1
|
Svyatova G, Berezina G, Murtazaliyeva A, Dyussupov A, Belyayeva T, Faizova R, Dyussupova A. Genetic Predisposition to Prediabetes in the Kazakh Population. Curr Issues Mol Biol 2024; 46:10913-10922. [PMID: 39451528 PMCID: PMC11505754 DOI: 10.3390/cimb46100648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
The aim of this study was to conduct a comparative analysis of the population frequencies of the minor allele of polymorphic variants in the genes TCF7L2 (rs7903146) and PPARG (rs1801282), based on the genome-wide association studies analysis data associated with the risk of developing prediabetes, in an ethnically homogeneous Kazakh population compared to previously studied populations worldwide. This study utilized a genomic database consisting of 1800 ethnically Kazakh individuals who were considered in healthy condition. Whole-genome genotyping was performed using Illumina OmniChip 2.5-8 arrays, which interrogated approximately 2.5 million single nucleotide polymorphisms. The distribution of genotypes for the TCF7L2 (rs7903146) and PPARG (rs1801282) polymorphisms in the Kazakh sample was found to be in Hardy-Weinberg equilibrium (p > 0.05). The minor G allele of the "Asian" protective polymorphism rs1801282 in the PPARG gene was observed at a frequency of 13.8% in the Kazakh population. This suggests a potentially more significant protective effect of this polymorphism in reducing the risk of prediabetes among Kazakhs. The frequency of the unfavorable T allele of the insulin secretion-disrupting gene TCF7L2 (rs7903146) in Kazakhs was 15.2%. Studying the associations of genetic markers for prediabetes enables the timely identification of "high-risk groups" and facilitates the implementation of effective preventive measures. Further results from replicative genomic research will help identify significant polymorphic variants of genes underlying the alteration of prediabetes status.
Collapse
Affiliation(s)
- Gulnara Svyatova
- Laboratory of Republican Medical Genetic Consultation, Scientific Center of Obstetrics, Gynecology, and Perinatology, Almaty 050020, Kazakhstan; (G.S.); (G.B.); (A.M.)
| | - Galina Berezina
- Laboratory of Republican Medical Genetic Consultation, Scientific Center of Obstetrics, Gynecology, and Perinatology, Almaty 050020, Kazakhstan; (G.S.); (G.B.); (A.M.)
| | - Alexandra Murtazaliyeva
- Laboratory of Republican Medical Genetic Consultation, Scientific Center of Obstetrics, Gynecology, and Perinatology, Almaty 050020, Kazakhstan; (G.S.); (G.B.); (A.M.)
| | - Altay Dyussupov
- Department of General Medical Practice, Semey Medical University, Semey 071400, Kazakhstan; (A.D.); (T.B.); (R.F.)
| | - Tatyana Belyayeva
- Department of General Medical Practice, Semey Medical University, Semey 071400, Kazakhstan; (A.D.); (T.B.); (R.F.)
| | - Raida Faizova
- Department of General Medical Practice, Semey Medical University, Semey 071400, Kazakhstan; (A.D.); (T.B.); (R.F.)
| | - Azhar Dyussupova
- Department of General Medical Practice, Semey Medical University, Semey 071400, Kazakhstan; (A.D.); (T.B.); (R.F.)
| |
Collapse
|
2
|
Carullo N, Zicarelli M, Michael A, Faga T, Battaglia Y, Pisani A, Perticone M, Costa D, Ielapi N, Coppolino G, Bolignano D, Serra R, Andreucci M. Childhood Obesity: Insight into Kidney Involvement. Int J Mol Sci 2023; 24:17400. [PMID: 38139229 PMCID: PMC10743690 DOI: 10.3390/ijms242417400] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
This review examines the impact of childhood obesity on the kidney from an epidemiological, pathogenetic, clinical, and pathological perspective, with the aim of providing pediatricians and nephrologists with the most current data on this topic. The prevalence of childhood obesity and chronic kidney disease (CKD) is steadily increasing worldwide, reaching epidemic proportions. While the impact of obesity in children with CKD is less pronounced than in adults, recent studies suggest a similar trend in the child population. This is likely due to the significant association between obesity and the two leading causes of end-stage renal disease (ESRD): diabetes mellitus (DM) and hypertension. Obesity is a complex, systemic disease that reflects interactions between environmental and genetic factors. A key mechanism of kidney damage is related to metabolic syndrome and insulin resistance. Therefore, we can speculate about an adipose tissue-kidney axis in which neurohormonal and immunological mechanisms exacerbate complications resulting from obesity. Adipose tissue, now recognized as an endocrine organ, secretes cytokines called adipokines that may induce adaptive or maladaptive responses in renal cells, leading to kidney fibrosis. The impact of obesity on kidney transplant-related outcomes for both donors and recipients is also significant, making stringent preventive measures critical in the pre- and post-transplant phases. The challenge lies in identifying renal involvement as early as possible, as it is often completely asymptomatic and not detectable through common markers of kidney function. Ongoing research into innovative technologies, such as proteomics and metabolomics, aims to identify new biomarkers and is constantly evolving. Many aspects of pediatric disease progression in the population of children with obesity still require clarification. However, the latest scientific evidence in the field of nephrology offers glimpses into various new perspectives, such as genetic factors, comorbidities, and novel biomarkers. Investigating these aspects early could potentially improve the prognosis of these young patients through new diagnostic and therapeutic strategies. Hence, the aim of this review is to provide a comprehensive exploration of the pathogenetic mechanisms and prevalent pathological patterns of kidney damage observed in children with obesity.
Collapse
Affiliation(s)
- Nazareno Carullo
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (N.C.); (M.Z.); (A.M.); (T.F.); (G.C.)
| | - Mariateresa Zicarelli
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (N.C.); (M.Z.); (A.M.); (T.F.); (G.C.)
| | - Ashour Michael
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (N.C.); (M.Z.); (A.M.); (T.F.); (G.C.)
| | - Teresa Faga
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (N.C.); (M.Z.); (A.M.); (T.F.); (G.C.)
| | - Yuri Battaglia
- Department of Medicine, University of Verona, 37129 Verona, Italy;
| | - Antonio Pisani
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy;
| | - Maria Perticone
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.P.); (D.C.); (D.B.)
| | - Davide Costa
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.P.); (D.C.); (D.B.)
- Interuniversity Center of Phlebolymphology (CIFL), “Magna Graecia” University, 88100 Catanzaro, Italy;
| | - Nicola Ielapi
- Interuniversity Center of Phlebolymphology (CIFL), “Magna Graecia” University, 88100 Catanzaro, Italy;
- Department of Public Health and Infectious Disease, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Giuseppe Coppolino
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (N.C.); (M.Z.); (A.M.); (T.F.); (G.C.)
| | - Davide Bolignano
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.P.); (D.C.); (D.B.)
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (M.P.); (D.C.); (D.B.)
- Interuniversity Center of Phlebolymphology (CIFL), “Magna Graecia” University, 88100 Catanzaro, Italy;
| | - Michele Andreucci
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (N.C.); (M.Z.); (A.M.); (T.F.); (G.C.)
| |
Collapse
|
3
|
Wang G, Chiou J, Zeng C, Miller M, Matta I, Han JY, Kadakia N, Okino ML, Beebe E, Mallick M, Camunas-Soler J, Dos Santos T, Dai XQ, Ellis C, Hang Y, Kim SK, MacDonald PE, Kandeel FR, Preissl S, Gaulton KJ, Sander M. Integrating genetics with single-cell multiomic measurements across disease states identifies mechanisms of beta cell dysfunction in type 2 diabetes. Nat Genet 2023; 55:984-994. [PMID: 37231096 PMCID: PMC10550816 DOI: 10.1038/s41588-023-01397-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 04/12/2023] [Indexed: 05/27/2023]
Abstract
Dysfunctional pancreatic islet beta cells are a hallmark of type 2 diabetes (T2D), but a comprehensive understanding of the underlying mechanisms, including gene dysregulation, is lacking. Here we integrate information from measurements of chromatin accessibility, gene expression and function in single beta cells with genetic association data to nominate disease-causal gene regulatory changes in T2D. Using machine learning on chromatin accessibility data from 34 nondiabetic, pre-T2D and T2D donors, we identify two transcriptionally and functionally distinct beta cell subtypes that undergo an abundance shift during T2D progression. Subtype-defining accessible chromatin is enriched for T2D risk variants, suggesting a causal contribution of subtype identity to T2D. Both beta cell subtypes exhibit activation of a stress-response transcriptional program and functional impairment in T2D, which is probably induced by the T2D-associated metabolic environment. Our findings demonstrate the power of multimodal single-cell measurements combined with machine learning for characterizing mechanisms of complex diseases.
