1
|
Coppedè F, Franzago M, Giardina E, Nigro CL, Matullo G, Moltrasio C, Nacmias B, Pileggi S, Sirchia SM, Stoccoro A, Storlazzi CT, Stuppia L, Tricarico R, Merla G. A perspective on diet, epigenetics and complex diseases: where is the field headed next? Epigenomics 2022; 14:1281-1304. [DOI: 10.2217/epi-2022-0239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dietary factors can regulate epigenetic processes during life, modulating the intracellular pools of metabolites necessary for epigenetic reactions and regulating the activity of epigenetic enzymes. Their effects are strong during the prenatal life, when epigenetic patterns are written, allowing organogenesis. However, interactions between diet and the epigenome continue throughout life and likely contribute to the onset and progression of various complex diseases. Here, we review the contribution of dietary factors to the epigenetic changes observed in complex diseases and suggest future steps to better address this issue, focusing on neurobehavioral, neuropsychiatric and neurodegenerative disorders, cardiovascular diseases, obesity and Type 2 diabetes, cancer and inflammatory skin diseases.
Collapse
Affiliation(s)
- Fabio Coppedè
- Department of Translational Research & of New Surgical & Medical Technologies, University of Pisa, Pisa, 56126, Italy
| | - Marica Franzago
- Department of Medicine & Aging, School of Medicine & Health Sciences, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
- Center for Advanced Studies & Technology, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, Rome, 00179, Italy
- Department of Biomedicine & Prevention, Tor Vergata University of Rome, Rome, 00133, Italy
| | | | - Giuseppe Matullo
- Department of Medical Sciences, University of Turin, Turin, 10126, Italy
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
- Department of Medical Surgical & Health Sciences, University of Trieste, Trieste, 34137, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research & Child Health, University of Florence, Florence, 50139, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, 50143, Italy
| | - Silvana Pileggi
- Department of Health Sciences, Medical Genetics, University of Milan, Milan, 20142, Italy
| | - Silvia Maria Sirchia
- Department of Health Sciences, Medical Genetics, University of Milan, Milan, 20142, Italy
| | - Andrea Stoccoro
- Department of Translational Research & of New Surgical & Medical Technologies, University of Pisa, Pisa, 56126, Italy
| | | | - Liborio Stuppia
- Center for Advanced Studies & Technology, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
- Department of Psychological, Health & Territorial Sciences, School of Medicine & Health Sciences, “G. d'Annunzio” University of Chieti–Pescara, Chieti, 66100, Italy
| | - Rossella Tricarico
- Department of Biology & Biotechnology, University of Pavia, Pavia, 27100, Italy
| | - Giuseppe Merla
- Laboratory of Regulatory & Functional Genomics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, 71013, Italy
- Department of Molecular Medicine & Medical Biotechnology, University of Naples Federico II, Naples, 80131, Italy
| |
Collapse
|
2
|
Dawes K, Philibert W, Darbro B, Simons RL, Philibert R. Additive and Interactive Genetically Contextual Effects of HbA1c on cg19693031 Methylation in Type 2 Diabetes. Genes (Basel) 2022; 13:genes13040683. [PMID: 35456489 PMCID: PMC9025650 DOI: 10.3390/genes13040683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) has a complex genetic and environmental architecture that underlies its development and clinical presentation. Despite the identification of well over a hundred genetic variants and CpG sites that associate with T2D, a robust biosignature that could be used to prevent or forestall clinical disease has not been developed. Based on the premise that underlying genetic variation influences DNA methylation (DNAm) independently of or in combination with environmental exposures, we assessed the ability of local and distal gene x methylation (GxMeth) interactive effects to improve cg19693031 models for predicting T2D status in an African American cohort. Using genome-wide genetic data from 506 subjects, we identified a total of 1476 GxMeth terms associated with HbA1c values. The GxMeth SNPs map to biological pathways associated with the development and complications of T2D, with genetically contextual differences in methylation observed only in diabetic subjects for two GxMeth SNPs (rs2390998 AG vs. GG, p = 4.63 × 10−11, Δβ = 13%, effect size = 0.16 [95% CI = 0.05, 0.32]; rs1074390 AA vs. GG, p = 3.93 × 10−4, Δβ = 9%, effect size = 0.38 [95% CI = 0.12, 0.56]. Using a repeated stratified k-fold cross-validation approach, a series of balanced random forest classifiers with random under-sampling were built to evaluate the addition of GxMeth terms to cg19693031 models to discriminate between normoglycemic controls versus T2D subjects. The results were compared to those obtained from models incorporating only the covariates (age, sex and BMI) and the addition of cg19693031. We found a post-pruned classifier incorporating 10 GxMeth SNPs and cg19693031 adjusted for covariates predicted the T2D status, with the AUC, sensitivity, specificity and precision of the positive target class being 0.76, 0.81, 0.70 and 0.63, respectively. Comparatively, the AUC, sensitivity, specificity and precision using the covariates and cg19693031 were only 0.71, 0.74, 0.67 and 0.59, respectively. Collectively, we demonstrate correcting for genetic confounding of cg19693031 improves its ability to detect type 2 diabetes. We conclude that an integrated genetic–epigenetic approach could inform personalized medicine programming for more effective prevention and treatment of T2D.
Collapse
Affiliation(s)
- Kelsey Dawes
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (W.P.); (R.P.)
- Correspondence: ; Tel.: +1-319-361-2081
| | - Willem Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (W.P.); (R.P.)
| | - Benjamin Darbro
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Ronald L. Simons
- Department of Sociology, University of Georgia, Athens, GA 30602, USA;
| | - Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (W.P.); (R.P.)
- Behavioral Diagnostics LLC, Coralville, IA 52246, USA
- Cardio Diagnostics Inc., Coralville, IA 52246, USA
| |
Collapse
|
3
|
Miljkovic I, Cvejkus R, An P, Thyagarajan B, Christensen K, Wojczynski M, Schupf N, Zmuda JM. Low Risk for Developing Diabetes Among the Offspring of Individuals With Exceptional Longevity and Their Spouses. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:753986. [PMID: 36992755 PMCID: PMC10012150 DOI: 10.3389/fcdhc.2022.753986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022]
Abstract
Little is known about the risk of type 2 diabetes (T2D) among the offspring of individuals with exceptional longevity. We determined the incidence of and potential risk and protective factors for T2D among the offspring of probands and offspring’s spouses (mean age=60 years, range 32-88 years) in the Long Life Family Study (LLFS), a multicenter cohort study of 583 two-generation families with a clustering of healthy aging and exceptional longevity. Incident T2D was defined as fasting serum glucose ≥126 mg/dl, or HbA1c of ≥6.5%, or self-reported with doctor diagnosis of T2D, or the use of anti-diabetic medication during a mean follow-up 7.9 ± 1.1 years. Among offspring (n=1105) and spouses (n=328) aged 45-64 years without T2D at baseline visit, the annual incident rate of T2D was 3.6 and 3.0 per 1000 person-years, respectively, while among offspring (n=444) and spouses (n=153) aged 65+ years without T2D at baseline, the annual incident rate of T2D was 7.2 and 7.4 per 1000 person-years, respectively. By comparison, the annual incident rate of T2D per 1000 person-years in the U.S. general population was 9.9 among those aged 45-64, and 8.8 among those aged 65+ years (2018 National Health Interview Survey). Baseline BMI, waist circumference, and fasting serum triglycerides were positively associated with incident T2D, whereas fasting serum HDL-C, adiponectin, and sex hormone binding globulin were protective against incident T2D among the offspring (all P<0.05). Similar associations were observed among their spouses (all P<0.05, except sex hormone binding globulin). In addition, we observed that among spouses, but not offspring, fasting serum interleukin 6 and insulin-like growth factor 1 were positively associated with incident T2D (P<0.05 for both). Our study suggests that both offspring of long-living individuals and their spouses, especially middle-aged, share a similar low risk for developing T2D as compared with the general population. Our findings also raise the possibility that distinct biological risk and protective factors may contribute to T2D risk among offspring of long-lived individuals when compared with their spouses. Future studies are needed to identify the mechanisms underlying low T2D risk among the offspring of individuals with exceptional longevity, and also among their spouses.
Collapse
Affiliation(s)
- Iva Miljkovic
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- *Correspondence: Iva Miljkovic,
| | - Ryan Cvejkus
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ping An
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Kaare Christensen
- Department of Epidemiology, Biostatistics and Biodemography, Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| | - Mary Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Nicole Schupf
- Taub Institute, Columbia University, New York, NY, United States
| | - Joseph M. Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Gaddas M, Latiri I, Kebaili R, Kacem I, Jaballah N, Maatoug J, Salaani M, Boughammoura L, Ben Saad H. Reversibility of pancreatic β-cells dysfunction after vitamin D and calcium supplementation: a pilot study in a population of obese and prepubescent North-African children. Libyan J Med 2022; 17:2059896. [PMID: 35388742 PMCID: PMC9004520 DOI: 10.1080/19932820.2022.2059896] [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] [Indexed: 11/18/2022] Open
Abstract
The mechanisms of diabetogenesis in children remain largely obscure. This study aimed to determine the impact of vitamin D and calcium supplementation on pancreatic β-cells function in terms of insulin secretion and sensitivity. This was a quasi-experimental study involving 30 obese and prepubescent Tunisian children (57% boys). During three months, the children received calcium and vitamin D supplementation at therapeutic doses. An oral glucose tolerance test (OGTT) was performed at the beginning and at the end of the study. The following metabolic definitions were applied: i) hyperinsulinism: insulinemia sum > 300 μ UI/ml during OGTT, ii) insulin-resistance: homeostatic model assessment of insulin-resistance > 2, iii) normal glycaemic profile: normal plasma levels during OGTT without any spike, and iv) pancreatic β-cells dysfunction reversibility: disappearance of the aforementioned disorders. The means ± standard-deviation of age and body mass index were 10.87 ± 1.9 years, and 30.17 ± 4.99 kg/m2, respectively. All children were at the stage of hyperinsulinism associated with insulin-resistance. These disturbances were noted even in children having a normal glycaemic profile at OGTT. After calcium and vitamin D supplementation, glycaemic profile as well as insulin-secretion improved significantly (p < 0.0001). Hyperinsulinism and insulin-resistance decreased significantly by 56.67% (p < 0.0001) and 70.00% (p < 0.0001), respectively. Complete reversibility of these two disorders was noted in 26.6% of children. To conclude, in obese and prepubescent children, vitamin D and calcium supplementation led to the reversibility of the pancreatic β-cells dysfunction.
