1
|
Zhou X, Yang N, Xu W, Li X, Spiliopoulou A, Theodoratou E. Associations of genetic factors with vascular diabetes complications: an umbrella review. J Glob Health 2025; 15:04081. [PMID: 40116328 PMCID: PMC11927039 DOI: 10.7189/jogh.15.04081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Abstract
Background To comprehensively assess evidence from published systematic review and meta-analyses (SRMAs) on the genetics of vascular diabetes complications. Methods A systematic literature search conducted in Medline and Embase identified 63 non-overlapping SRMAs. We re-conducted meta-analyses to compare diabetes with and without complications using multiple genetic models; evaluated associations using Venice criteria and Bayesian false-discovery probability (BFDP); and graded as highly credible, credible, and not credible. We also contrasted highly credible and credible associations to recent genome-wide association studies (GWASs). Results Highly credible evidence was discovered for single nucleotide polymorphisms (SNPs) rs1024611 at MCP-1 gene and SNP rs3025039 at VEGF gene with diabetic retinopathy (DR) in type 2 diabetes; SNP rs2268388 at ACACB gene, insertion/deletion (Ins/Del) variant at ACE gene, SNP rs1801133 at MTHFR gene, and SNP rs7903146 at TCF7L2 gene with diabetic kidney disease (DKD) in type 2 diabetes; and SNP rs4880 at SOD2 gene with diabetic peripheral neuropathy (DPN) in type 1 diabetes. Combining type 1 and 2 diabetes, highly credible evidence was discovered for insertion/deletion variant at ACE gene, SNP rs759853 at AKR1B1 gene, SNP rs1044498 at ENPP1 gene and DKD, and SNP rs1617640 at EPO gene for the combined endpoint of DR and DKD. None of these associations was directly replicated in the latest GWASs for DR and DKD, however, another SNP, rs55853916 at TCF7L2 gene had been detected as a GWAS hit for DKD. Conclusions This umbrella review rigorously assessed evidence on the genetics of vascular diabetes complications, complemented findings in recent GWASs and yielded insight into the optimal selection of genetic models for the design of GWASs on vascular diabetes complications. Mechanistic or bioinformatic studies are warranted to further assess the role of these genes in the pathology of vascular diabetes complications and their potential as drug targets. Registration PROSPERO: CRD42022384423.
Collapse
Affiliation(s)
- Xuan Zhou
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Nan Yang
- Department of Big Data in Health Science School of Public Health, and Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Xu
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, and Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Athina Spiliopoulou
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Cespuglio R, Gorlova A, Zabegalov K, Chaprov K, Svirin E, Sitdikova K, Burova A, Shulgin B, Lebedeva K, Deikin AV, Morozov S, Strekalova T. SERT-Deficient Mice Fed Western Diet Reveal Altered Metabolic and Pro-Inflammatory Responses of the Liver: A Link to Abnormal Behaviors. FRONT BIOSCI-LANDMRK 2025; 30:26778. [PMID: 39862090 DOI: 10.31083/fbl26778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/15/2024] [Accepted: 11/28/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND The inheritance of the short SLC6A4 allele, encoding the serotonin transporter (SERT) in humans, increases susceptibility to neuropsychiatric and metabolic disorders, with aging and female sex further exacerbating these conditions. Both central and peripheral mechanisms of the compromised serotonin (5-HT) system play crucial roles in this context. Previous studies on SERT-deficient (Sert-/-) mice, which model human SERT deficiency, have demonstrated emotional and metabolic disturbances, exacerbated by exposure to a high-fat Western diet (WD). Growing evidence suggests the significance of hepatic regulatory mechanisms in the neurobiology of central nervous system disorders, supporting the 'liver-brain' concept. However, the relationship between aberrant behavior and hepatic alterations under conditions of SERT deficiency remains poorly investigated. METHODS One-year-old female Sert-/- mice and their wild-type (WT) littermates were subjected to a control diet (CD) or the WD for a duration of three weeks. The WD had a higher caloric content and was characterized by an elevated saturated fat content (21%) compared to the CD (4.5%) and contained 0.2% cholesterol. Mice were evaluated for anxiety-like behavior, exploration and locomotor activity in the open field test, as well as glucose tolerance and histological indicators of hepatic steatosis. Hepatic pro-inflammatory and metabolism-related gene expression and markers of nitrosative stress, were analyzed utilizing real-time polymerase chain reaction (RT-PCR) and correlated with behavioral and histological outcomes. RESULTS In comparison to unchallenged mice, Sert-/-/WD mutants, but not the WT/WD group, had increased locomotion and anxiety-like