Collapse
Affiliation(s)
- Gaowei Wang
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Joshua Chiou
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
- Biomedical Graduate Studies Program, University of California San Diego, La Jolla, CA, USA
| | - Chun Zeng
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Michael Miller
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Ileana Matta
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Jee Yun Han
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Nikita Kadakia
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Mei-Lin Okino
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Elisha Beebe
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Medhavi Mallick
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | | | - Theodore Dos Santos
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Xiao-Qing Dai
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Cara Ellis
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Medicine and of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Medicine and of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Fouad R Kandeel
- Department of Clinical Diabetes, Endocrinology & Metabolism, City of Hope, Duarte, CA, USA
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA.
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Kyle J Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Maike Sander
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
4
|
Wang D, Liu L, Zhang C, Lu W, Wu F, He X. Evaluation of Association Studies and Meta-Analyses of eNOS Polymorphisms in Type 2 Diabetes Mellitus Risk. Front Genet 2022; 13:887415. [PMID: 35832187 PMCID: PMC9271911 DOI: 10.3389/fgene.2022.887415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Numerous studies reported the associations between endothelial nitric oxide synthase (eNOS) polymorphisms (4b/a VNTR (rs869109213), G894T (rs1799983) and T786C (rs2070744)) and type 2 diabetes mellitus (T2DM) risk. However, the conclusions were incongruent. Moreover, since no published meta-analyses were performed, a key issue regarding false-positive results needs to be addressed. Furthermore, four new articles have been published on these issues. Therefore, an updated meta-analysis was conducted to further explore these associations. Objectives: To investigate the association between eNOS 4b/a, G894T and T786C polymorphisms and T2DM risk. Methods: Studies were searched by using the PubMed, China National Knowledge Infrastructure (CNKI), Medline, Embase, International Statistical Institute (ISI) and the China Wanfang databases. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to evaluate the associations using five genetic models. Furthermore, the false-positive report probability (FPRP), Bayesian false discovery probability (BFDP), and the Venice criteria were employed to assess the credibility of statistically significant associations. Results: Overall, the eNOS 4b/a polymorphism was associated with a significantly decreased T2DM risk in Asians (bb vs. aa: OR = 0.44, 95% CI = 0.23–0.84; ab + bb vs. aa: OR = 0.45, 95% CI = 0.24–0.86; bb vs. aa + ab: OR = 0.73, 95% CI = 0.59–0.91; b vs. a: OR = 0.71, 95% CI = 0.57–0.88); the eNOS G894T polymorphism was associated with a significantly increased T2DM risk in Asians (GT vs. GG: OR = 1.52, 95% CI = 1.15–2.01; GT + TT vs. GG: OR = 1.52, 95% CI = 1.15–2.01; T vs. G: OR = 1.39, 95% CI = 1.09–1.76); the eNOS T786C polymorphism was associated with a significantly increased T2DM risk in Indian (TC vs. TT: OR = 1.93, 95% CI = 1.27–2.94; TC + CC vs. TT: OR = 2.06, 95%CI = 1.26–3.36; C vs. T: OR = 1.90, 95%CI = 1.17–3.08). However, when a sensitivity analysis was performed after excluding low quality and Hardy–Weinberg Disequilibrium (HWD) studies, no significant association was found for the eNOS G894T polymorphism. After credibility assessment, we identified “less-credible positive results” for the statistically significant associations in the current meta-analysis. Conclusion: In conclusion, this article suggests that all substantial relationships between eNOS 4b/a, G894T, and T786C polymorphisms and T2DM risk are most likely due to false positive results rather than real connections or biological variables.
Collapse
Affiliation(s)
- Di Wang
- Changzhi Medical College, Changzhi, China
| | | | | | - Wensheng Lu
- Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- *Correspondence: Wensheng Lu, ; Feifei Wu, ; Xiaofeng He,
| | - Feifei Wu
- Department of Endocrinology and Metabolism, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
- *Correspondence: Wensheng Lu, ; Feifei Wu, ; Xiaofeng He,
| | - Xiaofeng He
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
- Institute of Evidence-Based Medicine, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
- *Correspondence: Wensheng Lu, ; Feifei Wu, ; Xiaofeng He,
| |
Collapse
|
5
|
Giha HA, AlDehaini DMB, Joatar FE, Ali ME, Al-Harbi EM, Al Qarni AA. Hormonal and metabolic profiles of obese and nonobese type 2 diabetes patients: implications of plasma insulin, ghrelin, and vitamin D levels. Cardiovasc Endocrinol Metab 2022; 11:e0256. [PMID: 35098041 PMCID: PMC8789207 DOI: 10.1097/xce.0000000000000256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022]
Abstract
UNLABELLED Type 2 diabetes (T2D) is associated with obesity whereas loss of weight is a feature of the disease; however, the two states are not mutually exclusive. Obesity is linked with changes in hormonal activity and overall body metabolism. MATERIALS AND METHODS In this study, 408 T2D patients were recruited in three distinct studies conducted in Bahrain, Saudi Arabia, and Kuwait in three different intervals between 2001 and 2019. In addition to demographics, glycemic and lipid profiles were obtained in all studies, whereas plasma insulin and HOMA-IR, vitamin D, and ghrelin were analyzed in Saudi Arabia. Different techniques such as chemical auto-analyzer, ELISA, chemiluminescent immunoassay, radioimmunoassay were used. RESULTS The obese (BMI ≥ 30 kg/m2) compared with nonobese (BMI 18.5 to <30) patients with diabetes were more likely to be women (P < 0.001), smaller in age (P = 0.028), and with shorter disease duration (P = 0.018). Unexpectedly, the glycemic and lipid profiles were consistently comparable between the two groups in the three sites. Furthermore, vitamin D was strikingly lower in obese patients with diabetes (P = 0.007). Finally, plasma ghrelin (P = 0.163), insulin (P = 0.063), and HOMA-IR (P = 0.166) were comparable between obese and nonobese patients with diabetes. CONCLUSION Diabetic obesity was significantly associated with female sex, young age, short disease duration, and noticeably low vitamin D, and a trend of high insulin levels. However, the obese and nonobese patients had comparable metabolic profiles with no differences in insulin resistance and ghrelin levels. Further studies, especially at a molecular level, are needed to explore this topic which is barely investigated.