Collapse
Affiliation(s)
- Meriem Gaddas
- Faculté de Médecine de Sousse, Service de Physiologie et Explorations Fonctionnelles, Université de Sousse, Sousse, Tunisie.,University of Sousse, Farhat HACHED Hospital, Departement of physiology and functional explorations, Sousse, Tunisia Sousse
| | - Imed Latiri
- Faculté de Médecine de Sousse, Service de Physiologie et Explorations Fonctionnelles, Université de Sousse, Sousse, Tunisie.,Laboratoire de recherche LR12SP09 «Insuffisance cardiaque», Université de Sousse, Hôpital Farhat HACHED, Sousse, Tunisie
| | - Raoudha Kebaili
- Paediatrics Department, Farhat HACHED Hospital of Sousse, University of Sousse, Sousse, Tunisia
| | - Ilhem Kacem
- Basic Health Center «Sousse Jawhara", Outpatient consultation, Sousse, Tunisia
| | - Nesrine Jaballah
- Paediatrics Department, Farhat HACHED Hospital of Sousse, University of Sousse, Sousse, Tunisia
| | - Jihene Maatoug
- Epidemiology Department, Farhat HACHED Hospital of Sousse, University of Sousse, Sousse, Tunisia
| | - Mohamed Salaani
- Paediatrics Department, Farhat HACHED Hospital of Sousse, University of Sousse, Sousse, Tunisia
| | - Lamia Boughammoura
- Paediatrics Department, Farhat HACHED Hospital of Sousse, University of Sousse, Sousse, Tunisia
| | - Helmi Ben Saad
- Faculté de Médecine de Sousse, Service de Physiologie et Explorations Fonctionnelles, Université de Sousse, Sousse, Tunisie.,University of Sousse, Farhat HACHED Hospital, Departement of physiology and functional explorations, Sousse, Tunisia Sousse.,Laboratoire de recherche LR12SP09 «Insuffisance cardiaque», Université de Sousse, Hôpital Farhat HACHED, Sousse, Tunisie
| |
Collapse
|
5
|
Li X, Qi L. Epigenetics in Precision Nutrition. J Pers Med 2022; 12:jpm12040533. [PMID: 35455649 PMCID: PMC9027461 DOI: 10.3390/jpm12040533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/14/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Precision nutrition is an emerging area of nutrition research, with primary focus on the individual variability in response to dietary and lifestyle factors, which are mainly determined by an individual’s intrinsic variations, such as those in genome, epigenome, and gut microbiome. The current research on precision nutrition is heavily focused on genome and gut microbiome, while epigenome (DNA methylation, non-coding RNAs, and histone modification) is largely neglected. The epigenome acts as the interface between the human genome and environmental stressors, including diets and lifestyle. Increasing evidence has suggested that epigenetic modifications, particularly DNA methylation, may determine the individual variability in metabolic health and response to dietary and lifestyle factors and, therefore, hold great promise in discovering novel markers for precision nutrition and potential targets for precision interventions. This review summarized recent studies on DNA methylation with obesity, diabetes, and cardiovascular disease, with more emphasis put in the relations of DNA methylation with nutrition and diet/lifestyle interventions. We also briefly reviewed other epigenetic events, such as non-coding RNAs, in relation to human health and nutrition, and discussed the potential role of epigenetics in the precision nutrition research.
Collapse
Affiliation(s)
- Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA;
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Correspondence: ; Tel.: +1-504-988-7259
| |
Collapse
|
6
|
Li X, Zhao W, Peng L, Li Y, Nie S, Yu H, Qin Y, Zhang H. Elevated serum extracellular vesicle arginase 1 in type 2 diabetes mellitus: a cross-sectional study in middle-aged and elderly population. BMC Endocr Disord 2022; 22:62. [PMID: 35277141 PMCID: PMC8917686 DOI: 10.1186/s12902-022-00982-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 03/01/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Serum extracellular vesicle (EV)-derived arginase 1 (ARG 1) plays a critical role in diabetes-associated endothelial dysfunction. This study was performed to determine the levels of serum EV-derived ARG 1 in T2DM and non-T2DM participants and to examine the association of serum EV-derived ARG 1 with T2DM incidence. METHODS We performed a cross-sectional study in 103 Chinese, including 73 T2DM patients and 30 non-T2DM. Serum EVs were prepared via ultracentrifugation. Serum EV-derived ARG 1 levels were measured by enzyme-linked immunosorbent assay. The correlations between serum EV-derived ARG 1 and clinical variables were analyzed. The association of serum EV-derived ARG 1 levels with T2DM was determined by multivariate logistic regression analysis. Interaction subgroup analysis was used to evaluate the interaction of the relevant baselines on the association between serum EV-derived ARG 1 levels and T2DM. RESULTS Serum EV-derived ARG 1 levels were significantly higher in T2DM patients compared with non-T2DM patients (p < 0.001). Correlation analysis revealed that serum EV-derived ARG 1 levels were positively associated with fasting plasma glucose (FPG) (r = 0.316, p = 0.001) and glycated hemoglobin (HbA1c) (r = 0.322, p = 0.001). Serum EV-derived ARG 1 levels were significantly associated with T2DM, especially in the subgroup of T2DM for more than 10 years (OR 1.651, 95% CI = 1.066-2.557; P value, 0.025), after adjusting for confounding factors. CONCLUSIONS Elevated concentration of serum EV-derived ARG 1 is closely associated with T2DM.
Collapse
Affiliation(s)
- Xinwei Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Wen Zhao
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Lu Peng
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Yu Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Shaoping Nie
- Department of Emergency, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huahui Yu
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Yanwen Qin
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Huina Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China.
| |
Collapse
|
7
|
De Sousa RAL, Improta-Caria AC. Regulation of microRNAs in Alzheimer´s disease, type 2 diabetes, and aerobic exercise training. Metab Brain Dis 2022; 37:559-580. [PMID: 35075500 DOI: 10.1007/s11011-022-00903-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia. The evolution and aggregation of amyloid beta (β) oligomers is linked to insulin resistance in AD, which is also the major characteristic of type 2 diabetes (T2D). Being physically inactive can contribute to the development of AD and/or T2D. Aerobic exercise training (AET), a type of physical exercise, can be useful in preventing or treating the negative outcomes of AD and T2D. AD, T2D and AET can regulate the expression of microRNAs (miRNAs). Here, we review some of the changes in miRNAs expression regulated by AET, AD and T2D. MiRNAs play an important role in the gene regulation of key signaling pathways in both pathologies, AD and T2D. MiRNA dysregulation is evident in AD and has been associated with several neuropathological alterations, such as the development of a reactive gliosis. Expression of miRNAs are associated with many pathophysiological mechanisms involved in T2D like insulin synthesis, insulin resistance, glucose intolerance, hyperglycemia, intracellular signaling, and lipid profile. AET regulates miRNAs levels. We identified 5 miRNAs (miR-21, miR-29a/b, miR-103, miR-107, and miR-195) that regulate gene expression and are modulated by AET on AD and T2D. The identified miRNAs are potential targets to treat the symptoms of AD and T2D. Thus, AET is a non-pharmacological tool that can be used to prevent and fight the negative outcomes in AD and T2D.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Programa Multicêntrico de Pós-Graduação Em Ciências Fisiológicas- Sociedade Brasileira de Fisiologia (SBFis), Universidade Federal Dos Vales Do Jequitinhonha E Mucuri (UFVJM), Campus JK, Rodovia MGT 367, Km 583, Alto da Jacuba, nº 5000, Diamantina, Minas Gerais, CEP 39100-000, Brazil.
| | - Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Bahia, Brazil
| |
Collapse
|
8
|
Nettore IC, Franchini F, Palatucci G, Macchia PE, Ungaro P. Epigenetic Mechanisms of Endocrine-Disrupting Chemicals in Obesity. Biomedicines 2021; 9:biomedicines9111716. [PMID: 34829943 PMCID: PMC8615468 DOI: 10.3390/biomedicines9111716] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
The incidence of obesity has dramatically increased over the last decades. Recently, there has been a growing interest in the possible association between the pandemics of obesity and some endocrine-disrupting chemicals (EDCs), termed “obesogens”. These are a heterogeneous group of exogenous compounds that can interfere in the endocrine regulation of energy metabolism and adipose tissue structure. Oral intake, inhalation, and dermal absorption represent the major sources of human exposure to these EDCs. Recently, epigenetic changes such as the methylation of cytosine residues on DNA, post-translational modification of histones, and microRNA expression have been considered to act as an intermediary between deleterious effects of EDCs and obesity development in susceptible individuals. Specifically, EDCs exposure during early-life development can detrimentally affect individuals via inducing epigenetic modifications that can permanently change the epigenome in the germline, enabling changes to be transmitted to the next generations and predisposing them to a multitude of diseases. The purpose of this review is to analyze the epigenetic alterations putatively induced by chemical exposures and their ability to interfere with the control of energy metabolism and adipose tissue regulation, resulting in imbalances in the control of body weight, which can lead to obesity.
Collapse
Affiliation(s)
- Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Fabiana Franchini
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Giuseppe Palatucci
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Paola Ungaro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale del CNR “G. Salvatore”, Via S. Pansini, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-770-4795
| |
Collapse
|
9
|
DNA Methylation and Type 2 Diabetes: Novel Biomarkers for Risk Assessment? Int J Mol Sci 2021; 22:ijms222111652. [PMID: 34769081 PMCID: PMC8584054 DOI: 10.3390/ijms222111652] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes is a severe threat to global health. Almost 500 million people live with diabetes worldwide. Most of them have type 2 diabetes (T2D). T2D patients are at risk of developing severe and life-threatening complications, leading to an increased need for medical care and reduced quality of life. Improved care for people with T2D is essential. Actions aiming at identifying undiagnosed diabetes and at preventing diabetes in those at high risk are needed as well. To this end, biomarker discovery and validation of risk assessment for T2D are critical. Alterations of DNA methylation have recently helped to better understand T2D pathophysiology by explaining differences among endophenotypes of diabetic patients in tissues. Recent evidence further suggests that variations of DNA methylation might contribute to the risk of T2D even more significantly than genetic variability and might represent a valuable tool to predict T2D risk. In this review, we focus on recent information on the contribution of DNA methylation to the risk and the pathogenesis of T2D. We discuss the limitations of these studies and provide evidence supporting the potential for clinical application of DNA methylation marks to predict the risk and progression of T2D.