behavior, increased hepatic steatosis, and elevated expression of insulin receptor B and pro-inflammatory cytokines interleukin-1β (Il-1β) and Tnf, as well as decreased expression of leptin receptor B. The two genotypes displayed distinct gene expression patterns of nitric oxide (NO)-related molecules inducible NO synthase (iNos) and arginase (Arg2), insulin receptor-related signaling factors: cluster of differentiation 36 (Cd36), ecto-nucleotide pyrophosphatase/phosphodiesterase (Enpp), protein tyrosine phosphatase N1 (Ptpn1), cytochrome P450 omega-hydroxylase 4A14 (Cyp4a14), acyl-CoA synthetase 1 (Acsl1) and phosphatase and tensin homolog (Pten). Furthermore, there were profound differences in correlations between molecular, histological, and behavioral measurements across the two genotypes. CONCLUSIONS Our findings suggest that the genetic deficiency of SERT results in abnormal hepatic pro-inflammatory and metabolic adaptations in response to WD. The significant correlations observed between behavioral measures and pro-inflammatory and metabolic alterations in WD-fed mice suggest the importance of liver-brain interactions and their role in the aberrant behaviors exhibited by Sert-/- mutants. This study presents the first evidence that altered liver functions are associated with pathological behaviors arising from genetic SERT deficiency.
Collapse
Affiliation(s)
- Raymond Cespuglio
- Neuroscience Research Center of Lyon, Claude-Bernard Lyon-1 University, 69675 Bron, France
| | - Anna Gorlova
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | | | - Kirill Chaprov
- National Laboratory of Astana, Nazarbaev University, 010000 Astana, Kazakhstan
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia
| | - Evgeniy Svirin
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Kseniia Sitdikova
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Alisa Burova
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Boris Shulgin
- Laboratory of Engineering Profile Physical and Chemical Methods of Analysis, Korkyt Ata Kyzylorda State University, 120014 Kyzylorda, Kazakhstan
- Department of Normal Physiology, Sechenov University, 117198 Moscow, Russia
| | - Ksenia Lebedeva
- Department of Normal Physiology, Sechenov University, 117198 Moscow, Russia
| | - Alexei V Deikin
- Laboratory of Genetic Technology and gene editing for Biomedicine and Veterinary, National Research Belgorod state University, 308015 Belgorod, Russia
| | - Sergey Morozov
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Hospital Würzburg, 97080 Würzburg, Germany
- Maastricht University, Department of Psychiatry and Neuropsychology, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
3
|
Nair N, Chang J, Evans MS. Congenital hypopigmentation, hyperpigmentation, and punctate palmoplantar keratoderma. Pediatr Dermatol 2022; 39:476-477. [PMID: 36156296 DOI: 10.1111/pde.14984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/01/2022] [Accepted: 03/06/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Nikhil Nair
- University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas, USA
| | - Joshua Chang
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Megan S Evans
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
4
|
Felisbino K, Granzotti JG, Bello-Santos L, Guiloski IC. Nutrigenomics in Regulating the Expression of Genes Related to Type 2 Diabetes Mellitus. Front Physiol 2021; 12:699220. [PMID: 34366888 PMCID: PMC8334860 DOI: 10.3389/fphys.2021.699220] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
Nutrigenomics is the study of the gene-nutrient interaction and it indicates that some nutrients, called bioactive compounds, can mold the genetic expression or change the nucleotide chain. Polyphenols are secondary metabolites found in plants that are regularly consumed in functional foods and help prevent or delay the onset of type 2 diabetes mellitus (T2DM) and its complications. This article objected to review studies about the interaction of diet with polyphenols and Mediterranean diet in the expression of human genes related to T2DM. Resveratrol acts as an antioxidant, anti-inflammatory, and increases mitochondrial function. Regular consumption of quercetin resulted in improvement of hypertension and suppression of diabetes-induced vasoconstriction. Genistein also showed positive results in T2DM, such as increased cell mass and improved glucose tolerance and insulin levels. Catechins showed efficiency in inducing genes in triacylglycerol biosynthesis, inhibition of fatty acids and cholesterol, and resulting in their participation in mitigating complications of diabetes. Lastly, curcumin was demonstrated to be a protector of the pancreatic islets against streptozotocin-induced oxidative stress. Growing evidence suggest that bioactive compounds such as polyphenols have an important role in T2DM and the prevention and treatment of its complication, as they cause activation or inhibition of related genes.
