetic variants and environmental risk factors. This complex disorder is also characterized by changes in various adipokines. In this study, our objective was to estimate the levels of adiponectin, leptin, and resistin (ALR) in T2DM patients, besides studying the effect of various drugs on their levels. Study participants included 400 diabetic and 300 normal patients from the Department of Endocrinology and Department of Biochemistry, Govt Medical College Srinagar. Subjects were categorized under various groups, i.e., Group 1 (metformin treated) and Group 2 (glimepiride treated), and cases were also categorized as obese with T2DM (Group A), obese without T2DM (Group B), and T2DM only (Group C). The serum ALR levels were estimated by ELISA (Alere), and biochemical parameters were also evaluated before and after treatment. Adiponectin levels were found to be significantly lower in T2DM cases as compared to controls (12 ± 5.5 versus 22.5 ± 7.9 μg/ml), while leptin and resistin levels were found to be significantly higher than controls (14.3 ± 7.4 versus 7.36 ± 3.73 ng/ml) (13.4 ± 1.56 versus 7.236 ± 2.129 pg/ml). Taking the effect of drugs into consideration, the effect on adiponectin and resistin levels was found to be highly significant in Group 2 before and after treatment (11 ± 5 versus 19.2 ± 4.5 μg/ml) (13.6 ± 2.5 versus 7.3 ± 2.9 pg/ml), while more effect was observed in leptin among Group 1 (metformin)-treated cases (27 ± 15 ng/ml versus 15 ± 15 ng/ml). Further the adiponectin levels were found to be significantly lower in Group B, while leptin and resistin levels were found to be significantly higher among obese cases when compared to T2DM cases only. Glimepiride also shows more effect on FBG, HbA1c% levels, while metformin shows more effect on Lipid profile levels. From the study, it can be concluded that ALR levels are affected by use of antidiabetic drugs among which glimepiride shows more effect on adiponectin and resistin levels, while leptin gets affected more by metformin. It can also be proposed that ALR levels are not affected by diabetes only, suggesting that their alterations in T2DM may be due to obesity as we observed more ALR changes in obese cases when compared to T2DM cases, and so there might be an important link between adiposity and insulin resistance.
Collapse
Affiliation(s)
- Rabia Farooq
- a Department of Biochemistry , Govt Medical College , Srinagar , India
| | - Shajrul Amin
- b Department of Biochemistry , University of Kashmir , Srinagar , India , and
| | - M Hayat Bhat
- c Department of Medicine , Govt Medical College , Srinagar , India
| | - Rawoof Malik
- a Department of Biochemistry , Govt Medical College , Srinagar , India
| | - Hilal Ahmad Wani
- a Department of Biochemistry , Govt Medical College , Srinagar , India
| | - Sabhiya Majid
- a Department of Biochemistry , Govt Medical College , Srinagar , India
| |
Collapse
|
45
|
Génin E, Clerget-Darpoux F. Revisiting the Polygenic Additive Liability Model through the Example of Diabetes Mellitus. Hum Hered 2016; 80:171-7. [DOI: 10.1159/000447683] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
46
|
Eze IC, Imboden M, Kumar A, von Eckardstein A, Stolz D, Gerbase MW, Künzli N, Pons M, Kronenberg F, Schindler C, Probst-Hensch N. Air pollution and diabetes association: Modification by type 2 diabetes genetic risk score. ENVIRONMENT INTERNATIONAL 2016; 94:263-271. [PMID: 27281273 DOI: 10.1016/j.envint.2016.04.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/11/2016] [Accepted: 04/22/2016] [Indexed: 05/26/2023]
Abstract
Exposure to ambient air pollution (AP) exposure has been linked to type 2 diabetes (T2D) risk. Evidence on the impact of T2D genetic variants on AP susceptibility is lacking. Compared to single variants, joint genetic variants contribute substantially to disease risk. We investigated the modification of AP and diabetes association by a genetic risk score (GRS) covering 63 T2D genes in 1524 first follow-up participants of the Swiss cohort study on air pollution and lung and heart diseases in adults. Genome-wide data and covariates were available from a nested asthma case-control study design. AP was estimated as 10-year mean residential particulate matter <10μm (PM10). We computed count-GRS and weighted-GRS, and applied PM10 interaction terms in mixed logistic regressions, on odds of diabetes. Analyses were stratified by pathways of diabetes pathology and by asthma status. Diabetes prevalence was 4.6% and mean exposure to PM10 was 22μg/m(3). Odds of diabetes increased by 8% (95% confidence interval: 2, 14%) per T2D risk allele and by 35% (-8, 97%) per 10μg/m(3) exposure to PM10. We observed a positive interaction between PM10 and count-GRS on diabetes [ORinteraction=1.10 (1.01, 1.20)], associations being strongest among participants at the highest quartile of count-GRS [OR: 1.97 (1.00, 3.87)]. Stronger interactions were observed with variants of the GRS involved in insulin resistance [(ORinteraction=1.22 (1.00, 1.50)] than with variants related to beta-cell function. Interactions with count-GRS were stronger among asthma cases. We observed similar results with weighted-GRS. Five single variants near GRB14, UBE2E2, PTPRD, VPS26A and KCNQ1 showed nominally significant interactions with PM10 (P<0.05). Our results suggest that genetic risk for T2D may modify susceptibility to air pollution through alterations in insulin sensitivity. These results need confirmation in diabetes cohort consortia.