Collapse
|
10
|
Capparelli R, Iannelli D. Role of Epigenetics in Type 2 Diabetes and Obesity. Biomedicines 2021; 9:977. [PMID: 34440181 PMCID: PMC8393970 DOI: 10.3390/biomedicines9080977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022] Open
Abstract
Epigenetic marks the genome by DNA methylation, histone modification or non-coding RNAs. Epigenetic marks instruct cells to respond reversibly to environmental cues and keep the specific gene expression stable throughout life. In this review, we concentrate on DNA methylation, the mechanism often associated with transgenerational persistence and for this reason frequently used in the clinic. A large study that included data from 10,000 blood samples detected 187 methylated sites associated with body mass index (BMI). The same study demonstrates that altered methylation results from obesity (OB). In another study the combined genetic and epigenetic analysis allowed us to understand the mechanism associating hepatic insulin resistance and non-alcoholic disease in Type 2 Diabetes (T2D) patients. The study underlines the therapeutic potential of epigenetic studies. We also account for seemingly contradictory results associated with epigenetics.
Collapse
Affiliation(s)
- Rosanna Capparelli
- Department of Agriculture Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy
| | - Domenico Iannelli
- Department of Agriculture Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy
| |
Collapse
|
11
|
Juvinao-Quintero DL, Marioni RE, Ochoa-Rosales C, Russ TC, Deary IJ, van Meurs JBJ, Voortman T, Hivert MF, Sharp GC, Relton CL, Elliott HR. DNA methylation of blood cells is associated with prevalent type 2 diabetes in a meta-analysis of four European cohorts. Clin Epigenetics 2021; 13:40. [PMID: 33622391 PMCID: PMC7903628 DOI: 10.1186/s13148-021-01027-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/11/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a heterogeneous disease with well-known genetic and environmental risk factors contributing to its prevalence. Epigenetic mechanisms related to changes in DNA methylation (DNAm), may also contribute to T2D risk, but larger studies are required to discover novel markers, and to confirm existing ones. RESULTS We performed a large meta-analysis of individual epigenome-wide association studies (EWAS) of prevalent T2D conducted in four European studies using peripheral blood DNAm. Analysis of differentially methylated regions (DMR) was also undertaken, based on the meta-analysis results. We found three novel CpGs associated with prevalent T2D in Europeans at cg00144180 (HDAC4), cg16765088 (near SYNM) and cg24704287 (near MIR23A) and confirmed three CpGs previously identified (mapping to TXNIP, ABCG1 and CPT1A). We also identified 77 T2D associated DMRs, most of them hypomethylated in T2D cases versus controls. In adjusted regressions among diabetic-free participants in ALSPAC, we found that all six CpGs identified in the meta-EWAS were associated with white cell-types. We estimated that these six CpGs captured 11% of the variation in T2D, which was similar to the variation explained by the model including only the common risk factors of BMI, sex, age and smoking (R2 = 10.6%). CONCLUSIONS This study identifies novel loci associated with T2D in Europeans. We also demonstrate associations of the same loci with other traits. Future studies should investigate if our findings are generalizable in non-European populations, and potential roles of these epigenetic markers in T2D etiology or in determining long term consequences of T2D.
Collapse
Affiliation(s)
- Diana L. Juvinao-Quintero
- MRC Integrative Epidemiology, Bristol Medical School, Bristol, BS8 2BN UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN UK
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA 02215 USA
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN UK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Carolina Ochoa-Rosales
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA The Netherlands
- Centro de Vida Saludable de La Universidad de Concepción, Victoria 580, Concepción, Chile
| | - Tom C. Russ
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ UK
- Edinburgh Dementia Prevention Research Group, University of Edinburgh, Edinburgh, EH16 4UX UK
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ UK
| | - Ian J. Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ UK
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ UK
| | - Joyce B. J. van Meurs
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, 3000 CA The Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA The Netherlands
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA 02215 USA
| | - Gemma C. Sharp
- MRC Integrative Epidemiology, Bristol Medical School, Bristol, BS8 2BN UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN UK
| | - Caroline L. Relton
- MRC Integrative Epidemiology, Bristol Medical School, Bristol, BS8 2BN UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN UK
- Bristol NIHR Biomedical Research Centre, Oakfield House, Oakfield Grove, Bristol, BS8 2BN UK
| | - Hannah R. Elliott
- MRC Integrative Epidemiology, Bristol Medical School, Bristol, BS8 2BN UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN UK
| |
Collapse
|
12
|
Bele S, Girada SB, Ray A, Gupta A, Oruganti S, Prakash Babu P, Rayalla RSR, Kalivendi SV, Ibrahim A, Puri V, Adalla V, Katika MR, DiMarchi R, Mitra P. MS-275, a class 1 histone deacetylase inhibitor augments glucagon-like peptide-1 receptor agonism to improve glycemic control and reduce obesity in diet-induced obese mice. eLife 2020; 9:e52212. [PMID: 33349332 PMCID: PMC7755393 DOI: 10.7554/elife.52212] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/06/2020] [Indexed: 12/20/2022] Open
Abstract
Given its glycemic efficacy and ability to reduce the body weight, glucagon-like peptide 1 receptor (GLP-1R) agonism has emerged as a preferred treatment for diabetes associated with obesity. We here report that a small-molecule Class 1 histone deacetylase (HDAC) inhibitor Entinostat (MS-275) enhances GLP-1R agonism to potentiate glucose-stimulated insulin secretion and decrease body weight in diet-induced obese (DIO) mice. MS-275 is not an agonist or allosteric activator of GLP-1R but enhances the sustained receptor-mediated signaling through the modulation of the expression of proteins involved in the signaling pathway. MS-275 and liraglutide combined therapy improved fasting glycemia upon short-term treatment and a chronic administration causes a reduction of obesity in DIO mice. Overall, our results emphasize the therapeutic potential of MS-275 as an adjunct to GLP-1R therapy in the treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Shilpak Bele
- Dr. Reddy’s Institute of Life Sciences University of Hyderabad CampusHyderabadIndia
- Manipal Academy of Higher EducationManipalIndia
| | - Shravan Babu Girada
- Dr. Reddy’s Institute of Life Sciences University of Hyderabad CampusHyderabadIndia
| | - Aramita Ray
- Dr. Reddy’s Institute of Life Sciences University of Hyderabad CampusHyderabadIndia
| | - Abhishek Gupta
- Department of Biomedical Sciences and Diabetes Institute, Ohio UniversityAthensUnited States
| | - Srinivas Oruganti
- Dr. Reddy’s Institute of Life Sciences University of Hyderabad CampusHyderabadIndia
| | | | | | | | - Ahamed Ibrahim
- Division of Lipid Chemistry, National Institute of Nutrition HyderabadHyderabadIndia
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes Institute, Ohio UniversityAthensUnited States
| | - Venkateswar Adalla
- Medical Genomics, QIMR Berghofer Medical Research InstituteHerstonAustralia
| | - Madhumohan R Katika
- Stem Cell and Regenerative Medicine Department, Nizam’s Institute of Medical SciencesHyderabadIndia
| | - Richard DiMarchi
- Department of Chemistry, Indiana UniversityBloomingtonUnited States
| | - Prasenjit Mitra
- Dr. Reddy’s Institute of Life Sciences University of Hyderabad CampusHyderabadIndia
| |
Collapse
|
13
|
Vitamin D Supplementation Is Associated with Increased Glutathione Peroxidase-1 Levels in Arab Adults with Prediabetes. Antioxidants (Basel) 2020; 9:antiox9020118. [PMID: 32013162 PMCID: PMC7070325 DOI: 10.3390/antiox9020118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/26/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
Vitamin D supplementation may be used to lower oxidative stress. This interventional study aimed to investigate the effects of vitamin D supplementation on glutathione peroxidase 1 (GPx1) levels and other parameters in Arab adults with prediabetes. A total of 203 Saudi adults with prediabetes and vitamin D deficiency [intervention group, N = 146 (53 males and 93 females); control group, N = 57 (25 males and 32 females)] were included in this non-randomized, six-month intervention study. The intervention group received 50,000 international units (IU) cholecalciferol tablets once a week for two months, then twice a month for the next two months, followed by 1000 IU daily for the last two months. The control group received no supplementation. Serum 25(OH)D, lipid profile, glucose, C-reactive protein (CRP) and GPx1 were measured at baseline and after six months. Post-intervention, GPx1 concentrations increased significantly in the intervention group [17.3 (11.5–59.0) vs 26.7 (11.4–59.9) p < 0.01] while no changes were observed in the control group (p = 0.15). This significant increase in 25(OH)D and GPx1 levels persisted after adjusting for age and BMI. Stratification according to sex revealed that this favourable increase in GPx1 was true only for males (p = 0.002). In all groups, baseline GPx1 was inversely correlated with low density lipoprotein (LDL)-cholesterol (r = −0.26, p < 0.01) and body mass index (BMI) (r = −0.20, p < 0.05), while positively correlated with age (r = 0.18, p < 0.05) and systolic blood pressure (r = 0.19, p < 0.05). In conclusion, vitamin D supplementation favourably enhanced GPx1 levels in adult Arabs with prediabetes, particularly in males.