Collapse
Affiliation(s)
- Karoline Felisbino
- Centro de Ensino Superior de Maringá (CESUMAR), Curitiba, Brazil
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Brazil
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba, Brazil
| | | | | | - Izonete Cristina Guiloski
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Brazil
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba, Brazil
| |
Collapse
|
5
|
Wu X, Di F, Shen S, Wang S, Li Q, Dong Z, Guan J, He J, Wang Y. Levels of serum ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) predicts severity of abdominal aortic calcification in end-stage renal disease patients receiving regular dialysis. Hemodial Int 2021; 26:23-29. [PMID: 34235847 DOI: 10.1111/hdi.12969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the correlation between serum ectonucleotide pyrophosphatase/phosphodiesterase-1 (ENPP1) level and severity of abdominal vascular calcification in end-stage renal disease (ESRD) patients receiving dialysis. METHODS A total of 124 patients were consecutively enrolled into the study in our local institution. Based on the Kidney Disease Improving Global Outcomes (KDIGO) guidelines and recommendations, abdomen lateral X-ray was used to determine abdominal aortic calcification score (AACS) for each patient at enrollment. Patients were divided into three groups based on AACS: no or mild calcification group, moderate calcification group, and severe calcification group. The relationships between ENPP1 levels and AACS were assessed by Spearman analysis and the value of ENPP1 in predicting severity of abdominal aortic calcification was evaluated by receiver operating characteristic (ROC). RESULTS The level of ENPP1 in dialysis patients was (7.68 ± 1.67) ng/ml. There was no significant difference in serum ENPP1 level between peritoneal dialysis patients and hemodialysis patients (p > 0.05). The AACS of dialysis patients was negatively correlated with ENPP1 value (r = -0.70). Compared to no/mild calcification patients, the levels of serum ENPP1 in patients with moderate/severe calcification were decreased significantly (p < 0.01). The severity of vascular calcification was correlated with serum ENPP1 value, the severer the vascular calcification, the lower the serum ENPP1 level, and the difference was statistically significant (all p < 0.05). The area under ROC curve of ENPP1 was 0.90, the corresponding sensitivity was 0.86, and the specificity was 0.87. CONCLUSION Levels of serum ENPP1 in non-diabetic ESRD patients are negatively related to the severity of abdominal aortic vascular calcification.
Collapse
Affiliation(s)
- Xiujuan Wu
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Feng Di
- Department of Respiratory, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Shuijuan Shen
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Shimin Wang
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Qinghua Li
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Zhichao Dong
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Jichao Guan
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Jianling He
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Yu Wang
- Department of Nephrology, Shaoxing People's Hospital, (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| |
Collapse
|
6
|
Arikoglu H, Erkoc-Kaya D, Ipekci SH, Gokturk F, Iscioglu F, Korez MK, Baldane S, Gonen MS. Type 2 diabetes is associated with the MTNR1B gene, a genetic bridge between circadian rhythm and glucose metabolism, in a Turkish population. Mol Biol Rep 2021; 48:4181-4189. [PMID: 34117605 DOI: 10.1007/s11033-021-06431-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/21/2021] [Indexed: 01/06/2023]
Abstract
Type 2 diabetes (T2D) is a complicated public health problem in Turkey as well as worldwide. Genome-wide approaches have been guiding in very challenging situations, such as the elucidation of genetic variations underlying complex diseases such as T2D. Despite intensive studies worldwide, few studies have determined the genetic susceptibility to T2D in Turkish populations. In this study, we investigated the effect of genes that are strongly associated with T2D in genome-wide association (GWA) studies, including MTNR1B, CDKAL1, THADA, ADAMTS9 and ENPP1, on T2D and its characteristic traits in a Turkish population. In 824 nonobese individuals (454 T2D patients and 370 healthy individuals), prominent variants of these GWA genes were genotyped by real-time PCR using the LightSNiP Genotyping Assay System. The SNP rs1387153 C/T, which is located 28 kb upstream of the MTNR1B gene, was significantly associated with T2D and fasting blood glucose levels (P < 0.05). The intronic SNP rs10830963 C/G in the MTNR1B gene was not associated with T2D, but it was associated with fasting blood glucose, HbA1C and LDL levels (P < 0.05). The other important GWA loci investigated in our study were not found to be associated with T2D or its traits. Only the SNP rs1044498 (A/C variation) in the ENPP1 gene was determined to be related to fasting blood glucose (P < 0.05). Our study suggests, consistent with the literature, that the MTNR1B locus, which has a prominent role in glucose regulation, is associated with T2D development by affecting blood glucose levels in our population.