Collapse
Affiliation(s)
- Hayder A. Giha
- Medical Biochemistry and Molecular Biology, Khartoum, Sudan (current)
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Kingdom of Bahrain
| | | | - Faris E. Joatar
- Clinical Biochemistry Laboratory, King Abdulaziz Hospital, Ministry of National Guard Health Affairs, Al Ahsa, Saudi Arabia
| | - Muhalab E. Ali
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Kingdom of Bahrain
| | - Einas M. Al-Harbi
- Al-Jawhara Center for Molecular Medicine, Arabian Gulf University, Manama, Bahrain
| | - Ali A. Al Qarni
- Endocrinology and Metabolism Section, King Abdulaziz Hospital, Ministry of National Guard Health Affairs, King Abdullah Medical Research Center-Eastern Region, Al Ahsa, Saudi Arabia
| |
Collapse
|
6
|
Grigoriadis A, Koutounidou S, Räisänen I, Arsenakis M, Sakellari D. Interaction between TCF7L2 rs7903146 Genotype, HbA1c Levels, and the Periodontal Status of Dental Patients. Eur J Dent 2021; 15:495-501. [PMID: 34041727 PMCID: PMC8382464 DOI: 10.1055/s-0041-1725578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
OBJECTIVE The aim of the study was to investigate the potential interaction between TCF7L2 rs7903146 genotype, which is implicated for type-2 diabetes mellitus genetic susceptibility, HbA1c levels, and the periodontal status of dental patients. MATERIALS AND METHODS HbA1c levels, clinical periodontal parameters (probing depth, clinical attachment level, bleeding on probing, and plaque index), and several parameters (such as body mass index [BMI], smoking habits, education level, and age) were recorded in 150 patients who fulfilled the criteria for screening for prediabetes/diabetes of the Centers for Disease Control and Prevention. DNA was extracted and the TCF7L2 single nucleotide polymorphism (SNP) rs7903146 was genotyped in all participants. RESULTS Thirty-one patients out of 150 tested were found with unknown hyperglycemia (20.7%). Regarding sex, education, parent with diabetes, normal BMI, smoking, age ≥45 years and prior testing for diabetes, no differences were observed between patients displaying HbA1c < 5.7 and ≥ 5.7% (Pearson's Chi-square test, p > 0.05). Regarding periodontal parameters and differences between subgroups (HbA1c levels ≥ 5.7 and HbA1c levels < 5.7), statistically significant differences were observed for probing depth (3.20 ± 0.94 vs. 2.81 ± 0.78 mm), clinical attachment level (3.54 ± 1.20 vs. 3.18 ± 1.06 mm) and bleeding on probing (0.62 ± 0.25 vs. 0.50 ± 0.24%) with hyperglycemic patients exhibiting worse periodontal conditions (Mann-Whitney test p < 0.05). The allelic and genotype frequencies for the transcription factor 7-like 2 (TCF7L2) gene, SNPs 7903146 did not exhibit a significant difference between the HbA1c > 5.7 and HbA1c < 5.7 groups and the periodontitis and nonperiodontitis subgroups respectively (Fisher's exact test >0.05). Statistical Analysis Patient characteristics and their association with prediabetes were tested by Pearson's Chi-square test (asymptotic, two sided). Differences of periodontal parameters between subgroups were tested with the Mann-Whitney U-test. The associations of allele and genotype frequencies in the patient and control groups were analyzed using the Fisher's exact test of independence.The significance level was set at the 0.05 for all tests. CONCLUSION A statistically significant association between TCF7L2 rs7903146 genotype and periodontal condition or HbA1c levels was not observed in contrast to statistically significant differences of clinical parameters of periodontitis in patients with hyperglycemia.
Collapse
Affiliation(s)
- Andreas Grigoriadis
- Department of Preventive Dentistry, Periodontology and Implant Biology, Dental School, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Periodontology, 424 General Military Training Hospital, Thessaloniki, Greece
| | - Symela Koutounidou
- Department of Genetics and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ismo Räisänen
- Department of Oral and Maxillofacial Diseases, Helsinki University and University Hospital, Helsinki, Finland
| | - Minas Arsenakis
- Department of Genetics and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitra Sakellari
- Department of Preventive Dentistry, Periodontology and Implant Biology, Dental School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
7
|
Aylward A, Okino ML, Benaglio P, Chiou J, Beebe E, Padilla JA, Diep S, Gaulton KJ. Glucocorticoid signaling in pancreatic islets modulates gene regulatory programs and genetic risk of type 2 diabetes. PLoS Genet 2021; 17:e1009531. [PMID: 33983929 PMCID: PMC8183998 DOI: 10.1371/journal.pgen.1009531] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/07/2021] [Accepted: 04/06/2021] [Indexed: 02/08/2023] Open
Abstract
Glucocorticoids are key regulators of glucose homeostasis and pancreatic islet function, but the gene regulatory programs driving responses to glucocorticoid signaling in islets and the contribution of these programs to diabetes risk are unknown. In this study we used ATAC-seq and RNA-seq to map chromatin accessibility and gene expression from eleven primary human islet samples cultured in vitro with the glucocorticoid dexamethasone at multiple doses and durations. We identified thousands of accessible chromatin sites and genes with significant changes in activity in response to glucocorticoids. Chromatin sites up-regulated in glucocorticoid signaling were prominently enriched for glucocorticoid receptor binding sites and up-regulated genes were enriched for ion transport and lipid metabolism, whereas down-regulated chromatin sites and genes were enriched for inflammatory, stress response and proliferative processes. Genetic variants associated with glucose levels and T2D risk were enriched in glucocorticoid-responsive chromatin sites, including fine-mapped variants at 51 known signals. Among fine-mapped variants in glucocorticoid-responsive chromatin, a likely casual variant at the 2p21 locus had glucocorticoid-dependent allelic effects on beta cell enhancer activity and affected SIX2 and SIX3 expression. Our results provide a comprehensive map of islet regulatory programs in response to glucocorticoids through which we uncover a role for islet glucocorticoid signaling in mediating genetic risk of T2D.
Collapse
Affiliation(s)
- Anthony Aylward
- Bioinformatics and Systems Biology graduate program, University of California San Diego, La Jolla, California, United States of America
| | - Mei-Lin Okino
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Paola Benaglio
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Joshua Chiou
- Biomedical Sciences graduate program, University of California San Diego, La Jolla, California, United States of America
| | - Elisha Beebe
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Jose Andres Padilla
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Sharlene Diep
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
8
|
Öhlund M, Müllner E, Moazzami A, Hermansson U, Pettersson A, Anderson F, Häggström J, Hansson-Hamlin H, Holst BS. Differences in metabolic profiles between the Burmese, the Maine coon and the Birman cat-Three breeds with varying risk for diabetes mellitus. PLoS One 2021; 16:e0249322. [PMID: 33886598 PMCID: PMC8062062 DOI: 10.1371/journal.pone.0249322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/15/2021] [Indexed: 12/31/2022] Open
Abstract
Feline diabetes mellitus shares many features with type 2 diabetes in people, regarding clinical presentation, physiology, and pathology. A breed predisposition for type 2 diabetes has been identified, with the Burmese breed at a fivefold increased risk of developing the condition compared to other purebred cats. We aimed to characterize the serum metabolome in cats (n = 63) using nuclear magnetic resonance metabolomics, and to compare the metabolite pattern of Burmese cats with that of two cat breeds of medium or low risk of diabetes, the Maine coon (MCO) and Birman cat, respectively. Serum concentrations of adiponectin, insulin and insulin-like growth factor-1 were also measured (n = 94). Burmese cats had higher insulin and lower adiponectin concentrations than MCO cats. Twenty one metabolites were discriminative between breeds using a multivariate statistical approach and 15 remained significant after adjustment for body weight and body condition score. Burmese cats had higher plasma levels of 2-hydroxybutyrate relative to MCO and Birman cats and increased concentrations of 2-oxoisocaproic acid, and tyrosine, and lower concentrations of dimethylglycine relative to MCO cats. The metabolic profile of MCO cats was characterized by high concentrations of arginine, asparagine, methionine, succinic acid and low levels of acetylcarnitine while Birman cats had the highest creatinine and the lowest taurine plasma levels, compared with MCO and Burmese. The pattern of metabolites in Burmese cats is similar to that in people with insulin resistance. In conclusion, the metabolic profile differed between healthy cats of three breeds. Detection of an abnormal metabolome might identify cats at risk of developing diabetes.
Collapse
Affiliation(s)
- Malin Öhlund
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Elisabeth Müllner
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ali Moazzami
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ulrika Hermansson
- University Animal Hospital, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ann Pettersson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Jens Häggström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Helene Hansson-Hamlin
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Bodil S. Holst
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
9
|
Risk Factors for Atonic Postpartum Hemorrhage: A Systematic Review and Meta-analysis. Obstet Gynecol 2021; 137:305-323. [PMID: 33417319 DOI: 10.1097/aog.0000000000004228] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/29/2020] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To identify and quantify risk factors for atonic postpartum hemorrhage. DATA SOURCES PubMed, CINAHL, EMBASE, Web of Science, and and ClinicalTrials.gov databases were searched for English language studies with no restrictions on date or location. Studies included randomized trials, prospective or retrospective cohort studies, and case-control studies of pregnant patients who developed atonic postpartum hemorrhage and reported at least one risk factor. METHODS OF STUDY SELECTION Title, abstract, and full-text screening were performed using the Raayan web application. Of 1,239 records screened, 27 studies were included in this review. Adjusted or unadjusted odds ratios (ORs), relative risks, or rate ratios were recorded or calculated. For each risk factor, a qualitative synthesis of low and moderate risk of bias studies classifies the risk factor as definite, likely, unclear, or not a risk factor. For risk factors with sufficiently homogeneous definitions and reference ranges, a quantitative meta-analysis of low and moderate risk of bias studies was implemented to estimate a combined OR. TABULATION, INTEGRATION, AND RESULTS Forty-seven potential risk factors for atonic postpartum hemorrhage were identified in this review, of which 15 were judged definite or likely risk factors. The remaining 32 assessed risk factors showed no association with atonic postpartum hemorrhage or had conflicting or unclear evidence. CONCLUSION A substantial proportion of postpartum hemorrhage occurs in the absence of recognized risk factors. Many risk factors for atonic hemorrhage included in current risk-assessment tools were confirmed, with the greatest risk conferred by prior postpartum hemorrhage of any etiology, placenta previa, placental abruption, uterine rupture, and multiple gestation. Novel risk factors not currently included in risk-assessment tools included hypertension, diabetes, and ethnicity. Obesity and magnesium were not associated with atonic postpartum hemorrhage in this review. SYSTEMATIC REVIEW REGISTRATION PROSPERO, CRD42020157521.