Collapse
|
14
|
Sangalli E, Tagliabue E, Sala LL, Prattichizzo F, Uccellatore A, Spada D, Lorino F, de Candia P, Lupini S, Cantone L, Favero C, Madeddu P, Bollati V, Genovese S, Spinetti G. Circulating MicroRNA-15a Associates With Retinal Damage in Patients With Early Stage Type 2 Diabetes. Front Endocrinol (Lausanne) 2020; 11:254. [PMID: 32390950 PMCID: PMC7192007 DOI: 10.3389/fendo.2020.00254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/06/2020] [Indexed: 11/30/2022] Open
Abstract
Circulating microRNAs are potential biomarkers of type 2 diabetes mellitus (T2DM) and related complications. Here, we investigated the association of microRNA-15a with early retinal damage in T2DM. A cohort of untreated subjects screened for intermediate/high risk of T2DM, according to a score assessment questionnaire, and then recognized to have a normal (NGT) or impaired (IGT) glucose tolerance or T2DM was studied. The thickness of the ganglion cell complex (GCC), an early marker of retinal degeneration anteceding overt retinopathy was assessed by Optical Coherence Tomography. Total and extracellular vesicles (EV)-associated microRNA-15a quantity was measured in plasma by real time PCR. MicroRNA-15a level was significantly higher in subjects with IGT and T2DM compared with NGT. MicroRNA-15a abundance was correlated to body mass index and classical diabetes biomarkers, including fasting glucose, HbA1c, insulinemia, and HOMA-IR. Moreover, GCC thickness was significantly reduced in IGT and T2DM subjects compared with NGT controls. Importantly, total microRNA-15a correlated with GCC in IGT subjects, while in T2DM subjects, EV-microRNA-15a negatively correlated with GCC, suggesting that microRNA-15a may monitor initial retinal damage. The assessment of plasma microRNA-15a may help refining risk assessment and secondary prevention in patients with preclinical T2DM.
Collapse
Affiliation(s)
| | | | | | | | - AnnaChiara Uccellatore
- Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | | | | | | | - Silvia Lupini
- Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Laura Cantone
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Chiara Favero
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Paolo Madeddu
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Gaia Spinetti
- IRCCS MultiMedica, Milan, Italy
- *Correspondence: Gaia Spinetti
| |
Collapse
|
15
|
Middleton TL, Brooks BA, Constantino MI, Wu T, Wong J, Yue DK. Maternal vs paternal diabetes: The parental history is different in younger onset versus older onset type 2 diabetes. J Diabetes Complications 2019; 33:107440. [PMID: 31676253 DOI: 10.1016/j.jdiacomp.2019.107440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/08/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND A number of previous studies exploring family history of type 2 diabetes have reported a predominance of maternal diabetes. These studies have not explicitly compared parental history of diabetes across the spectrum of disease onset from youth to later adulthood. METHODS Family history data from 11,467 patients with type 2 diabetes were extracted from the RPA Diabetes Centre database. Parental histories of diabetes were compared across a range of age of diagnosis strata (15-<30, 30-<40, 40-<50, 50-<60 and 60-<70 years). For the young-onset group (diagnosed between 15 and 30 years of age), associations between parental history of diabetes and the presence of cardio-metabolic risk factors and diabetic complications were also explored. RESULTS For the total cohort and within each age of diagnosis strata, more individuals reported maternal history than paternal history of diabetes. The young-onset group demonstrated the highest prevalence of any parental history of diabetes (60.7%), the highest combined maternal and paternal history (15.8%) and the smallest differential between maternal (25.1%) and paternal (19.7%) history of diabetes. Within the young-onset group, no significant association between parental history and cardio-metabolic risk factors or diabetic complications were identified after a median of 15.0 years of diabetes exposure. CONCLUSION Overall, our results demonstrate a consistent maternal excess of diabetes which could be consistent with an underlying epigenetic effect. However, the differential between maternal and paternal history is significantly lower in the young-onset group. Earlier emergence of type 2 diabetes may therefore reflect a different interaction and impact of genetic and environmental factors.
Collapse
Affiliation(s)
- Timothy L Middleton
- Diabetes Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; School of Medicine, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Belinda A Brooks
- Diabetes Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; Susan Wakil School of Nursing and Midwifery, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | - Maria I Constantino
- Diabetes Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; School of Medicine, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ted Wu
- Diabetes Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Jencia Wong
- Diabetes Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; School of Medicine, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Dennis K Yue
- Diabetes Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; School of Medicine, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Werissa NA, Piko P, Fiatal S, Kosa Z, Sandor J, Adany R. SNP-Based Genetic Risk Score Modeling Suggests No Increased Genetic Susceptibility of the Roma Population to Type 2 Diabetes Mellitus. Genes (Basel) 2019; 10:genes10110942. [PMID: 31752367 PMCID: PMC6896051 DOI: 10.3390/genes10110942] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/12/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND In a previous survey, an elevated fasting glucose level (FG) and/or known type 2 diabetes mellitus (T2DM) were significantly more frequent in the Roma population than in the Hungarian general population. We assessed whether the distribution of 16 single nucleotide polymorphisms (SNPs) with unequivocal effects on the development of T2DM contributes to this higher prevalence. METHODS Genetic risk scores, unweighted (GRS) and weighted (wGRS), were computed and compared between the study populations. Associations between GRSs and FG levels and T2DM status were investigated in separate and combined study populations. RESULTS The Hungarian general population carried a greater genetic risk for the development of T2DM (GRSGeneral = 15.38 ± 2.70 vs. GRSRoma = 14.80 ± 2.68, p < 0.001; wGRSGeneral = 1.41 ± 0.32 vs. wGRSRoma = 1.36 ± 0.31, p < 0.001). In the combined population models, GRSs and wGRSs showed significant associations with elevated FG (p < 0.001) and T2DM (p < 0.001) after adjusting for ethnicity, age, sex, body mass index (BMI), high-density Lipoprotein Cholesterol (HDL-C), and triglyceride (TG). In these models, the effect of ethnicity was relatively strong on both outcomes (FG levels: βethnicity = 0.918, p < 0.001; T2DM status: ORethnicity = 2.484, p < 0.001). CONCLUSIONS The higher prevalence of elevated FG and/or T2DM among Roma does not seem to be directly linked to their increased genetic load but rather to their environmental/cultural attributes. Interventions targeting T2DM prevention among Roma should focus on harmful environmental exposures related to their unhealthy lifestyle.
Collapse
Affiliation(s)
- Nardos Abebe Werissa
- MTA−DE Public Health Research Group of the Hungarian Academy of Sciences, Public Health Research Institute, University of Debrecen, 4028 Debrecen, Hungary; (N.A.W.); (P.P.)
- Doctorial School of Health Sciences, University of Debrecen, 4028 Debrecen, Hungary
| | - Peter Piko
- MTA−DE Public Health Research Group of the Hungarian Academy of Sciences, Public Health Research Institute, University of Debrecen, 4028 Debrecen, Hungary; (N.A.W.); (P.P.)
| | - Szilvia Fiatal
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, 4028 Debrecen, Hungary; (S.F.); (J.S.)
- WHO Collaborating Centre on Vulnerability and Health, University of Debrecen, 4028 Debrecen, Hungary
| | - Zsigmond Kosa
- Department of Health Visitor Methodology and Public Health, Faculty of Health, University of Debrecen, 4400 Nyíregyháza, Hungary;
| | - Janos Sandor
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, 4028 Debrecen, Hungary; (S.F.); (J.S.)
- WHO Collaborating Centre on Vulnerability and Health, University of Debrecen, 4028 Debrecen, Hungary
| | - Roza Adany
- MTA−DE Public Health Research Group of the Hungarian Academy of Sciences, Public Health Research Institute, University of Debrecen, 4028 Debrecen, Hungary; (N.A.W.); (P.P.)
- WHO Collaborating Centre on Vulnerability and Health, University of Debrecen, 4028 Debrecen, Hungary
- Correspondence: ; Tel: +36-5251-2764
| |
Collapse
|
17
|
Juvinao-Quintero DL, Hivert MF, Sharp GC, Relton CL, Elliott HR. DNA Methylation and Type 2 Diabetes: the Use of Mendelian Randomization to Assess Causality. CURRENT GENETIC MEDICINE REPORTS 2019; 7:191-207. [PMID: 32274260 PMCID: PMC7145450 DOI: 10.1007/s40142-019-00176-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose of Review This review summarises recent advances in the field of epigenetics in order to understand the aetiology of type 2 diabetes (T2D). Recent Findings DNA methylation at a number of loci has been shown to be robustly associated with T2D, including TXNIP, ABCG1, CPT1A, and SREBF1. However, due to the cross-sectional nature of many epidemiological studies and predominant analysis in samples derived from blood rather than disease relevant tissues, inferring causality is difficult. We therefore outline the use of Mendelian randomisation (MR) as one method able to assess causality in epigenetic studies of T2D. Summary Epidemiological studies have been fruitful in identifying epigenetic markers of T2D. Triangulation of evidence including utilisation of MR is essential to delineate causal from non-causal biomarkers of disease. Understanding the causality of epigenetic markers in T2D more fully will aid prioritisation of CpG sites as early biomarkers to detect disease or in drug development to target epigenetic mechanisms in order to treat patients.
Collapse
Affiliation(s)
- Diana L Juvinao-Quintero
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, USA
| | - Gemma C Sharp
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Bristol NIHR Biomedical Research Centre, Bristol, UK
| | - Hannah R Elliott
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| |
Collapse
|
18
|
Mononen N, Lyytikäinen LP, Seppälä I, Mishra PP, Juonala M, Waldenberger M, Klopp N, Illig T, Leiviskä J, Loo BM, Laaksonen R, Oksala N, Kähönen M, Hutri-Kähönen N, Raitakari O, Lehtimäki T, Raitoharju E. Whole blood microRNA levels associate with glycemic status and correlate with target mRNAs in pathways important to type 2 diabetes. Sci Rep 2019; 9:8887. [PMID: 31222113 PMCID: PMC6586838 DOI: 10.1038/s41598-019-43793-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 04/29/2019] [Indexed: 12/25/2022] Open
Abstract
We analyzed the associations between whole blood microRNA profiles and the indices of glucose metabolism and impaired fasting glucose and examined whether the discovered microRNAs correlate with the expression of their mRNA targets. MicroRNA and gene expression profiling were performed for the Young Finns Study participants (n = 871). Glucose, insulin, and glycated hemoglobin (HbA1c) levels were measured, the insulin resistance index (HOMA2-IR) was calculated, and the glycemic status (normoglycemic [n = 534]/impaired fasting glucose [IFG] [n = 252]/type 2 diabetes [T2D] [n = 24]) determined. Levels of hsa-miR-144-5p, -122-5p, -148a-3p, -589-5p, and hsa-let-7a-5p associated with glycemic status. hsa-miR-144-5p and -148a-3p associated with glucose levels, while hsa-miR-144-5p, -122-5p, -184, and -339-3p associated with insulin levels and HOMA2-IR, and hsa-miR-148a-3p, -15b-3p, -93-3p, -146b-5p, -221-3p, -18a-3p, -642a-5p, and -181-2-3p associated with HbA1c levels. The targets of hsa-miR-146b-5p that correlated with its levels were enriched in inflammatory pathways, and the targets of hsa-miR-221-3p were enriched in insulin signaling and T2D pathways. These pathways showed indications of co-regulation by HbA1c-associated miRNAs. There were significant differences in the microRNA profiles associated with glucose, insulin, or HOMA-IR compared to those associated with HbA1c. The HbA1c-associated miRNAs also correlated with the expression of target mRNAs in pathways important to the development of T2D.