Collapse
Affiliation(s)
- Hilal Arikoglu
- Department of Medical Biology, Faculty of Medicine, Selcuk University, Konya, Turkey.
| | - Dudu Erkoc-Kaya
- Department of Medical Biology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Suleyman Hilmi Ipekci
- Department of Endocrinology and Metabolic Diseases, Hisar Hospital Intercontinental, Istanbul, Turkey
| | - Fatma Gokturk
- Department of Medical Biology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Funda Iscioglu
- Department of Statistics, Faculty of Science, Ege University, Izmir, Turkey
| | - Muslu Kazim Korez
- Department of Biostatistics, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Suleyman Baldane
- Department of Endocrinology and Metabolic Diseases, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Mustafa Sait Gonen
- Department of Endocrinology and Metabolic Diseases, Faculty of Cerrahpasa Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
7
|
Luo X, Cui H, Cai L, Zhu W, Yang WC, Patrick M, Zhu S, Huang J, Yao X, Yao Y, He Y, Ji Y. Selection of a Clinical Lead TCR Targeting Alpha-Fetoprotein-Positive Liver Cancer Based on a Balance of Risk and Benefit. Front Immunol 2020; 11:623. [PMID: 32425926 PMCID: PMC7203609 DOI: 10.3389/fimmu.2020.00623] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with a poor prognosis and limited therapeutic options. Alpha-fetoprotein (AFP), an established clinical biomarker of HCC, has been employed as an attractive target for T cell-based immunotherapy against this disease given its high expression in the tumor and restricted expression in normal tissues. We have identified a number of T cell receptors (TCRs) recognizing the HLA-A*02:01 restricted AFP158-166 peptide FMNKFIYEI, providing a TCR candidate pool for identifying TCRs with optimal clinical benefit. To select the ideal AFP TCR for clinical use, we evaluated the efficacy and safety profile of 7 TCRs by testing their potency toward AFP-expressing HCC cells and their specificity based upon reactivity to normal and transformed cells covering a wide variety of primary cell types and HLA serotypes. Furthermore, we assessed their cross-reactivity to potential protein candidates in the human genome by an extensive alanine scan (X-scan). We first selected three TCR candidates based on the in vitro anti-tumor activity. Next we eliminated two potential cross-reactive TCRs based on their reactivity against normal and transformed cells covering a variety of primary cell types and HLA serotypes, respectively. We then excluded the potential cross-reactivity of the selected TCR with a protein candidate identified by X-scan. At present we have selected an AFP TCR with the optimal affinity, function, and safety profile, bearing properties that are expected to allow AFP TCR redirected T cells to specifically differentiate between AFP levels on tumor and normal tissues. An early phase clinical trial using T cells transduced with this TCR to treat HCC patients (NCT03971747) has been initiated.