Collapse
|
10
|
AlDehaini DMB, Al-Bustan SA, Malalla ZHA, Ali ME, Sater M, Giha HA. The influence of TERC, TERT and ACYP2 genes polymorphisms on plasma telomerase concentration, telomeres length and T2DM. Gene 2021; 766:145127. [PMID: 32937184 DOI: 10.1016/j.gene.2020.145127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/20/2020] [Accepted: 09/01/2020] [Indexed: 11/21/2022]
Abstract
Telomeres are duplex tandem repeats of DNA sequence 5'-TTAGGG-3' at chromosomal ends synthesized by telomerase enzyme (TE). Telomeres length (TL) shortening is associated with age and age-related disorders. Recently, we demonstrated marked leukocytes TL (LTL) shortening in T2DM. To set the relationship between the TE, LTL and T2DM, we analyzed samples from 212 Kuwaiti subjects, 112 patients withT2DM and 100 non-diabetic subjects. The plasma TE and fasting insulin were measured by ELISA, the LTL was estimated by qPCR and three SNPs of genes related to TL; TERC rs12696304 (C/G), TERT rs2736100 (C/A) and ACYP2 rs6713088 (C/G) were genotyped by rtPCR. Results revealed comparable TE levels and alleles/genotypes between the cases and controls with no influence of either on the LTL. Interestingly, although the plasma concentration of the TE was generally low, it was significantly influenced by the TERT and ACYP2 but not TERC polymorphisms. The CC genotype carriers of rs2736100 (C/A) had significantly higher plasma TE levels compared to CA and AA carriers, p 0.009 and p 0.047, respectively, and the A-allele was associated with low TE, p 0.018. Similarly, significantly higher TE levels were detected in CC carriers of ACYP2 rs6713088 (C/G) compared with GC carriers, p 0.002, and the G-allele was associated with low TE, p 0.009. Finally, the TERT and ACYP2 polymorphisms had an influence on blood glucose levels. In conclusion, the telomeres shortening in T2DM was not due to TE deficiency or gene polymorphisms, while the TE levels were significantly associated with the TERT and ACYP2 but not TERC polymorphisms.
Collapse
Affiliation(s)
- Dhuha M B AlDehaini
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain; Farwaniya Hospital, Biochemistry Laboratory, PO Box 13373, Farwaniya 81004, Kuwait
| | - Suzanne A Al-Bustan
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Zainab Hasan Abdulla Malalla
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain
| | - Muhalab E Ali
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain
| | - Mai Sater
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain
| | - Hayder A Giha
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain.
| |
Collapse
|
11
|
Denyer AL, Catchpole B, Davison LJ. Genetics of canine diabetes mellitus part 2: Current understanding and future directions. Vet J 2021; 270:105612. [PMID: 33641811 DOI: 10.1016/j.tvjl.2021.105612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023]
Abstract
Part 1 of this 2-part review outlined the importance of disease classification in diabetes genetic studies, as well as the ways in which genetic variants may contribute to risk of a complex disease within an individual, or within a particular group of individuals. Part 2, presented here, describes in more detail our current understanding of the genetics of canine diabetes mellitus compared to our knowledge of the human disease. Ongoing work to improve our knowledge, using new technologies, is also introduced.
Collapse
Affiliation(s)
- Alice L Denyer
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - Brian Catchpole
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - Lucy J Davison
- Department of Clinical Sciences and Services, Royal Veterinary College, Hatfield, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | |
Collapse
|
12
|
Dai Y, Pracana R, Holland PWH. Divergent genes in gerbils: prevalence, relation to GC-biased substitution, and phenotypic relevance. BMC Evol Biol 2020; 20:134. [PMID: 33076817 PMCID: PMC7574485 DOI: 10.1186/s12862-020-01696-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/29/2020] [Indexed: 11/25/2022] Open
Abstract
Background Two gerbil species, sand rat (Psammomys obesus) and Mongolian jird (Meriones unguiculatus), can become obese and show signs of metabolic dysregulation when maintained on standard laboratory diets. The genetic basis of this phenotype is unknown. Recently, genome sequencing has uncovered very unusual regions of high guanine and cytosine (GC) content scattered across the sand rat genome, most likely generated by extreme and localized biased gene conversion. A key pancreatic transcription factor PDX1 is encoded by a gene in the most extreme GC-rich region, is remarkably divergent and exhibits altered biochemical properties. Here, we ask if gerbils have proteins in addition to PDX1 that are aberrantly divergent in amino acid sequence, whether they have also become divergent due to GC-biased nucleotide changes, and whether these proteins could plausibly be connected to metabolic dysfunction exhibited by gerbils. Results We analyzed ~ 10,000 proteins with 1-to-1 orthologues in human and rodents and identified 50 proteins that accumulated unusually high levels of amino acid change in the sand rat and 41 in Mongolian jird. We show that more than half of the aberrantly divergent proteins are associated with GC biased nucleotide change and many are in previously defined high GC regions. We highlight four aberrantly divergent gerbil proteins, PDX1, INSR, MEDAG and SPP1, that may plausibly be associated with dietary metabolism. Conclusions We show that through the course of gerbil evolution, many aberrantly divergent proteins have accumulated in the gerbil lineage, and GC-biased nucleotide substitution rather than positive selection is the likely cause of extreme divergence in more than half of these. Some proteins carry putatively deleterious changes that could be associated with metabolic and physiological phenotypes observed in some gerbil species. We propose that these animals provide a useful model to study the ‘tug-of-war’ between natural selection and the excessive accumulation of deleterious substitutions mutations through biased gene conversion.
Collapse
Affiliation(s)
- Yichen Dai
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford, OX1 3SZ, UK
| | - Rodrigo Pracana
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford, OX1 3SZ, UK
| | - Peter W H Holland
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford, OX1 3SZ, UK.
| |
Collapse
|
13
|
Fan J, Liang R. Quantitative assessment of TLR4 gene polymorphisms and T2DM risk: A meta-analysis. Mol Genet Genomic Med 2020; 8:e1466. [PMID: 32822111 PMCID: PMC7549608 DOI: 10.1002/mgg3.1466] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Numerous studies have evaluated the association between TLR4 gene polymorphisms and T2DM risk. However, the findings were inconsistent and controversial. METHODS In order to drive a more precise estimation, we carried out a meta-analysis based on 41 studies involving 23,250 cases and 24,760 controls. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to assess the strength of association. RESULTS Our meta-analysis provides evidence that rs4986790 polymorphism was associated with an increased risk of T2DM in Asian (AG vs. AA, OR = 1.23, 95% CI = 1.01-1.50, p = 0.042; G vs. A, OR = 1.21, 95% CI = 1.01-1.44, p = 0.041). Rs4986791 polymorphism was related to an increased risk of T2DM both in Asian (AG vs. AA, OR = 1.76, 95% CI = 1.11-2.80, p = 0.017; G vs. A, OR = 1.63, 95% CI = 1.04-2.55, p = 0.034) and Caucasian (GG vs. AA, OR = 2.42, 95% CI = 1.23-4.75, p = 0.010). Rs11536889 polymorphism may have a protective effect on T2DM in Chinese populations (CC vs. GG, OR = 0.62, 95% CI = 0.40-0.96, p = 0.031; GC vs. GG, OR = 0.77, 95% CI = 0.61-0.98, p = 0.034; CC vs. GC/GG, OR = 0.81, 95% CI = 0.69-0.96, p = 0.013; C vs. G, OR = 0.76, 95% CI = 0.59-0.97, p = 0.027), whereas rs1927911 may have no impact. CONCLUSIONS These findings supported that rs4986790, rs4986791, and rs11536889 may contribute to the risk of T2DM.