Collapse
Affiliation(s)
- Nina Mononen
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Markus Juonala
- Division of Medicine, Turku University Hospital, and Department of Medicine, University of Turku, Turku, Finland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum, German Research Center for Environmental Health, Munich, Germany
| | - Norman Klopp
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany.,Institute for Human Genetics, Hannover Medical School, Hanover, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum, German Research Center for Environmental Health, Munich, Germany.,Hannover Unified Biobank, Hannover Medical School, Hannover, Germany.,Institute for Human Genetics, Hannover Medical School, Hanover, Germany
| | - Jaana Leiviskä
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital HUSLAB, Helsinki, Finland
| | - Britt-Marie Loo
- Joint Clinical Biochemistry Laboratory of the University of Turku and Turku University Central Hospital and Department of Chronic Disease Prevention, National Institute for Health and Welfare, Turku, Finland
| | - Reijo Laaksonen
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Niku Oksala
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Centre for Vascular Surgery and Interventional Radiology, Tampere University Hospital, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Olli Raitakari
- Research Centre for Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine and Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories, and the Finnish Cardiovascular Research Center, Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
19
|
Parrillo L, Spinelli R, Nicolò A, Longo M, Mirra P, Raciti GA, Miele C, Beguinot F. Nutritional Factors, DNA Methylation, and Risk of Type 2 Diabetes and Obesity: Perspectives and Challenges. Int J Mol Sci 2019; 20:ijms20122983. [PMID: 31248068 PMCID: PMC6627657 DOI: 10.3390/ijms20122983] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022] Open
Abstract
A healthy diet improves life expectancy and helps to prevent common chronic diseases such as type 2 diabetes (T2D) and obesity. The mechanisms driving these effects are not fully understood, but are likely to involve epigenetics. Epigenetic mechanisms control gene expression, maintaining the DNA sequence, and therefore the full genomic information inherited from our parents, unchanged. An interesting feature of epigenetic changes lies in their dynamic nature and reversibility. Accordingly, they are susceptible to correction through targeted interventions. Here we will review the evidence supporting a role for nutritional factors in mediating metabolic disease risk through DNA methylation changes. Special emphasis will be placed on the potential of using DNA methylation traits as biomarkers to predict risk of obesity and T2D as well as on their response to dietary and pharmacological (epi-drug) interventions.
Collapse
Affiliation(s)
- Luca Parrillo
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Rosa Spinelli
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Antonella Nicolò
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Michele Longo
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Paola Mirra
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Gregory Alexander Raciti
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Claudia Miele
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Francesco Beguinot
- Department of Translation Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| |
Collapse
|
20
|
Porta M, Amione C, Barutta F, Fornengo P, Merlo S, Gruden G, Albano L, Ciccarelli M, Ungaro P, Durazzo M, Beguinot F, Berchialla P, Cavallo F, Trento M. The co-activator-associated arginine methyltransferase 1 (CARM1) gene is overexpressed in type 2 diabetes. Endocrine 2019; 63:284-292. [PMID: 30173329 DOI: 10.1007/s12020-018-1740-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 08/23/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE We examined the expression of a panel of epigenetic enzymes catalyzing histone tails post-transcriptional modifications, together with effectors of metabolic and inflammatory alterations, in type 2 diabetes. METHODS Cross-sectional, case-control study of 21 people with type 2 diabetes and 21 matched controls. Total RNA was extracted from white cells and reverse transcribed. PCR primer assays for 84 key genes encoding enzymes known to modify genomic DNA and histones were performed. Western blot was performed on lysates using primary antibodies for abnormally expressed enzymes. Hormones and cytokines were measured by multiplex kits. A Bayesian network was built to investigate the relationships between epigenetic, cytokine, and endocrine variables. RESULTS Co-activator-associated aRginine Methyltransferase 1 (CARM1) expression showed a five-fold higher median value, matched by higher protein levels, among patients who also had increased GIP, IL-4, IL-7, IL-13, IL-17, FGF basic, G-CSF, IFN-γ, and TNFα and decreased IP-10. In a Bayesian network approach, CARM1 expression showed a conditional dependence on diabetes, but was independent of all other variables nor appeared to influence any. CONCLUSIONS Increased CARM1 expression in type 2 diabetes suggests that epigenetic mechanisms are altered in human diabetes. The impact of lifestyle and pharmacological treatment on regulation of this enzyme should be further investigated.
Collapse
Affiliation(s)
- Massimo Porta
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy.
| | - Cristina Amione
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| | - Federica Barutta
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| | - Paolo Fornengo
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| | - Stefano Merlo
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| | - Gabriella Gruden
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| | - Luigi Albano
- National Research Council, URT of the Institute of Experimental Endocrinology Oncology "G. Salvatore", Naples, Italy
| | - Marco Ciccarelli
- National Research Council, URT of the Institute of Experimental Endocrinology Oncology "G. Salvatore", Naples, Italy
| | - Paola Ungaro
- National Research Council, URT of the Institute of Experimental Endocrinology Oncology "G. Salvatore", Naples, Italy
| | - Marilena Durazzo
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| | - Francesco Beguinot
- National Research Council, URT of the Institute of Experimental Endocrinology Oncology "G. Salvatore", Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paola Berchialla
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Franco Cavallo
- Department of Public Health and Paediatric Sciences, University of Turin, Turin, Italy
| | - Marina Trento
- Department of Medical Sciences, Laboratory of Clinical Pedagogy, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T, Wang X. A Maternal High-Fat Diet Induces DNA Methylation Changes That Contribute to Glucose Intolerance in Offspring. Front Endocrinol (Lausanne) 2019; 10:871. [PMID: 31920981 PMCID: PMC6923194 DOI: 10.3389/fendo.2019.00871] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
Scope: Overnutrition in utero is a critical contributor to the susceptibility of diabetes by programming, although the exact mechanism is not clear. In this paper, we aimed to study the long-term effect of a maternal high-fat (HF) diet on offspring through epigenetic modifications. Procedures: Five-week-old female C57BL6/J mice were fed a HF diet or control diet for 4 weeks before mating and throughout gestation and lactation. At postnatal week 3, pups continued to consume a HF or switched to a control diet for 5 weeks, resulting in four groups of offspring differing by their maternal and postweaning diets. Results: The maternal HF diet combined with the offspring HF diet caused hyperglycemia and insulin resistance in male pups. Even after changing to the control diet, male pups exposed to the maternal HF diet still exhibited hyperglycemia and glucose intolerance. The livers of pups exposed to a maternal HF diet had a hypermethylated insulin receptor substrate 2 (Irs2) gene and a hypomethylated mitogen-activated protein kinase kinase 4 (Map2k4) gene. Correspondingly, the expression of the Irs2 gene decreased and that of Map2k4 increased in pups exposed to a maternal HF diet. Conclusion: Maternal overnutrition programs long-term epigenetic modifications, namely, Irs2 and Map2k4 gene methylation in the offspring liver, which in turn predisposes the offspring to diabetes later in life.
Collapse
|
22
|
Silveira AC, Dias JP, Santos VM, Oliveira PF, Alves MG, Rato L, Silva BM. The Action of Polyphenols in Diabetes Mellitus and Alzheimer's Disease: A Common Agent for Overlapping Pathologies. Curr Neuropharmacol 2019; 17:590-613. [PMID: 30081787 PMCID: PMC6712293 DOI: 10.2174/1570159x16666180803162059] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/30/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Diabetes Mellitus (DM) and Alzheimer's disease (AD) are two prevalent diseases in modern societies, which are caused mainly by current lifestyle, aging and genetic alterations. It has already been demonstrated that these two diseases are associated, since individuals suffering from DM are prone to develop AD. Conversely, it is also known that individuals with AD are more susceptible to DM, namely type 2 diabetes (T2DM). Therefore, these two pathologies, although completely different in terms of symptomatology, end up sharing several mechanisms at the molecular level, with the most obvious being the increase of oxidative stress and inflammation. Polyphenols are natural compounds widely spread in fruits and vegetables whose dietary intake has been considered inversely proportional to the incidence of DM and AD. So, it is believed that this group of phytochemicals may have preventive and therapeutic potential, not only by reducing the risk and delaying the development of these pathologies, but also by improving brain's metabolic profile and cognitive function. The aim of this review is to understand the extent to which DM and AD are related pathologies, the degree of similarity and the relationship between them, to detail the molecular mechanisms by which polyphenols may exert a protective effect, such as antioxidant and anti-inflammatory effects, and highlight possible advantages of their use as common preventive and therapeutic alternatives.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Branca M. Silva
- Address correspondence to this author at the Faculty of Health Sciences, University of Beira Interior, Av. Infante D.Henrique, 6201-506 Covilhã, Portugal; Tel: +351 275319700; Fax: +351 275 329 183; E-mail:
| |
Collapse
|
23
|
Huang Q, You W, Li Y, Sun Y, Zhou Y, Zhang Y, Liu D, Zhan S, Zhu Y, Han X. Glucolipotoxicity-Inhibited miR-299-5p Regulates Pancreatic β-Cell Function and Survival. Diabetes 2018; 67:2280-2292. [PMID: 30131392 DOI: 10.2337/db18-0223] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/07/2018] [Indexed: 11/13/2022]
Abstract
Inhibition of microRNAs (miRNAs) essential for pancreatic β-cell biology (e.g., miR-375) results in β-cell failure and diabetes in rodent models. Whether the downregulation of miRNAs in pancreatic islets is involved in the development of human type 2 diabetes remains unclear. Here, with the use of an miRNA microarray, we identified a set of miRNAs that were differentially expressed in healthy human islets under glucolipotoxic conditions. A downregulated miRNA, miR-299-5p, was preferentially studied because its inhibition causes dramatic β-cell dysfunction and apoptosis. Proteomic profiling and bioinformatics methods identified four target genes, including a Trp53 effector, Perp, that were further confirmed by luciferase reporter assays. We narrowed down the effector of miR-299-5p downregulation to PERP owing to its upregulation in islets from diabetic rodents. Indeed, Perp inhibition prevented the β-cell impairment caused by either miR-299-5p reduction or glucolipotoxicity. Additional investigations confirmed the modulatory effect of PERP on insulin secretion. Collectively, miR-299-5p appears to be an essential regulator of β-cell biology, and its downregulation links PERP enhancement to β-cell dysfunction and apoptosis in glucolipotoxic settings. Our work demonstrates a novel mechanism of glucolipotoxicity-induced β-cell failure mediated through miR-299-5p downregulation.