Collapse
Affiliation(s)
- Ikenna C Eze
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Ashish Kumar
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Karolinska Institutet, Stockholm, Sweden
| | | | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, Basel, Switzerland
| | | | - Nino Künzli
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Marco Pons
- Department of Internal Medicine, Regional Hospital of Lugano, Lugano, Switzerland
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Nicole Probst-Hensch
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
47
|
Ley SH, Ardisson Korat AV, Sun Q, Tobias DK, Zhang C, Qi L, Willett WC, Manson JE, Hu FB. Contribution of the Nurses' Health Studies to Uncovering Risk Factors for Type 2 Diabetes: Diet, Lifestyle, Biomarkers, and Genetics. Am J Public Health 2016; 106:1624-30. [PMID: 27459454 DOI: 10.2105/ajph.2016.303314] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To review the contribution of the Nurses' Health Study (NHS) and the NHS II to addressing hypotheses regarding risk factors for type 2 diabetes. METHODS We carried out a narrative review of 1976 to 2016 NHS and NHS II publications. RESULTS The NHS and NHS II have uncovered important roles in type 2 diabetes for individual nutrients, foods, dietary patterns, and physical activity independent of excess body weight. Up to 90% of type 2 diabetes cases are potentially preventable if individuals follow a healthy diet and lifestyle. The NHS investigations have also identified novel biomarkers for diabetes, including adipokines, inflammatory cytokines, nutrition metabolites, and environmental pollutants, offering new insights into the pathophysiology of the disease. Global collaborative efforts have uncovered many common genetic variants associated with type 2 diabetes and improved our understanding of gene-environment interactions. Continued efforts to identify epigenetic, metagenomic, and metabolomic risk factors for type 2 diabetes have the potential to reveal new pathways and improve prediction and prevention. CONCLUSIONS Over the past several decades, the NHS and NHS II have made major contributions to public health recommendations and strategies designed to reduce the global burden of diabetes.
Collapse
Affiliation(s)
- Sylvia H Ley
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Andres V Ardisson Korat
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Qi Sun
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Deirdre K Tobias
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Cuilin Zhang
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Lu Qi
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Walter C Willett
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - JoAnn E Manson
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Frank B Hu
- Sylvia H. Ley, Andres V. Ardisson Korat, Qi Sun, Lu Qi, Walter C. Willett, and Frank B. Hu are with the Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA. Deirdre K. Tobias and JoAnn E. Manson are with the Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston. Cuilin Zhang is with Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| |
Collapse
|
48
|
Cigliola V, Populaire C, Pierri CL, Deutsch S, Haefliger JA, Fadista J, Lyssenko V, Groop L, Rueedi R, Thorel F, Herrera PL, Meda P. A Variant of GJD2, Encoding for Connexin 36, Alters the Function of Insulin Producing β-Cells. PLoS One 2016; 11:e0150880. [PMID: 26959991 PMCID: PMC4784816 DOI: 10.1371/journal.pone.0150880] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/20/2016] [Indexed: 01/16/2023] Open
Abstract
Signalling through gap junctions contributes to control insulin secretion and, thus, blood glucose levels. Gap junctions of the insulin-producing β-cells are made of connexin 36 (Cx36), which is encoded by the GJD2 gene. Cx36-null mice feature alterations mimicking those observed in type 2 diabetes (T2D). GJD2 is also expressed in neurons, which share a number of common features with pancreatic β-cells. Given that a synonymous exonic single nucleotide polymorphism of human Cx36 (SNP rs3743123) associates with altered function of central neurons in a subset of epileptic patients, we investigated whether this SNP also caused alterations of β-cell function. Transfection of rs3743123 cDNA in connexin-lacking HeLa cells resulted in altered formation of gap junction plaques and cell coupling, as compared to those induced by wild type (WT) GJD2 cDNA. Transgenic mice expressing the very same cDNAs under an insulin promoter revealed that SNP rs3743123 expression consistently lead to a post-natal reduction of islet Cx36 levels and β-cell survival, resulting in hyperglycemia in selected lines. These changes were not observed in sex- and age-matched controls expressing WT hCx36. The variant GJD2 only marginally associated to heterogeneous populations of diabetic patients. The data document that a silent polymorphism of GJD2 is associated with altered β-cell function, presumably contributing to T2D pathogenesis.