Collapse
Affiliation(s)
- Xiaobing Luo
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Huijuan Cui
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Lun Cai
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA, United States
| | - Wei Zhu
- CodexSage LLC., Germantown, MD, United States
| | - Wei-Chih Yang
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Michael Patrick
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Shigui Zhu
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Jiaqi Huang
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Xin Yao
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Yihong Yao
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yun Ji
- Cellular Biomedicine Group Inc., Gaithersburg, MD, United States
| |
Collapse
|
8
|
Yahaya TO, Salisu TF. A Review of Type 2 Diabetes Mellitus Predisposing Genes. Curr Diabetes Rev 2019; 16:52-61. [PMID: 30514191 DOI: 10.2174/1573399815666181204145806] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/22/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Scientists are considering the possibility of treating diabetes mellitus (DM) using a personalized approach in which various forms of the diseases will be treated based on the causal gene and its pathogenesis. To this end, scientists have identified mutations in certain genes as probable causes of Type 2 diabetes mellitus (T2DM) with diverse mechanisms. AIM This review was aimed at articulating already identified T2DM genes with their mechanisms of action and phenotypic presentations for the awareness of all stakeholders. METHOD The Google search engine was used to retrieve relevant information on the subject from reliable academic databases such as PubMed, Medline, and Google Scholar, among others. RESULTS At least seventy (70) genes are currently being suspected in the biogenesis of T2DM. However, mutations in, or variants of KCNJ11, PPARG, HNF1B and WFS1 genes, are the most suspected and reported in the pathogenesis of the disease. Mutations in these genes can cause disruption of insulin biosynthesis through the destruction of pancreatic beta cells, change of beta cell morphology, destruction of insulin receptors, among others. These cellular events may lead to insulin resistance and hyperglycemia and, along with environmental triggers such as obesity and overweight, culminate in T2DM. It was observed that each identified gene has its distinct mechanism by which it interacts with other genes and environmental factors to cause T2DM. CONCLUSION Healthcare providers are advised to formulate T2DM drugs or treatment by targeting the causal genes along with their mechanisms.
Collapse
Affiliation(s)
- Tajudeen O Yahaya
- Department of Biology, Federal University Birnin Kebbi, Kebbi State, Nigeria
| | - Titilola F Salisu
- Department of Cell Biology and Genetics, University of Lagos, Lagos, Nigeria
| |
Collapse
|
9
|
ATP-degrading ENPP1 is required for survival (or persistence) of long-lived plasma cells. Sci Rep 2017; 7:17867. [PMID: 29259245 PMCID: PMC5736562 DOI: 10.1038/s41598-017-18028-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/05/2017] [Indexed: 02/05/2023] Open
Abstract
Survival of antibody-secreting plasma cells (PCs) is vital for sustained antibody production. However, it remains poorly understood how long-lived PCs (LLPCs) are generated and maintained. Here we report that ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is preferentially upregulated in bone marrow LLPCs compared with their splenic short-lived counterparts (SLPCs). We studied ENPP1-deficient mice (Enpp1−/−) to determine how the enzyme affects PC biology. Although Enpp1−/− mice generated normal levels of germinal center B cells and plasmablasts in periphery, they produced significantly reduced numbers of LLPCs following immunization with T-dependent antigens or infection with plasmodium C. chabaudi. Bone marrow chimeric mice showed B cell intrinsic effect of ENPP1 selectively on generation of bone marrow as well as splenic LLPCs. Moreover, Enpp1−/− PCs took up less glucose and had lower levels of glycolysis than those of wild-type controls. Thus, ENPP1 deficiency confers an energetic disadvantage to PCs for long-term survival and antibody production.
Collapse
|
10
|
Integrating genome and transcriptome profiling for elucidating the mechanism of muscle growth and lipid deposition in Pekin ducks. Sci Rep 2017. [PMID: 28630415 PMCID: PMC5476626 DOI: 10.1038/s41598-017-04178-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Muscle growth and lipid deposition are co-ordinately regulated processes. Cherry Valley Pekin duck is a lean-type duck breed with high growth rate, whereas the native Pekin duck of China has high lipid deposition. Phenotypic analysis showed that native Pekin ducks have smaller fibre diameter and larger density in the breast muscle at 3 weeks of age and higher intramuscular fat content at 6 weeks of age than those in Cherry Valley Pekin ducks. We detected 17 positively selected genes (PSGs) by comparing genes mainly involved with muscle organ development, muscle contraction, peroxisome proliferator activated receptor signalling pathway, and fatty acid metabolism. In all, 52 and 206 differentially expressed genes (DEGs) were identified in transcriptomic comparisons between the two breeds at 3 and 6 weeks of age, respectively, which could potentially affect muscle growth and lipid deposition. Based on the integration of PSGs and DEGs and their functional annotations, we found that 11 and 10 genes were correlated with muscle growth and lipid deposition, respectively. Identification of candidate genes controlling quantitative traits of duck muscle might aid in elucidating the mechanisms of muscle growth and lipid deposition and could help in improving duck breeding.