Collapse
Affiliation(s)
- Qiqing Huang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiyan You
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory for Neurodegenerative Disease of Jiangsu Province, Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuncai Zhou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dechen Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shanshan Zhan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
24
|
Wang B, Yu J, Wang T, Shen Y, Lin D, Xu X, Wang Y. Identification of megakaryocytes as a target of advanced glycation end products in diabetic complications in bone marrow. Acta Diabetol 2018; 55:419-427. [PMID: 29417230 DOI: 10.1007/s00592-018-1109-z] [Citation(s) in RCA: 3] [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: 12/05/2017] [Accepted: 01/17/2018] [Indexed: 10/18/2022]
Abstract
AIMS To define the possible effect of diabetic conditions on megakaryocytes, the long-know precursors of platelets and lately characterized modulator of hematopoietic stem quiescence-activation transition. METHODS Megakaryoblastic MEG-01 cell culture and TPO/SCF/IL-3-induced differentiation of human umbilical blood mononuclear cells toward megakaryocytes were used to test effects of glycated bovine serum albumin (BSA-AGEs). The ob/ob mice and streptozotocin-treated mice were used as models of hyperglycemia. MTT was used to measure cell proliferation, FACS for surface marker and cell cycle, and RT-qPCR for the expression of interested genes. Megakaryocytes at different stages in marrow smear were checked under microscope. RESULTS When added in MEG-01 cultures at 200 μg/ml, BSA-AGEs increased proliferation of cells and enhanced mRNA expression of RAGE, VEGFα and PF4 in the cells. None of cell cycle distribution, PMA-induced platelet-like particles production, expression of GATA1/NF-E2/PU-1/IL-6/OPG/PDGF in MEG-01 cells nor TPO/SCF/IL-3 induced umbilical cord blood cells differentiation into megakaryocyte was affected by BSA-AGEs. In the ob/ob diabetic mice, MKs percentages in marrow cells and platelets in peripheral blood were significantly increased compared with control mice. In streptozotocin-induced diabetic mice, however, MKs percentage in marrow cells was decreased though peripheral platelet counts were not altered. Gene expression assay showed that the change in MKs in these two diabetic conditions might be explained by the alteration of GATA1 and NF-E2 expression, respectively. CONCLUSIONS Diabetic condition in animals might exert its influence on hematopoiesis via megakaryocytes-the newly identified modulator of hematopoietic stem cells in bone marrow.
Collapse
Affiliation(s)
- Benfang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Jianjiang Yu
- Department of Clinical Laboratory, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin, 214400, China
| | - Ting Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Ying Shen
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Dandan Lin
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China
| | - Xin Xu
- Department of Hematology, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin, 214400, China
| | - Yiqiang Wang
- MOH Key Lab of Thrombosis and Hemostasis, Collaborative Innovation Center of Hematology-Thrombosis and Hemostasis Group, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 708 Renmin Road, Suzhou, 215007, China.
| |
Collapse
|
25
|
Schleger F, Linder K, Walter L, Heni M, Brändle J, Brucker S, Pauluschke-Fröhlich J, Weiss M, Häring HU, Preissl H, Fritsche A. Family History of Diabetes Is Associated With Delayed Fetal Postprandial Brain Activity. Front Endocrinol (Lausanne) 2018; 9:673. [PMID: 30524370 PMCID: PMC6256935 DOI: 10.3389/fendo.2018.00673] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/29/2018] [Indexed: 12/24/2022] Open
Abstract
Introduction: We have previously shown that fetuses of mothers with gestational diabetes mellitus (GDM) and insulin resistance exhibit a prolongation of fetal auditory event-related brain responses (fAER) compared to fetuses of normal glucose tolerant women during an oral glucose tolerance test (oGTT). This implies that maternal metabolism may program the developing fetal brain. We now asked whether a family history of type 2 diabetes without metabolic programing also impacts fetal brain activity. We therefore investigated brain activity in fetuses of normal glucose tolerant mothers with and without family history of type 2 diabetes (FHD+ and FHD-). Methods: A 75 g oGTT was performed in healthy pregnant women. Plasma glucose and insulin levels were measured after 0, 60, and 120 min. Each blood draw was preceded by fetal magnetoencephalographic (fMEG) recordings of fAER. From a group of 167 participants, a subsample of 52 metabolically healthy women, 37 with a negative, and 15 with a positive FHD (at least one first- or second-degree relative) was carefully selected based on the following inclusion criteria: inconspicuous pregnancy, no GDM, BMI 18.5-30 kg/m2, no preterm birth and at least two fMEG with detectable fetal responses during oGTT. Results: An ANOVA showed a significant interaction between fMEG measurement time during the oGTT and FHD on fAER latency [F (2) = 4.163, p = 0.018]. Fetuses of mothers with FHD+ had a prolonged fAER (273 ± 113 ms) compared to fetuses of mothers with FHD- (219 ± 69 ms) at 60 min during the oGTT [F (1) = 4.902, p = 0.032]. There were no significant differences in age, BMI before pregnancy, weight gain during pregnancy and gestational age between the groups. Maternal glucose levels and insulin sensitivity were also not significantly different. Discussion: In addition to the previously shown influence of maternal metabolism on fetal brain activity, maternal family history of diabetes (FHD) is also linked to fetal postprandial brain activity. This indicates that genetic and/or epigenetic factors modulate the postprandial brain response of the developing fetus.
Collapse
Affiliation(s)
- Franziska Schleger
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Katarzyna Linder
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Laura Walter
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Johanna Brändle
- Department of Obstetrics and Gynecology, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Sara Brucker
- Department of Obstetrics and Gynecology, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Jan Pauluschke-Fröhlich
- Department of Obstetrics and Gynecology, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Magdalene Weiss
- Department of Obstetrics and Gynecology, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
- Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls University, Tübingen, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- *Correspondence: Hubert Preissl
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
26
|
Mirra P, Nigro C, Prevenzano I, Leone A, Raciti GA, Formisano P, Beguinot F, Miele C. The Destiny of Glucose from a MicroRNA Perspective. Front Endocrinol (Lausanne) 2018; 9:46. [PMID: 29535681 PMCID: PMC5834423 DOI: 10.3389/fendo.2018.00046] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucose serves as a primary, and for some tissues the unique, fuel source in order to generate and maintain the biological functions. Hyperglycemia is a hallmark of type 2 diabetes and is the direct consequence of perturbations in the glucose homeostasis. Insulin resistance, referred to as a reduced response of target tissues to the hormone, contributes to the development of hyperglycemia. The molecular mechanisms responsible for the altered glucose homeostasis are numerous and not completely understood. MicroRNAs (miRNAs) are now recognized as regulators of the lipid and glucose metabolism and are involved in the onset of metabolic diseases. Indeed, these small non-coding RNA molecules operate in the RNA silencing and posttranscriptional regulation of gene expression and may modulate the levels of kinases and enzymes in the glucose metabolism. Therefore, a better characterization of the function of miRNAs and a deeper understanding of their role in disease may represent a fundamental step toward innovative treatments addressing the causes, not only the symptoms, of hyperglycemia, using approaches aimed at restoring either miRNAs or their specific targets. In this review, we outline the current understanding regarding the impact of miRNAs in the glucose metabolism and highlight the need for further research focused on altered key kinases and enzymes in metabolic diseases.
Collapse
Affiliation(s)
- Paola Mirra
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Cecilia Nigro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Immacolata Prevenzano
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessia Leone
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Gregory Alexander Raciti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Francesco Beguinot
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Claudia Miele
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- *Correspondence: Claudia Miele,
| |
Collapse
|
27
|
Raciti GA, Spinelli R, Desiderio A, Longo M, Parrillo L, Nigro C, D'Esposito V, Mirra P, Fiory F, Pilone V, Forestieri P, Formisano P, Pastan I, Miele C, Beguinot F. Specific CpG hyper-methylation leads to Ankrd26 gene down-regulation in white adipose tissue of a mouse model of diet-induced obesity. Sci Rep 2017; 7:43526. [PMID: 28266632 PMCID: PMC5339897 DOI: 10.1038/srep43526] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/27/2017] [Indexed: 12/16/2022] Open
Abstract
Epigenetic modifications alter transcriptional activity and contribute to the effects of environment on the individual risk of obesity and Type 2 Diabetes (T2D). Here, we have estimated the in vivo effect of a fat-enriched diet (HFD) on the expression and the epigenetic regulation of the Ankyrin repeat domain 26 (Ankrd26) gene, which is associated with the onset of these disorders. In visceral adipose tissue (VAT), HFD exposure determined a specific hyper-methylation of Ankrd26 promoter at the −436 and −431 bp CpG sites (CpGs) and impaired its expression. Methylation of these 2 CpGs impaired binding of the histone acetyltransferase/transcriptional coactivator p300 to this same region, causing hypo-acetylation of histone H4 at the Ankrd26 promoter and loss of binding of RNA Pol II at the Ankrd26 Transcription Start Site (TSS). In addition, HFD increased binding of DNA methyl-transferases (DNMTs) 3a and 3b and methyl-CpG-binding domain protein 2 (MBD2) to the Ankrd26 promoter. More importantly, Ankrd26 down-regulation enhanced secretion of pro-inflammatory mediators by 3T3-L1 adipocytes as well as in human sera. Thus, in mice, the exposure to HFD induces epigenetic silencing of the Ankrd26 gene, which contributes to the adipose tissue inflammatory secretion profile induced by high-fat regimens.