Collapse
Affiliation(s)
- Valentina Cigliola
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Celine Populaire
- Centre Hospitalier Régional Universitaire Besançon, Besançon, France
| | - Ciro L. Pierri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Samuel Deutsch
- Joint Genome Institute, Walnut Creek, California, United States of America
| | | | - João Fadista
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
- Steno Diabetes Center A/S, Gentofte, Denmark
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Rue du Bugnon 27, 1011, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Fabrizio Thorel
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Pedro Luis Herrera
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
49
|
Sandovici I, Hammerle CM, Cooper WN, Smith NH, Tarry-Adkins JL, Dunmore BJ, Bauer J, Andrews SR, Yeo GSH, Ozanne SE, Constância M. Ageing is associated with molecular signatures of inflammation and type 2 diabetes in rat pancreatic islets. Diabetologia 2016; 59:502-11. [PMID: 26699651 PMCID: PMC4742511 DOI: 10.1007/s00125-015-3837-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/17/2015] [Indexed: 01/04/2023]
Abstract
AIMS/HYPOTHESIS Ageing is a major risk factor for development of metabolic diseases such as type 2 diabetes. Identification of the mechanisms underlying this association could help to elucidate the relationship between age-associated progressive loss of metabolic health and development of type 2 diabetes. We aimed to determine molecular signatures during ageing in the endocrine pancreas. METHODS Global gene transcription was measured in pancreatic islets isolated from young and old rats by Ilumina BeadChip arrays. Promoter DNA methylation was measured by Sequenom MassArray in 46 genes that showed differential expression with age, and correlations with expression were established. Alterations in morphological and cellular processes with age were determined by immunohistochemical methods. RESULTS Age-related changes in gene expression were found at 623 loci (>1.5-fold, false discovery rate [FDR] <5%), with a significant (FDR < 0.05) enrichment in genes previously implicated in islet-cell function (Enpp1, Abcc8), type 2 diabetes (Tspan8, Kcnq1), inflammatory processes (Cxcl9, Il33) and extracellular matrix organisation (Col3a1, Dpt). Age-associated transcriptional differences negatively correlated with promoter DNA methylation at several loci related to inflammation, glucose homeostasis, cell proliferation and cell-matrix interactions (Il33, Cxcl9, Gpr119, Fbp2, Col3a1, Dpt, Spp1). CONCLUSIONS/INTERPRETATION Our findings suggest that a significant proportion of pancreatic islets develop a low-grade 'chronic' inflammatory status with ageing and this may trigger altered functional plasticity. Furthermore, we identified changes in expression of genes previously linked to type 2 diabetes and associated changes in DNA methylation that could explain their age-associated dysregulation. These findings provide new insights into key (epi)genetic signatures of the ageing process in islets.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital, Robinson Way, Cambridge, CB2 0SW, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Constanze M Hammerle
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital, Robinson Way, Cambridge, CB2 0SW, UK
| | - Wendy N Cooper
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital, Robinson Way, Cambridge, CB2 0SW, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Noel H Smith
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
| | - Jane L Tarry-Adkins
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
| | - Benjamin J Dunmore
- Cambridge Genomic Services, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Julien Bauer
- Cambridge Genomic Services, Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Giles S H Yeo
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- National Institute of Health Research, Cambridge Biomedical Research Centre, Cambridge, UK
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK.
- National Institute of Health Research, Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK.
- Department of Obstetrics and Gynaecology, University of Cambridge, The Rosie Hospital, Robinson Way, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- National Institute of Health Research, Cambridge Biomedical Research Centre, Cambridge, UK.
| |
Collapse
|
50
|
Joyce-Tan SM, Zain SM, Abdul Sattar MZ, Abdullah NA. Renin-Angiotensin System Gene Variants and Type 2 Diabetes Mellitus: Influence of Angiotensinogen. J Diabetes Res 2016; 2016:2161376. [PMID: 26682227 PMCID: PMC4670722 DOI: 10.1155/2016/2161376] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 04/24/2015] [Accepted: 05/11/2015] [Indexed: 12/27/2022] Open
Abstract
Genome-wide association studies (GWAS) have been successfully used to call for variants associated with diseases including type 2 diabetes mellitus (T2DM). However, some variants are not included in the GWAS to avoid penalty in multiple hypothetic testing. Thus, candidate gene approach is still useful even at GWAS era. This study attempted to assess whether genetic variations in the renin-angiotensin system (RAS) and their gene interactions are associated with T2DM risk. We genotyped 290 T2DM patients and 267 controls using three genes of the RAS, namely, angiotensin converting enzyme (ACE), angiotensinogen (AGT), and angiotensin II type 1 receptor (AGTR1). There were significant differences in allele frequencies between cases and controls for AGT variants (P = 0.05) but not for ACE and AGTR1. Haplotype TCG of the AGT was associated with increased risk of T2DM (OR 1.92, 95% CI 1.15-3.20, permuted P = 0.012); however, no evidence of significant gene-gene interactions was seen. Nonetheless, our analysis revealed that the associations of the AGT variants with T2DM were independently associated. Thus, this study suggests that genetic variants of the RAS can modestly influence the T2DM risk.
Collapse
Affiliation(s)
- Siew Mei Joyce-Tan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shamsul Mohd Zain
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Nor Azizan Abdullah
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- *Nor Azizan Abdullah:
| |
Collapse
|