Collapse
Affiliation(s)
- Jinzhuo Fan
- Department of Traditional Chinese Medicine and Acupuncture, Hainan Hospital of General Hospital of Chinese PLA, Sanya, China
| | - Renxian Liang
- Department of Cardiovascular and Endocrinology, Beibei Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
14
|
Hubáček JA, Šedová L, Olišarová V, Adámková V, Tóthová V. Different prevalence of T2DM risk alleles in Roma population in comparison with the majority Czech population. Mol Genet Genomic Med 2020; 8:e1361. [PMID: 32578971 PMCID: PMC7507457 DOI: 10.1002/mgg3.1361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The Czech governmental study suggests up to a 25% higher prevalence of type 2 diabetes mellitus (T2DM) in the Roma population than within the majority population. It is not known whether and to what extent these differences have a genetic background. METHODS To analyze whether the frequencies of the alleles/genotypes of the FTO, TCF7L2, CDKN2A/2B, MAEA, TLE4, IGF2BP2, ARAP1, and KCNJ11 genes differ between the two major ethnic groups in the Czech Republic, we examined them in DNA samples from 302 Roma individuals and 298 Czech individuals. RESULTS Compared to the majority population, Roma are more likely to carry risk alleles in the FTO (26% vs. 16% GG homozygotes, p < .01), IGF2BP2 (22% vs. 10% TT homozygotes, p < .0001), ARAP1 (98% vs. 95% of A allele carriers, p < .005), and CDKN2A/2B (81% vs. 66% of TT homozygotes, p < .001) genes; however, less frequently they are carriers of the TCF7L2 risk allele (34% vs. 48% of the T allele p < .0005). Finally, we found significant accumulation of T2DM-associated alleles between the Roma population in comparison with the majority population (25.4% vs. 15.2% of the carriers of at least 12 risk alleles; p < .0001). CONCLUSION The increased prevalence of T2DM in the Roma population may have a background in different frequencies of the risk alleles of genes associated with T2DM development.
Collapse
Affiliation(s)
- Jaroslav A. Hubáček
- Centre for Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Lenka Šedová
- Faculty of Health and Social SciencesUniversity of South BohemiaČeské BudějoviceCzech Republic
| | - Věra Olišarová
- Faculty of Health and Social SciencesUniversity of South BohemiaČeské BudějoviceCzech Republic
| | - Věra Adámková
- Department of Preventive CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Valérie Tóthová
- Faculty of Health and Social SciencesUniversity of South BohemiaČeské BudějoviceCzech Republic
| |
Collapse
|
15
|
Xi X, Ma J. A meta-analysis on genetic associations between Transcription Factor 7 Like 2 polymorphisms and type 2 diabetes mellitus. Genomics 2020; 112:1192-1196. [DOI: 10.1016/j.ygeno.2019.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/05/2019] [Indexed: 12/21/2022]
|
16
|
Souliotis K, Koutsovasilis A, Vatheia G, Golna C, Nikolaidi S, Hatziagelaki E, Kotsa K, Koufakis T, Melidonis A, Papazafiropoulou A, Tentolouris N, Siami E, Sotiropoulos A. Profile and factors associated with glycaemic control of patients with type 2 diabetes in Greece: results from the diabetes registry. BMC Endocr Disord 2020; 20:16. [PMID: 31992275 PMCID: PMC6986011 DOI: 10.1186/s12902-020-0496-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 01/20/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Strict glycaemic control early in the treatment process has been shown to reduce the occurrence of micro- and macro- vascular complications of diabetes in the long-term. Thus, treatment guidelines advise early intensification of treatment to achieve glycaemic control goals. However, evidence in Greece suggests that, despite guideline recommendations, glycaemic control among patients with T2DM remains challenging. This study presents the demographic and clinical characteristics of patients with T2DM in Greece using data from an electronic registry designed specifically for this treatment category and investigates the factors that are independently associated with glycaemic control. METHODS This is a multi-center, observational, cross-sectional study to investigate epidemiological and clinical factors affecting glycaemic control among patients with T2DM in Greece. Data was collected via a web-based disease registry, the Diabetes Registry, which operated from January 1st to December 31st, 2017. Five large specialized diabetes centers operating in Greek hospitals participated in the study. RESULTS Data for 1141 patients were retrieved (aged 63.02 ± 12.65 years, 56.9% male). Glycaemic control (Hb1Ac < 7%) was not achieved in 57.1% of patients. Factors independently associated with poor glycaemic control were: family history of diabetes [OR: 1.53, 95% CI: 1.06-2.23], BMI score between 25 to 30 [OR: 2.08, 95% CI: 1.05-4.13] or over 30 [OR: 2.12, 95% CI 1.12-4.07], elevated LDL levels [OR: 1.53, 95% 1.06-2.21] and low HDL levels [OR: 2.12, 95% CI: 1.44-3.12]. Lastly, use of injectable antidiabetic agents (in monotherapy or in combination) was less likely to be associated with poor glycaemic control versus treatment with combination of oral and injectable agents [OR: 0.50, 95% CI: 0.24-1.01]. This association was found to be marginally statistically significant. CONCLUSION Inadequate lipid control, family history of diabetes and presence of obesity (ΒΜΙ ≥ 30 kg/m2) were associated with poor glycaemic control among study sample, whereas use of injectable antidiabetic agents was less likely to be associated with poor glycaemic control. These findings indicate how complex optimal glycaemic control is, highlighting the need for tailored interventions in high-risk subpopulations with T2DM.
Collapse
Affiliation(s)
- Kyriakos Souliotis
- Department of Social and Education Policy, University of Peloponnese, Corinth, Greece
- Health Policy Institute, 8, Agisilaou Str, 15123 Maroussi, Athens Greece
| | - Anastasios Koutsovasilis
- 3rd Department of Internal Medicine and Diabetes Center, General Hospital of Nikaia, Piraeus, Greece
| | - Georgia Vatheia
- Department of Social and Education Policy, University of Peloponnese, Corinth, Greece
- Health Policy Institute, 8, Agisilaou Str, 15123 Maroussi, Athens Greece
| | | | - Sofia Nikolaidi
- Department of Social and Education Policy, University of Peloponnese, Corinth, Greece
- Health Policy Institute, 8, Agisilaou Str, 15123 Maroussi, Athens Greece
| | - Erifili Hatziagelaki
- 2nd Department of Internal Medicine-Propaedeutic, Research Institute and Diabetes Center “Attikon” University Hospital Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism and Diabetes Center, 1st Department of Internal Medicine, Medical School, Aristotle University, AHEPA University Hospital, Thessaloniki, Greece
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Center, 1st Department of Internal Medicine, Medical School, Aristotle University, AHEPA University Hospital, Thessaloniki, Greece
| | - Andreas Melidonis
- 1st Department of Internal Medicine and Diabetes Center, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - Athanasia Papazafiropoulou
- 1st Department of Internal Medicine and Diabetes Center, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - Nikolaos Tentolouris
- 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Evangelia Siami
- 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Alexios Sotiropoulos
- 3rd Department of Internal Medicine and Diabetes Center, General Hospital of Nikaia, Piraeus, Greece
| |
Collapse
|
17
|
A meta-analysis on associations of FTO, MTHFR and TCF7L2 polymorphisms with polycystic ovary syndrome. Genomics 2019; 112:1516-1521. [PMID: 31470081 DOI: 10.1016/j.ygeno.2019.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 07/31/2019] [Accepted: 08/26/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND We aimed to better clarify the relationship between FTO/MTHFR/TCF7L2 polymorphisms and PCOS in a larger combined population by performing a meta-analysis. METHODS Eligible articles were retrieved from Pubmed, Embase, Web of Science and CNKI. Review Manager Version was used to perform statistical analyses. RESULTS Forty-six studies were included for this meta-analysis. FTO rs9939609 polymorphism was found to be significantly associated with PCOS under dominant, recessive, over-dominant and allele comparisons, MTHFR rs1801131 polymorphism was found to be significantly associated with PCOS under recessive and allele comparisons, and MTHFR rs1801133 polymorphism was also found to be significantly associated with PCOS under dominant, recessive and allele comparisons in general population. In subgroup analyses, we found that positive results were mainly driven by the Asians. CONCLUSIONS Collectively, this meta-analysis proved that FTO rs9939609, MTHFR rs1801131 and MTHFR rs1801133 polymorphisms may serve as predisposing factors of PCOS, especially for Asians.