Collapse
|
11
|
Chen S, Han J, Liu Y. Dual Opposing Roles of Metallothionein Overexpression in C57BL/6J Mouse Pancreatic β-Cells. PLoS One 2015; 10:e0137583. [PMID: 26335571 PMCID: PMC4559390 DOI: 10.1371/journal.pone.0137583] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/18/2015] [Indexed: 11/20/2022] Open
Abstract
Background Growing evidence indicates that oxidative stress (OS), a persistent state of excess amounts of reactive oxygen species (ROS) along with reactive nitrogen species (RNS), plays an important role in insulin resistance, diabetic complications, and dysfunction of pancreatic β-cells. Pancreatic β-cells contain exceptionally low levels of antioxidant enzymes, rendering them susceptible to ROS-induced damage. Induction of antioxidants has been proposed to be a way for protecting β-cells against oxidative stress. Compared to other antioxidants that act against particular β-cell damages, metallothionein (MT) is the most effective in protecting β-cells from several oxidative stressors including nitric oxide, peroxynitrite, hydrogen peroxide, superoxide and streptozotocin (STZ). We hypothesized that MT overexpression in pancreatic β-cells would preserve β-cell function in C57BL/6J mice, an animal model susceptible to high fat diet-induced obesity and type 2 diabetes. Research Design and Methods The pancreatic β-cell specific MT overexpression was transferred to C57BL/6J background by backcrossing. We studied transgenic MT (MT-tg) mice and wild-type (WT) littermates at 8 weeks and 18 weeks of age. Several tests were performed to evaluate the function of islets, including STZ in vivo treatment, intraperitoneal glucose tolerance tests (IPGTT) and plasma insulin levels during IPGTT, pancreatic and islet insulin content measurement, insulin secretion, and islet morphology assessment. Gene expression in islets was performed by quantitative real-time PCR and PCR array analysis. Protein levels in pancreatic sections were evaluated by using immunohistochemistry. Results The transgenic MT protein was highly expressed in pancreatic islets. MT-tg overexpression significantly protected mice from acute STZ-induced ROS at 8 weeks of age; unexpectedly, however, MT-tg impaired glucose stimulated insulin secretion (GSIS) and promoted the development of diabetes. Pancreatic β-cell function was significantly impaired, and islet morphology was also abnormal in MT-tg mice, and more severe damage was detected in males. The unique gene expression pattern and abnormal protein levels were observed in MT-tg islets. Conclusions MT overexpression protected β-cells from acute STZ-induced ROS damages at young age, whereas it impaired GSIS and promoted the development of diabetes in adult C57BL/6J mice, and more severe damage was found in males.
Collapse
Affiliation(s)
- Suqin Chen
- The Research Institute for Children, Children’s Hospital at New Orleans, New Orleans, Louisiana, United States of America
- Department of Medical Genetics, Zhongshan Medical College, Sun Yat-sen University, Guangzhou, Guangdong Province, People’s Republic of China
- * E-mail:
| | - Junying Han
- The Research Institute for Children, Children’s Hospital at New Orleans, New Orleans, Louisiana, United States of America
| | - Yeqi Liu
- The Research Institute for Children, Children’s Hospital at New Orleans, New Orleans, Louisiana, United States of America
- Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| |
Collapse
|
12
|
Xue B, Nie J, Wang X, DuBois DC, Jusko WJ, Almon RR. Effects of High Fat Feeding on Adipose Tissue Gene Expression in Diabetic Goto-Kakizaki Rats. GENE REGULATION AND SYSTEMS BIOLOGY 2015; 9:15-26. [PMID: 26309393 PMCID: PMC4533846 DOI: 10.4137/grsb.s25172] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/24/2015] [Accepted: 06/17/2015] [Indexed: 12/15/2022]
Abstract
Development and progression of type 2 diabetes is a complex interaction between genetics and environmental influences. High dietary fat is one environmental factor that is conducive to the development of insulin-resistant diabetes. In the present report, we compare the responses of lean poly-genic, diabetic Goto-Kakizaki (GK) rats to those of control Wistar-Kyoto (WKY) rats fed a high fat diet from weaning to 20 weeks of age. This comparison included a wide array of physiological measurements along with gene expression profiling of abdominal adipose tissue using Affymetrix gene array chips. Animals of both strains fed a high fat diet or a normal diet were sacrificed at 4, 8, 12, 16, and 20 weeks for this comparison. The microarray analysis revealed that the two strains developed different adaptations to increased dietary fat. WKY rats decrease fatty acid synthesis and lipogenic processes whereas GK rats increase lipid elimination. However, on both diets the major differences between the two strains remained essentially the same. Specifically relative to the WKY strain, the GK strain showed lipoatrophy, chronic inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Bai Xue