Collapse
Affiliation(s)
- Gregory A Raciti
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Rosa Spinelli
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Antonella Desiderio
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Michele Longo
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Luca Parrillo
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Cecilia Nigro
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Vittoria D'Esposito
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Paola Mirra
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesca Fiory
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Vincenzo Pilone
- Bariatric and Metabolic Surgery Unit, University of Salerno, Salerno, 84084, Italy
| | - Pietro Forestieri
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Pietro Formisano
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Ira Pastan
- Laboratory of Molecular Biology (LMB), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Claudia Miele
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesco Beguinot
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|
28
|
Sala P, de Miranda Torrinhas RSM, Fonseca DC, Ravacci GR, Waitzberg DL, Giannella-Neto D. Tissue-specific methylation profile in obese patients with type 2 diabetes before and after Roux-en-Y gastric bypass. Diabetol Metab Syndr 2017; 9:15. [PMID: 28250848 PMCID: PMC5322591 DOI: 10.1186/s13098-017-0214-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/14/2017] [Indexed: 12/19/2022] Open
Abstract
Eating habits, lifestyles, and exposure to specific environmental factors can greatly impact the risk of developing type 2 diabetes (T2D), influence the genome epigenetically, and affect the expression of genes, including genes related to glycemic control, at any stage of life. The epigenetic mechanism underlying obesity and T2D pathogenesis remains poorly understood. Conventional strategies for the treatment of obesity and its comorbidities often have poor long-term adherence, and pharmacological interventions are limited. Bariatric surgery is the most effective current option to treat severe obesity, and Roux-en-Y gastric bypass (RYGB) is the most applied technique worldwide. Epigenetic changes differ depending on the approach used to treat obesity and its associated comorbidities (clinical or surgical). Compared to primary clinical care, bariatric surgery leads to much greater loss of body weight and higher remission rates of T2D and metabolic syndrome, with methylation profiles in promoter regions of genes in obese individuals becoming similar to those of normal-weight individuals. Bariatric surgery can influence DNA methylation in parallel with changes in gene expression pattern. Changes in clinical biomarkers that reflect improvements in glucose and lipid metabolism after RYGB often occur before major weight loss and are coordinated by surgery-induced changes in intestinal hormones. Therefore, the intestine methylation profile would assist in understanding the mechanisms involved in improved glycemic control after bariatric surgery. The main objectives in this area for the future are to identify epigenetic marks that could be used as early indicators of metabolic risk, and to develop treatments able to delay or even reverse these epigenetic changes. Studies that provide the "human epigenetic profile" will be of considerable value to identify tissue-specific epigenetic signatures and their role in the development of chronic diseases. Further studies should apply methods based on global analysis of the genome to identify methylated sites associated with disease and epigenetic marks associated with the remodeling response to bariatric surgery. This review describes the main epigenetic alterations associated with obesity and T2D and the potential role of RYGB in remodeling these changes.
Collapse
Affiliation(s)
- Priscila Sala
- FMUSP—Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, University of São Paulo (LIM 35), São Paulo, Brazil
| | | | - Danielle Cristina Fonseca
- FMUSP—Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, University of São Paulo (LIM 35), São Paulo, Brazil
| | - Graziela Rosa Ravacci
- FMUSP—Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, University of São Paulo (LIM 35), São Paulo, Brazil
| | - Dan Linetzky Waitzberg
- FMUSP—Department of Gastroenterology, Digestive Surgery Discipline, School of Medicine, University of São Paulo (LIM 35), São Paulo, Brazil
| | | |
Collapse
|
29
|
Olivieri F, Spazzafumo L, Bonafè M, Recchioni R, Prattichizzo F, Marcheselli F, Micolucci L, Mensà E, Giuliani A, Santini G, Gobbi M, Lazzarini R, Boemi M, Testa R, Antonicelli R, Procopio AD, Bonfigli AR. MiR-21-5p and miR-126a-3p levels in plasma and circulating angiogenic cells: relationship with type 2 diabetes complications. Oncotarget 2016; 6:35372-82. [PMID: 26498351 PMCID: PMC4742111 DOI: 10.18632/oncotarget.6164] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/02/2015] [Indexed: 01/15/2023] Open
Abstract
Innovative biomarkers are required to manage type 2 diabetic patients (T2DM). We focused our study on miR-126-3p and miR-21-5p levels, as biomarkers of endothelial function and inflammation. MiRNAs levels were measured in plasma from 107 healthy subjects (CTR) and 193 diabetic patients (T2DM), 76 without (T2DM NC) and 117 with (T2DM C) complications. When diabetic complication were analysed as a whole, miR-126-3p and miR-21-5p levels declined significantly from CTR to T2DM NC and T2DM C patients. When miRNAs levels were related to specific complications, significantly higher miR-21-5p levels (0.46 ± 0.44 vs. 0.26±0.33, p < 0.001) and significant lower miR-126-3p levels (0.21±0.21 vs. 0.28±0.22, p = 0.032) were found in T2DM with previous major cardiovascular events (MACE) vs. all the others T2DM patients. To confirm these results we focused on circulating angiogenic cells (CACs) from a subgroup of 10 CTR, 15 T2DM NC and 15 T2DM patients with MACE. CACs from T2DM patients expressed higher miR-21-5p and lower miR-126-3p levels than CACs from CTR. Furthermore, CACs from T2DM + MACE showed the highest levels of miR-21-5p. Circulating miR-21-5p and miR-126-3p emerge as dynamic biomarkers of systemic inflammatory/angiogenic status. Their expression levels in CACs from T2DM with MACE suggest a shift from a proangiogenic to a proinflammatory profile.
Collapse
Affiliation(s)
- Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - Liana Spazzafumo
- Center of Biostatistics, INRCA-IRCCS National Institute, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Bologna, Italy
| | - Rina Recchioni
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - Francesco Prattichizzo
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Fiorella Marcheselli
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - Luigina Micolucci
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Emanuela Mensà
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Gabriele Santini
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Mirko Gobbi
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Raffaella Lazzarini
- Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | - Massimo Boemi
- Metabolic Diseases and Diabetology Unit, INRCA-IRCCS National Institute, Ancona, Italy
| | - Roberto Testa
- Experimental Models in Clinical Pathology, INRCA-IRCCS National Institute, Ancona, Italy
| | | | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | | |
Collapse
|
30
|
Desiderio A, Spinelli R, Ciccarelli M, Nigro C, Miele C, Beguinot F, Raciti GA. Epigenetics: spotlight on type 2 diabetes and obesity. J Endocrinol Invest 2016; 39:1095-103. [PMID: 27180180 DOI: 10.1007/s40618-016-0473-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes (T2D) and obesity are the major public health problems. Substantial efforts have been made to define loci and variants contributing to the individual risk of these disorders. However, the overall risk explained by genetic variation is very modest. Epigenetics is one of the fastest growing research areas in biomedicine as changes in the epigenome are involved in many biological processes, impact on the risk for several complex diseases including diabetes and may explain susceptibility. In this review, we focus on the role of DNA methylation in contributing to the risk of T2D and obesity.
Collapse
Affiliation(s)
- A Desiderio
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - R Spinelli
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - M Ciccarelli
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - C Nigro
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - C Miele
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - F Beguinot
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy.
| | - G A Raciti
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| |
Collapse
|
31
|
Yan S, Wang T, Huang S, Di Y, Huang Y, Liu X, Luo Z, Han W, An B. Differential expression of microRNAs in plasma of patients with prediabetes and newly diagnosed type 2 diabetes. Acta Diabetol 2016; 53:693-702. [PMID: 27039347 DOI: 10.1007/s00592-016-0837-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 01/09/2016] [Indexed: 02/06/2023]
Abstract
AIMS MicroRNAs (miRNAs) are present in plasma and have emerged as critical regulators of gene expression at posttranscriptional level, and thus are involved in various human diseases, including diabetes. The objective of this study was to screen and validate differentially expressed plasma miRNAs in prediabetes and newly diagnosed type 2 diabetes (T2D). METHODS In this study, we screened differentially expressed plasma miRNAs in prediabetes and newly diagnosed T2D by miRNA microarray analysis, and validated the expression of candidate miRNAs using quantitative reverse transcription polymerase chain reaction assays. Furthermore, we performed gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway analyses to disclose functional enrichment of genes predicted to be regulated by the differentially expressed miRNAs. RESULTS Notably, our results revealed that hsa-miR-1249, hsa-miR-320b, and hsa-miR-572 (P < 0.05) were differentially expressed among the three groups, which yielded an area under the receiver operator characteristics curve (AUC) of 0.784 [95 % confidence interval (CI) 0.685-0.883], 0.946 (95 % CI 0.906-0.985), and 0.843 (95 % CI 0.766-0.920) discriminating T2D patients from NGT control groups, respectively, while the AUC was 0.887 (95 % CI 0.818-0.957), 0.635 (95 % CI 0.525-0.744), and 0.69 (95 % CI 0.580-0.793) discriminating prediabetes patients from NGT control groups, respectively. In addition, GO and KEGG pathway analyses showed that genes predicted to be regulated by differentially expressed miRNAs were significantly enriched in several related biological processes and pathways, including the development of multicellular organisms, signal transduction, cell differentiation, apoptosis, cell metabolism, ion transport regulation, and other biological functions. CONCLUSIONS Taken together, our results showed differentially expressed miRNAs in T2D and prediabetes. Plasma hsa-miR-1249, hsa-miR-320b, and hsa-miR-572 may serve as novel biomarkers for diagnosis and potential targets for the treatment for prediabetes and T2D.