Collapse
|
18
|
Liu Y, Shi L, Song X, Shi C, Lou W, Zhang D, Wang AD, Luo L. Altered Brain Regional Homogeneity in First-Degree Relatives of Type 2 Diabetics: A functional MRI Study. Exp Clin Endocrinol Diabetes 2019; 128:737-744. [PMID: 31137069 DOI: 10.1055/a-0883-4955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aimed to investigate regional homogeneity in the first-: degree relatives of type 2 diabetes patients. METHODS Seventy-eight subjects, including 26 type 2 diabetes patients, 26 first-: degree relatives, and 26 healthy controls, were assessed. All participants underwent resting-state functional magnetic resonance imaging scanning. The estimated regional homogeneity value was used to evaluate differences in brain activities. RESULTS In first-: degree relatives, we observed significantly decreased regional homogeneity in the left anterior cingulate cortex, left insula, and bilateral temporal lobes, and increased regional homogeneity in the left superior frontal gyrus, right anterior cingulate cortex, and bilateral posterior cingulate cortex compared to healthy controls. In type 2 diabetes patients, we detected altered regional homogeneity in the left anterior cingulate cortex, left insula, bilateral posterior cingulate cortex, and several other brain regions compared to healthy controls. Both first-: degree relatives and type 2 diabetes patients showed decreased regional homogeneity in the left superior temporal gyrus, right middle temporal gyrus, left anterior cingulate cortex, left insula, and increased regional homogeneity in the left superior frontal gyrus and bilateral posterior cingulate cortex. CONCLUSION These findings suggest that altered regional homogeneity in the left anterior cingulate cortex, left insula, left superior frontal gyrus, bilateral posterior cingulate cortex, and bilateral temporal lobes might be a neuroimaging biomarker of type 2 diabetes -: related brain dysfunction.
Collapse
Affiliation(s)
- Yiyong Liu
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, Research Centre for Medical Image Computing, The Chinese University of Hong Kong, China.,Chow Yuk Ho Technology Centre for Innovative Medicine, The Chinese University of Hong Kong, China
| | - Xiubao Song
- Department of Rehabilitation, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Changzheng Shi
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wutao Lou
- Department of Imaging and Interventional Radiology, Research Centre for Medical Image Computing, The Chinese University of Hong Kong, China
| | - Dong Zhang
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Alan D Wang
- Auckland Bioengineering Institute, and Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Shenzhen SmartView MedTech Limited, Shenzhen, China
| | - Liangping Luo
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
19
|
Associations between cytotoxic T-lymphocyte-associated antigen 4 gene polymorphisms and diabetes mellitus: a meta-analysis of 76 case-control studies. Biosci Rep 2019; 39:BSR20190309. [PMID: 30988065 PMCID: PMC6522704 DOI: 10.1042/bsr20190309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/02/2019] [Accepted: 04/09/2019] [Indexed: 01/11/2023] Open
Abstract
Background: Several genetic association studies already investigated potential roles of cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) gene polymorphisms in diabetes mellitus (DM), with inconsistent results. Therefore, we performed this meta-analysis to better assess the relationship between CTLA-4 gene polymorphisms and DM in a larger pooled population. Methods: PubMed, Embase, Web of Science, and CNKI were systematically searched for eligible studies. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to estimate the strength of associations between CTLA-4 gene polymorphisms and DM in all possible genetic models. Results: A total of 76 studies were finally included in our analyses. Significant associations with susceptibility to type 1 diabetes mellitus (T1DM) were detected for rs231775 (dominant model: P=0.008, OR = 0.83, 95%CI 0.73–0.95; recessive model: P=0.003, OR = 1.27, 95%CI 1.09–1.50; allele model: P=0.004, OR = 0.85, 95%CI 0.77–0.95) and rs5742909 (recessive model: P=0.02, OR = 1.50, 95%CI 1.05–2.13) polymorphisms in overall population. Further subgroup analyses revealed that rs231775 polymorphism was significantly associated with susceptibility to T1DM in Caucasians and South Asians, and rs5742909 polymorphism was significantly associated with susceptibility to T1DM in South Asians. Moreover, rs231775 polymorphism was also found to be significantly associated with susceptibility to type 2 diabetes mellitus (T2DM) in East Asians and South Asians. Conclusions: Our findings indicated that rs231775 and rs5742909 polymorphisms may serve as genetic biomarkers of T1DM, and rs231775 polymorphism may also serve as a genetic biomarker of T2DM.
Collapse
|
20
|
Pancreatic islet chromatin accessibility and conformation reveals distal enhancer networks of type 2 diabetes risk. Nat Commun 2019; 10:2078. [PMID: 31064983 PMCID: PMC6505525 DOI: 10.1038/s41467-019-09975-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
Genetic variants affecting pancreatic islet enhancers are central to T2D risk, but the gene targets of islet enhancer activity are largely unknown. We generate a high-resolution map of islet chromatin loops using Hi-C assays in three islet samples and use loops to annotate target genes of islet enhancers defined using ATAC-seq and published ChIP-seq data. We identify candidate target genes for thousands of islet enhancers, and find that enhancer looping is correlated with islet-specific gene expression. We fine-map T2D risk variants affecting islet enhancers, and find that candidate target genes of these variants defined using chromatin looping and eQTL mapping are enriched in protein transport and secretion pathways. At IGF2BP2, a fine-mapped T2D variant reduces islet enhancer activity and IGF2BP2 expression, and conditional inactivation of IGF2BP2 in mouse islets impairs glucose-stimulated insulin secretion. Our findings provide a resource for studying islet enhancer function and identifying genes involved in T2D risk. Risk loci for type 2 diabetes (T2D) reside in pancreatic islet enhancers. Here, the authors generate high-resolution maps of islet chromatin conformation using Hi-C which they combine with ATAC-seq and ChIP-seq data to annotate candidate target genes of enhancers and validate IGF2BP2 activity in mouse islets.
Collapse
|
21
|
Postolache TT, del Bosque-Plata L, Jabbour S, Vergare M, Wu R, Gragnoli C. Co-shared genetics and possible risk gene pathway partially explain the comorbidity of schizophrenia, major depressive disorder, type 2 diabetes, and metabolic syndrome. Am J Med Genet B Neuropsychiatr Genet 2019; 180:186-203. [PMID: 30729689 PMCID: PMC6492942 DOI: 10.1002/ajmg.b.32712] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 11/16/2018] [Accepted: 12/07/2018] [Indexed: 12/20/2022]
Abstract
Schizophrenia (SCZ) and major depressive disorder (MDD) in treatment-naive patients are associated with increased risk for type 2 diabetes (T2D) and metabolic syndrome (MetS). SCZ, MDD, T2D, and MetS are often comorbid and their comorbidity increases cardiovascular risk: Some risk genes are likely co-shared by them. For instance, transcription factor 7-like 2 (TCF7L2) and proteasome 26S subunit, non-ATPase 9 (PSMD9) are two genes independently reported as contributing to T2D and SCZ, and PSMD9 to MDD as well. However, there are scarce data on the shared genetic risk among SCZ, MDD, T2D, and/or MetS. Here, we briefly describe T2D, MetS, SCZ, and MDD and their genetic architecture. Next, we report separately about the comorbidity of SCZ and MDD with T2D and MetS, and their respective genetic overlap. We propose a novel hypothesis that genes of the prolactin (PRL)-pathway may be implicated in the comorbidity of these disorders. The inherited predisposition of patients with SCZ and MDD to psychoneuroendocrine dysfunction may confer increased risk of T2D and MetS. We illustrate a strategy to identify risk variants in each disorder and in their comorbid psychoneuroendocrine and mental-metabolic dysfunctions, advocating for studies of genetically homogeneous and phenotype-rich families. The results will guide future studies of the shared predisposition and molecular genetics of new homogeneous endophenotypes of SCZ, MDD, and metabolic impairment.