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jing Nie
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Xi Wang
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Debra C DuBois
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, USA
| | - Richard R Almon
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, USA
| |
Collapse
|
13
|
Huesa C, Zhu D, Glover JD, Ferron M, Karsenty G, Milne EM, Millan JL, Ahmed SF, Farquharson C, Morton NM, MacRae VE. Deficiency of the bone mineralization inhibitor NPP1 protects mice against obesity and diabetes. Dis Model Mech 2014; 7:1341-50. [PMID: 25368121 PMCID: PMC4257003 DOI: 10.1242/dmm.017905] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The emergence of bone as an endocrine regulator has prompted a re-evaluation of the role of bone mineralization factors in the development of metabolic disease. Ectonucleotide pyrophosphatase/phosphodiesterase-1 (NPP1) controls bone mineralization through the generation of pyrophosphate, and levels of NPP1 are elevated both in dermal fibroblast cultures and muscle of individuals with insulin resistance. We investigated the metabolic phenotype associated with impaired bone metabolism in mice lacking the gene that encodes NPP1 (Enpp1−/− mice). Enpp1−/− mice exhibited mildly improved glucose homeostasis on a normal diet but showed a pronounced resistance to obesity and insulin resistance in response to chronic high-fat feeding. Enpp1−/− mice had increased levels of the insulin-sensitizing bone-derived hormone osteocalcin but unchanged insulin signalling within osteoblasts. A fuller understanding of the pathways of NPP1 could inform the development of novel therapeutic strategies for treating insulin resistance.
Collapse
Affiliation(s)
- Carmen Huesa
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - Dongxing Zhu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK.
| | - James D Glover
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - Mathieu Ferron
- Integrative and Molecular Physiology Research Unit Institut de Recherches Cliniques de Montréal (IRCM), 110 Avenue des Pins Ouest - Laboratory 2750, Montréal, QC H2W 1R7, Canada
| | - Gerard Karsenty
- Department of Developmental Genetics, Columbia University, NY 10032, USA
| | - Elspeth M Milne
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - José Luis Millan
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - S Faisal Ahmed
- Developmental Medicine, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Colin Farquharson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - Nicholas M Morton
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| |
Collapse
|
14
|
Honardoost M, reza Sarookhani M, Arefian E, Soleimani M. Insulin Resistance Associated Genes and miRNAs. Appl Biochem Biotechnol 2014; 174:63-80. [DOI: 10.1007/s12010-014-1014-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 06/15/2014] [Indexed: 01/05/2023]
|
15
|
Coletta P, Barbarossa G, Pergolini D, D'Erasmo L, Renzi A, Mercuri L, Anatra MG, Ciociola E, Verrienti A, Maranghi M. Genetic determinants and early carotid atherosclerosis: is there a role for the ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP-1) K121Q polymorphism? Preliminary results in non diabetic individuals. Endocrine 2012; 42:749-51. [PMID: 22899099 DOI: 10.1007/s12020-012-9771-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/31/2012] [Indexed: 11/25/2022]
|
16
|
Abstract
Polygenic type 2 diabetes mellitus (T2DM) is a multi-factorial disease due to the interplay between genes and the environment. Over the years, several genes/loci have been associated with this type of diabetes, with the majority of them being related to β cell dysfunction. In this review, the available information on how polymorphisms in T2DM-associated genes/loci do directly affect the properties of human islet cells are presented and discussed, including some clinical implications and the role of epigenetic mechanisms.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy.