Collapse
Affiliation(s)
- Shaoying Yan
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou Province, People's Republic of China
| | - Tianqiong Wang
- Department of Clinical Laboratory, People's Hospital of Guizhou Province, Guiyang, Guizhou Province, People's Republic of China
| | - Shengwen Huang
- Department of Clinical Laboratory, People's Hospital of Guizhou Province, Guiyang, Guizhou Province, People's Republic of China
| | - Yanan Di
- Department of Clinical Laboratory, People's Hospital of Guizhou Province, Guiyang, Guizhou Province, People's Republic of China
| | - Yunzhu Huang
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou Province, People's Republic of China
| | - Xingmei Liu
- Department of Clinical Laboratory, People's Hospital of Guizhou Province, Guiyang, Guizhou Province, People's Republic of China
| | - Zhenyuan Luo
- Department of Clinical Laboratory, People's Hospital of Guizhou Province, Guiyang, Guizhou Province, People's Republic of China
| | - Wenping Han
- Department of Medical Laboratory, Guizhou Medical University, Guiyang, Guizhou Province, People's Republic of China
| | - Bangquan An
- Department of Clinical Laboratory, People's Hospital of Guizhou Province, Guiyang, Guizhou Province, People's Republic of China.
| |
Collapse
|
32
|
Khan S, Kumar S, Jena G. Valproic acid reduces insulin-resistance, fat deposition and FOXO1-mediated gluconeogenesis in type-2 diabetic rat. Biochimie 2016; 125:42-52. [DOI: 10.1016/j.biochi.2016.02.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 02/29/2016] [Indexed: 10/22/2022]
|
33
|
Lahiri DK, Maloney B, Bayon BL, Chopra N, White FA, Greig NH, Nurnberger JI. Transgenerational latent early-life associated regulation unites environment and genetics across generations. Epigenomics 2016; 8:373-87. [PMID: 26950428 DOI: 10.2217/epi.15.117] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The origin of idiopathic diseases is still poorly understood. The latent early-life associated regulation (LEARn) model unites environmental exposures and gene expression while providing a mechanistic underpinning for later-occurring disorders. We propose that this process can occur across generations via transgenerational LEARn (tLEARn). In tLEARn, each person is a 'unit' accumulating preclinical or subclinical 'hits' as in the original LEARn model. These changes can then be epigenomically passed along to offspring. Transgenerational accumulation of 'hits' determines a sporadic disease state. Few significant transgenerational hits would accompany conception or gestation of most people, but these may suffice to 'prime' someone to respond to later-life hits. Hits need not produce symptoms or microphenotypes to have a transgenerational effect. Testing tLEARn requires longitudinal approaches. A recently proposed longitudinal epigenome/envirome-wide association study would unite genetic sequence, epigenomic markers, environmental exposures, patient personal history taken at multiple time points and family history.
Collapse
Affiliation(s)
- Debomoy K Lahiri
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA.,Department of Medical & Molecular Genetics, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Bryan Maloney
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Baindu L Bayon
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Nipun Chopra
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Fletcher A White
- Department of Anesthesia, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Nigel H Greig
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - John I Nurnberger
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA.,Department of Medical & Molecular Genetics, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN 46202, USA
| |
Collapse
|
34
|
Zhao X, Guo L, Lin Y, Wang H, Gu C, Zhao L, Tong X. The top 100 most cited scientific reports focused on diabetes research. Acta Diabetol 2016; 53:13-26. [PMID: 26596851 DOI: 10.1007/s00592-015-0813-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/24/2015] [Indexed: 01/17/2023]
Abstract
AIMS The number of citations that a paper has received reflects the impact of the article within a particular medical research area. However, which articles are cited most frequently in diabetes research is unknown. We aimed to identify the most frequently cited 100 articles that describe advances focused on diabetes research. METHODS Science Citation Index Expanded database was used to determine the 100 most cited articles in diabetes research. The key characteristics of the top 100 cited articles, including citation ranking, year of publication, publishing journal, type of study, h-index, PMC citation, patent citation, country of origin, funding source, and authorship, were analyzed. RESULTS The number of citations ranged from 10292 to 1121. The 100 most cited articles were published between 1962 and 2010. New England Journal of Medicine published the largest number of the most cited articles (n = 23), followed by Diabetes Care (n = 14) and Nature (n = 12). The greatest number of contributions came from the USA, followed by the UK and Canada. RCT was the most popular article type, followed by basic science studies and observational studies. There was a positive correlation between total citations and h-indexes and PMC citations, while there was no correlation with patent citations. CONCLUSIONS The most influential report in diabetes research seems to be RCT, which is performed to confirm the effectiveness and outcome of anti-diabetes drugs in patients with diabetes. This study presents insight into the historical development of diabetes research, and provides foundations for further investigations.
Collapse
Affiliation(s)
- Xiyan Zhao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, XiCheng District, Beijing, 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liangqing Guo
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong Province, China
| | - Yiqun Lin
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, XiCheng District, Beijing, 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Han Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, XiCheng District, Beijing, 100053, China
| | - Chengjuan Gu
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, XiCheng District, Beijing, 100053, China
| | - Linhua Zhao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, XiCheng District, Beijing, 100053, China.
| | - Xiaolin Tong
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, XiCheng District, Beijing, 100053, China.
| |
Collapse
|
35
|
Schwartz SS, Epstein S, Corkey BE, Grant SFA, Gavin JR, Aguilar RB. The Time Is Right for a New Classification System for Diabetes: Rationale and Implications of the β-Cell-Centric Classification Schema. Diabetes Care 2016; 39:179-86. [PMID: 26798148 PMCID: PMC5317235 DOI: 10.2337/dc15-1585] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The current classification system presents challenges to the diagnosis and treatment of patients with diabetes mellitus (DM), in part due to its conflicting and confounding definitions of type 1 DM, type 2 DM, and latent autoimmune diabetes of adults (LADA). The current schema also lacks a foundation that readily incorporates advances in our understanding of the disease and its treatment. For appropriate and coherent therapy, we propose an alternate classification system. The β-cell-centric classification of DM is a new approach that obviates the inherent and unintended confusions of the current system. The β-cell-centric model presupposes that all DM originates from a final common denominator-the abnormal pancreatic β-cell. It recognizes that interactions between genetically predisposed β-cells with a number of factors, including insulin resistance (IR), susceptibility to environmental influences, and immune dysregulation/inflammation, lead to the range of hyperglycemic phenotypes within the spectrum of DM. Individually or in concert, and often self-perpetuating, these factors contribute to β-cell stress, dysfunction, or loss through at least 11 distinct pathways. Available, yet underutilized, treatments provide rational choices for personalized therapies that target the individual mediating pathways of hyperglycemia at work in any given patient, without the risk of drug-related hypoglycemia or weight gain or imposing further burden on the β-cells. This article issues an urgent call for the review of the current DM classification system toward the consensus on a new, more useful system.
Collapse
Affiliation(s)
- Stanley S Schwartz
- Main Line Health, Wynnewood, PA, and University of Pennsylvania, Philadelphia, PA
| | - Solomon Epstein
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Mount Sinai Hospital, New York, NY
| | - Barbara E Corkey
- Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Struan F A Grant
- Division of Human Genetics and Center for Applied Genomics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | |
Collapse
|
36
|
Abstract
Type 1 diabetes (T1D) is a multifactorial disease resulting from an immune-mediated destruction of the insulin-producing pancreatic β cells. Several environmental and genetic risk factors predispose to the disease. Genome-wide association studies (GWAS) have identified around 50 genetic regions that affect the risk of developing T1D, but the disease-causing variants and genes are still largely unknown. In this review, we discuss the current status of T1D susceptibility loci and candidate genes with focus on the β cell. At least 40 % of the genes in the T1D susceptibility loci are expressed in human islets and β cells, where they according to recent studies modulate the β-cell response to the immune system. As most of the risk variants map to noncoding regions of the genome, i.e., promoters, enhancers, intergenic regions, and noncoding genes, their possible involvement in T1D pathogenesis as gene regulators will also be addressed.
Collapse
Affiliation(s)
- Tina Fløyel
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.
| | - Simranjeet Kaur
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.
| | - Flemming Pociot
- Copenhagen Diabetes Research Center, Department of Pediatrics, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730, Herlev, Denmark.
| |
Collapse
|
37
|
Hao K, Di Narzo AF, Ho L, Luo W, Li S, Chen R, Li T, Dubner L, Pasinetti GM. Shared genetic etiology underlying Alzheimer's disease and type 2 diabetes. Mol Aspects Med 2015; 43-44:66-76. [PMID: 26116273 DOI: 10.1016/j.mam.2015.06.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 06/12/2015] [Indexed: 12/12/2022]
Abstract
Epidemiological evidence supports the observation that subjects with type 2 diabetes (T2D) are at higher risk to develop Alzheimer's disease (AD). However, whether and how these two conditions are causally linked is unknown. Possible mechanisms include shared genetic risk factors, which were investigated in this study based on recent genome wide association study (GWAS) findings. In order to achieve our goal, we retrieved single nucleotide polymorphisms (SNPs) associated with T2D and AD from large-scale GWAS meta-analysis consortia and tested for overlap among the T2D- and AD-associated SNPs at various p-value thresholds. We then explored the function of the shared T2D/AD GWAS SNPs by leveraging expressional quantitative trait loci, pathways, gene ontology data, and co-expression networks. We found 927 SNPs associated with both AD and T2D with p-value ≤0.01, an overlap significantly larger than random chance (overlapping p-value of 6.93E-28). Among these, 395 of the shared GWAS SNPs have the same risk allele for AD and T2D, suggesting common pathogenic mechanisms underlying the development of both AD and T2D. Genes influenced by shared T2D/AD SNPs with the same risk allele were first identified using a SNP annotation variation (ANNOVAR) software, followed by using Association Protein-Protein Link Evaluator (DAPPLE) software to identify additional proteins that are known to physically interact with the ANNOVAR gene annotations. We found that gene annotations from ANNOVAR and DAPPLE significantly enriched specific KEGG pathways pertaining to immune responses, cell signaling and neuronal plasticity, cellular processes in which abnormalities are known to contribute to both T2D and AD pathogenesis. Thus, our observation suggests that among T2D subjects with common genetic predispositions (e.g., SNPs with consistent risk alleles for T2D and AD), dysregulation of these pathogenic pathways could contribute to the elevated risks for AD in subjects. Interestingly, we found that 532 of the shared T2D/AD GWAS SNPs had divergent risk alleles in the two diseases. For individual shared T2D/AD SNPs with divergent alleles, one of the allelic forms may contribute to one of the diseases (e.g., T2D), but not necessarily to the other (e.g., AD), or vice versa. Collectively, our GWAS studies tentatively support the epidemiological observation of disease concordance between T2D and AD. Moreover, the studies provide the much needed information for the design of future novel therapeutic approaches, for a subpopulation of T2D subjects with genetic disposition to AD, that could benefit T2D and reduce the risk for subsequent development of AD.
Collapse
Affiliation(s)
- Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Antonio Fabio Di Narzo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Lap Ho
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Wei Luo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA; College of Computer Science and Technology, Huaqiao University, No.668 Jimei Avenue, Xiamen 361021, China
| | - Shuyu Li
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Rong Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Tongbin Li
- AccuraScience, LLC, 5721 Merle Hay Road, Johnston, IA, USA
| | - Lauren Dubner
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA; Geriatric Research, Education and Clinical Center (GRECC), James J. Peters Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, USA.
| |
Collapse
|