Collapse
Affiliation(s)
- Teodor T. Postolache
- Department of Psychiatry, Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, Maryland,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Denver, Colorado,Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, Maryland
| | - Laura del Bosque-Plata
- National Institute of Genomic Medicine, Nutrigenetics and Nutrigenomic Laboratory, Mexico City, Mexico
| | - Serge Jabbour
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolic Disease, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Michael Vergare
- Department of Psychiatry and Human Behavior, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rongling Wu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania,Department of Statistics, Penn State College of Medicine, Hershey, Pennsylvania
| | - Claudia Gragnoli
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolic Disease, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania,Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania,Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
22
|
Shen LL, Jin Y. Effects of MTNR1B genetic variants on the risk of type 2 diabetes mellitus: A meta-analysis. Mol Genet Genomic Med 2019; 7:e611. [PMID: 30811895 PMCID: PMC6503061 DOI: 10.1002/mgg3.611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 12/17/2022] Open
Abstract
Background Whether melatonin receptor 1B (MTNR1B) variants are associated with type 2 diabetes mellitus (T2DM) remains unclear. Therefore, we performed this meta‐analysis to better explore correlations between MTNR1B variants and T2DM. Methods Literature research was performed in PubMed, Medline, and Embase. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated. Results Totally 21 studies were enrolled to analyses. Pooled overall analyses showed that MTNR1B rs10830963 variant was significantly correlated with the susceptibility to T2DM (allele model: p = 0.02, OR = 0.97, 95% CI 0.95–1.00). Further subgroup analyses by ethnicity of participants revealed that rs10830963 variant was significantly correlated with the susceptibility to T2DM in South Asians, but not in Caucasians or East Asians. No any other positive results were found in overall and subgroup analyses. Conclusions Our findings indicated that MTNR1B rs10830963 variant might serve as a genetic biomarker of T2DM, especially in South Asians.
Collapse
Affiliation(s)
- Ling-Long Shen
- Department of Clinical Laboratory, Huzhou Maternity and Child Health Care Hospital, Huzhou, China
| | - Yin Jin
- Department of Clinical Laboratory, Huzhou Central Hospital, Huzhou, China
| |
Collapse
|
23
|
Lou L, Wang J, Wang J. Genetic associations between Transcription Factor 7 Like 2 rs7903146 polymorphism and type 2 diabetes mellitus: a meta-analysis of 115,809 subjects. Diabetol Metab Syndr 2019; 11:56. [PMID: 31312259 PMCID: PMC6612193 DOI: 10.1186/s13098-019-0451-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/24/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Some genetic association studies tried to investigate potential associations of Transcription Factor 7 Like 2 (TCF7L2) rs7903146 polymorphism with type 2 diabetes mellitus (T2DM). However, the results of these studies were not consistent. Thus, we performed the present meta-analysis to explore associations between TCF7L2 rs7903146 polymorphism and T2DM in a larger pooled population. METHODS Systematic literature research of PubMed, Web of Science and Embase was performed to identify eligible studies for pooled analyses. I2 statistics were employed to assess between-study heterogeneities. If I2 was greater than 50%, random-effect models (REMs) would be used to pool the data. Otherwise, fixed-effect models (FEMs) would be applied for synthetic analyses. RESULTS Totally 68 studies with 115,809 subjects were included for analyses. The pooled analyses showed that TCF7L2 rs7903146 (dominant model: p < 0.0001; recessive model: p < 0.0001; over-dominant model: p < 0.0001; allele model: p < 0.0001) polymorphism was significantly associated with susceptibility to T2DM in overall population. Further subgroup analyses revealed similar significant findings in both Asians and Caucasians. CONCLUSIONS In conclusion, our findings supported that TCF7L2 rs7903146 polymorphism could be used to identify individuals at high risk of developing T2DM in Asians and Caucasians.
Collapse
Affiliation(s)
- Liying Lou
- Department of Endocrinology, Shengzhou People’s Hospital, No. 666 Dangui Road of Sanjiang Street, Shaoxing, 312400 Zhejiang China
| | - Jingjing Wang
- Department of Endocrinology, Shengzhou People’s Hospital, No. 666 Dangui Road of Sanjiang Street, Shaoxing, 312400 Zhejiang China
| | - Jing Wang
- Department of Endocrinology, Shengzhou People’s Hospital, No. 666 Dangui Road of Sanjiang Street, Shaoxing, 312400 Zhejiang China
| |
Collapse
|
24
|
Dong J, Ping Y, Wang Y, Zhang Y. The roles of endothelial nitric oxide synthase gene polymorphisms in diabetes mellitus and its associated vascular complications: a systematic review and meta-analysis. Endocrine 2018; 62:412-422. [PMID: 30140993 DOI: 10.1007/s12020-018-1683-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 07/09/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE The roles of endothelial nitric oxide synthase (eNOS) gene polymorphisms in diabetes mellitus (DM) were intensively analyzed, but the results of these studies were inconsistent. Therefore, we performed this study to better assess the relationship between eNOS genetic variations and DM. METHODS Eligible studies were searched in PubMed, Medline, Embase, and Web of Science. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess correlations between eNOS polymorphisms and DM. RESULTS A total of 91 studies were finally included in our analyses. Significant associations with the susceptibility to DM were detected for the rs891512, rs1799983, rs2070744, and rs869109213 polymorphisms. As for vascular complications in DM, significant associations with the susceptibility to diabetic nephropathy were detected for the rs1799983 and rs2070744 polymorphisms. In addition, we also found that the rs1799983 polymorphism was significantly associated with the susceptibility to peripheral artery disease, whereas the rs2070744 polymorphism was significantly associated with the susceptibility to coronary artery disease in DM patients. Further subgroup analyses on the basis of type of disease and ethnicity of participants showed similar positive results. CONCLUSIONS In conclusion, our findings indicate that rs891512, rs1799983, rs2070744, and rs869109213 polymorphisms may serve as genetic biomarkers of DM, while rs1799983, rs2070744, and rs869109213 polymorphisms may contribute to the development of vascular complications in DM.
Collapse
Affiliation(s)
- Jinnan Dong
- Department of Clinical Nutrition, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, China
| | - Yu Ping
- Department of Clinical Nutrition, Yantaishan Hospital, Yantai, 264001, Shandong, China
| | - Yuhong Wang
- Hemodialysis Room, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, China
| | - Yang Zhang
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, Shandong, China.
| |
Collapse
|
25
|
Chao CT, Wang J, Huang JW, Chien KL. Acarbose Use and Liver Injury in Diabetic Patients With Severe Renal Insufficiency and Hepatic Diseases: A Propensity Score-Matched Cohort Study. Front Pharmacol 2018; 9:860. [PMID: 30131698 PMCID: PMC6090209 DOI: 10.3389/fphar.2018.00860] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Acarbose has been deemed contraindicated in diabetic patients with chronic kidney disease (CKD) or end-stage renal disease (ESRD), but such use is not uncommon. We tested whether this concept hold true in this population with different background hepatic diseases. Methods: All incident diabetic patients (n = 2,036,531) with stage 5 CKD/ESRD were enrolled from Taiwan between 2017 and 2013 and divided into those without chronic liver disease (CLD), with CLD but without cirrhosis, and those with cirrhosis. Among each group, acarbose users, defined as cumulative use >30 days within the preceding year, were propensity-score matched 1:2 to non-users. Our main outcome was the development of liver injury events during follow-up. Results: Acarbose users did not exhibit an increased incidence of liver injury during follow-up compared to non-users (hazard ratio and 95% confidence interval, 1.04 [0.88–1.25], 0.97 [0.61–1.56], and 0.71 [0.33–1.54] among those without CLD, with CLD but without cirrhosis, and those with cirrhosis, respectively), after adjusting for demographic profiles, comorbidities, potentially hepatotoxic medication use, and diabetic severity. Conclusions: The incidence of liver injury did not increase significantly among diabetic acarbose users with severe renal insufficiency than non-users, regardless of the presence or absence of chronic liver disease. Our findings support the renaissance of acarbose as a useful adjunct in diabetic patients with stage 5 and 5D chronic kidney disease.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Department of Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Geriatric and Community Medicine Research Center, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan
| | - Jui Wang
- College of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Jenq-Wen Huang
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-Liong Chien
- College of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
26
|
Dziewulska A, Dobosz AM, Dobrzyn A. High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes. Genes (Basel) 2018; 9:E374. [PMID: 30050001 PMCID: PMC6115814 DOI: 10.3390/genes9080374] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex disorder that is caused by a combination of genetic, epigenetic, and environmental factors. High-throughput approaches have opened a new avenue toward a better understanding of the molecular bases of T2D. A genome-wide association studies (GWASs) identified a group of the most common susceptibility genes for T2D (i.e., TCF7L2, PPARG, KCNJ1, HNF1A, PTPN1, and CDKAL1) and illuminated novel disease-causing pathways. Next-generation sequencing (NGS)-based techniques have shed light on rare-coding genetic variants that account for an appreciable fraction of T2D heritability (KCNQ1 and ADRA2A) and population risk of T2D (SLC16A11, TPCN2, PAM, and CCND2). Moreover, single-cell sequencing of human pancreatic islets identified gene signatures that are exclusive to α-cells (GCG, IRX2, and IGFBP2) and β-cells (INS, ADCYAP1, INS-IGF2, and MAFA). Ongoing epigenome-wide association studies (EWASs) have progressively defined links between epigenetic markers and the transcriptional activity of T2D target genes. Differentially methylated regions were found in TCF7L2, THADA, KCNQ1, TXNIP, SOCS3, SREBF1, and KLF14 loci that are related to T2D. Additionally, chromatin state maps in pancreatic islets were provided and several non-coding RNAs (ncRNA) that are key to T2D pathogenesis were identified (i.e., miR-375). The present review summarizes major progress that has been made in mapping the (epi)genomic landscape of T2D within the last few years.
Collapse
Affiliation(s)
- Anna Dziewulska
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland.
| | - Aneta M Dobosz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland.
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland.
| |
Collapse
|
27
|
Kong Y, Sharma RB, Ly S, Stamateris RE, Jesdale WM, Alonso LC. CDKN2A/B T2D Genome-Wide Association Study Risk SNPs Impact Locus Gene Expression and Proliferation in Human Islets. Diabetes 2018; 67:872-884. [PMID: 29432124 PMCID: PMC5910004 DOI: 10.2337/db17-1055] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/29/2018] [Indexed: 12/18/2022]
Abstract
Genome-wide association studies link the CDKN2A/B locus with type 2 diabetes (T2D) risk, but mechanisms increasing risk remain unknown. The CDKN2A/B locus encodes cell cycle inhibitors p14, p15, and p16; MTAP; and ANRIL, a long noncoding RNA. The goal of this study was to determine whether CDKN2A/B T2D risk SNPs impact locus gene expression, insulin secretion, or β-cell proliferation in human islets. Islets from donors without diabetes (n = 95) were tested for SNP genotype (rs10811661, rs2383208, rs564398, and rs10757283), gene expression (p14, p15, p16, MTAP, ANRIL, PCNA, KI67, and CCND2), insulin secretion (n = 61), and β-cell proliferation (n = 47). Intriguingly, locus genes were coregulated in islets in two physically overlapping cassettes: p14-p16-ANRIL, which increased with age, and MTAP-p15, which did not. Risk alleles at rs10811661 and rs2383208 were differentially associated with expression of ANRIL, but not p14, p15, p16, or MTAP, in age-dependent fashion, such that younger homozygous risk donors had higher ANRIL expression, equivalent to older donor levels. We identified several risk SNP combinations that may impact locus gene expression, suggesting possible mechanisms by which SNPs impact locus biology. Risk allele carriers at ANRIL coding SNP rs564398 had reduced β-cell proliferation index. In conclusion, CDKN2A/B locus SNPs may impact T2D risk by modulating islet gene expression and β-cell proliferation.
Collapse
Affiliation(s)
- Yahui Kong
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Rohit B Sharma
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Socheata Ly
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Rachel E Stamateris
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - William M Jesdale
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA
| | - Laura C Alonso
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
28
|
Wang L, Niu YM, Wu SS, Zhang C, Zhou L, Zuo HX, Wang P. A Study on the Association Between Polymorphisms in the Cytochrome P450 Family 17 Subfamily A Member 1 Gene Region and Type 2 Diabetes Mellitus in Han Chinese. Front Endocrinol (Lausanne) 2018; 9:323. [PMID: 29942286 PMCID: PMC6004380 DOI: 10.3389/fendo.2018.00323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/28/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cytochrome P450 family 17 subfamily A member 1 (CYP17A1) gene encodes a key enzyme in the synthesis and metabolism of steroid hormones and has been associated with various factors, such as hypertension, insulin resistance, and polycystic ovary syndrome. However, whether the gene was associated with type 2 diabetes mellitus (T2DM) has not been reported yet. Therefore, we sought to investigate whether CYP17A1 was associated with T2DM and related traits among Han Chinese. METHODS Three tagging single nucleotide polymorphisms (rs1004467, rs17115149, and rs12413409), in the CYP17A1 gene region were selected and genotyped in a case-control study that included 440 diabetes and 1,320 control subjects. Effects of genetic loci were studied using univariate unconditional logistic regression and multivariate logistic regression analysis adjusted for age, sex, family history, body mass index, smoking, and drinking. Bioinformatics analysis was also conducted using the GEO DataSets and PROMO database to gain hints of possible mechanism. RESULTS Rs17115149 and rs12413409 polymorphisms were significantly associated with the risk of T2DM, even after adjusting for age, sex, family history, body mass index, smoking, and drinking. In stratified analyses, rs1004467 and rs12413409 showed significant association with T2DM in the older age group (≥65 years) and, in the case of rs12413409, the risk of T2DM was significant in men but not in women. Rs17115149 had significant association with T2DM in the hypertension subgroup, and rs12413409 in the non-hypertension subgroup. Moreover, rs12413409 showed significant association with plasma glucose levels in the recessive model (P = 0.020) among subjects not taking hypoglycemic measures. Bioinformatics analysis revealed significantly higher CYP17A1 gene expression in T2DM patients compared to healthy controls. Finally, the mutant T allele of the rs17115149 polymorphism allowed binding to the RBP-Jkappa transcription factor. CONCLUSION This is the first report to identify that variants rs1004467, rs17115149, and rs12413409 of CYP17A1, are related to plasma glucose levels and T2DM among Han Chinese. Our results suggest that CYP17A1 might constitute a risk gene for progression to T2DM.
Collapse
Affiliation(s)
- Long Wang
- Department of Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yu-Ming Niu
- Department of Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Yu-Ming Niu, ; Shi-Shi Wu,
| | - Shi-Shi Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- *Correspondence: Yu-Ming Niu, ; Shi-Shi Wu,
| | - Chao Zhang
- Department of Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Zhou
- Research Center for Medicine and Social Development, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Hong-Xia Zuo
- Department of Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Peng Wang
- Department of Clinical Laboratory, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Genome-wide association studies (GWAS) for type 2 diabetes (T2D) risk have identified a large number of genetic loci associated with disease susceptibility. However, progress moving from association signals through causal genes to functional understanding has so far been slow, hindering clinical translation. This review discusses the benefits and limitations of emerging, unbiased approaches for prioritising causal genes at T2D risk loci. RECENT FINDINGS Candidate causal genes can be identified by a number of different strategies that rely on genetic data, genomic annotations, and functional screening of selected genes. To overcome the limitations of each particular method, integration of multiple data sets is proving essential for establishing confidence in the prioritised genes. Previous studies have also highlighted the need to support these efforts through identification of causal variants and disease-relevant tissues. Prioritisation of causal genes at T2D risk loci by integrating complementary lines of evidence promises to accelerate our understanding of disease pathology and promote translation into new therapeutics.
Collapse
Affiliation(s)
- Antje K Grotz
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- National Institute of Health Research Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Soren K Thomsen
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK.
| |
Collapse
|