| | | | | | | | | |
Collapse
|
17
|
Shi X, Wang L, Jin F, Sun J, Sun L, Tang L, Yang Z. The ENPP1 K121Q polymorphism is not associated with type 2 diabetes in northern Chinese. Acta Diabetol 2011; 48:303-310. [PMID: 21452007 DOI: 10.1007/s00592-011-0281-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 03/15/2011] [Indexed: 01/07/2023]
Abstract
The K121Q polymorphism of the ectoenzyme nucleotide pyrophosphate phosphodiesterase 1 (ENPP1) gene has been studied in relation to insulin resistance, type 2 diabetes, and obesity, and conflicting results were observed in various populations. The purpose of the present study was to investigate the prevalence of K121Q polymorphism of ENPP1 gene and to clarify whether this polymorphism is associated with type 2 diabetes susceptibility in northern Chinese population. We studied the association of the ENPP1 K121Q polymorphism with type 2 diabetes (T2D) in 639 unrelated patients and 885 control subjects with normal glucose tolerance of northern China. The patients were diagnosed in accordance with the guidelines of the American Diabetes Association (ADA). Genotypes were determined by the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. The distribution of KK, KQ, and QQ genotypes among patients was 79.5, 19.2, and 1.3%, similar to that of the control group (79.2, 20.1, and 0.7%). After readjusting for the confounding effects of age, gender, and BMI, no significant effect of genotypes on T2D was found for any of the genetic models tested (recessive model, dominant model, or additive model). All clinical characteristics tested were similar among the different genotypes, and no significant associations were observed both in T2D patients and in controls. When subgroup analyses of T2D patients and non-diabetic controls were stratified according to BMI and waist circumference, the variant was still not associated with T2D. The results showed that the ENPP1 K121Q polymorphism is not associated with genetic susceptibility of type 2 diabetes in the northern Chinese population.
Collapse
Affiliation(s)
- Xiaohong Shi
- Key Laboratory of Geriatrics, Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing, 100730, China.
| | - Li Wang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Junfeng Sun
- Key Laboratory of Geriatrics, Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing, 100730, China
| | - Liang Sun
- Key Laboratory of Geriatrics, Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing, 100730, China
| | - Lei Tang
- Key Laboratory of Geriatrics, Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing, 100730, China
| | - Ze Yang
- Key Laboratory of Geriatrics, Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing, 100730, China.
| |
Collapse
|
18
|
Choquet H, Meyre D. Molecular basis of obesity: current status and future prospects. Curr Genomics 2011; 12:154-68. [PMID: 22043164 PMCID: PMC3137001 DOI: 10.2174/138920211795677921] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 03/30/2011] [Accepted: 03/31/2011] [Indexed: 12/15/2022] Open
Abstract
Obesity is a global health problem that is gradually affecting each continent of the world. Obesity is a heterogeneous disorder, and the biological causes of obesity are complex. The rapid increase in obesity prevalence during the past few decades is due to major societal changes (sedentary lifestyle, over-nutrition) but who becomes obese at the individual level is determined to a great extent by genetic susceptibility. In this review, we evidence that obesity is a strongly heritable disorder, and provide an update on the molecular basis of obesity. To date, nine loci have been involved in Mendelian forms of obesity and 58 loci contribute to polygenic obesity, and rare and common structural variants have been reliably associated with obesity. Most of the obesity genes remain to be discovered, but promising technologies, methodologies and the use of “deep phenotyping” lead to optimism to chip away at the ‘missing heritability’ of obesity in the near future. In the longer term, the genetic dissection of obesity will help to characterize disease mechanisms, provide new targets for drug design, and lead to an early diagnosis, treatment, and prevention of obesity.
Collapse
Affiliation(s)
- Hélène Choquet
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, Emeryville, CA 94608, USA
| | | |
Collapse
|
19
|
Schäfer SA, Machicao F, Fritsche A, Häring HU, Kantartzis K. New type 2 diabetes risk genes provide new insights in insulin secretion mechanisms. Diabetes Res Clin Pract 2011; 93 Suppl 1:S9-24. [PMID: 21864758 DOI: 10.1016/s0168-8227(11)70008-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes results from the inability of beta cells to increase insulin secretion sufficiently to compensate for insulin resistance. Insulin resistance is thought to result mainly from environmental factors, such as obesity. However, there is compelling evidence that the decline of both insulin sensitivity and insulin secretion have also a genetic component. Recent genome-wide association studies identified several novel risk genes for type 2 diabetes. The vast majority of these genes affect beta cell function by molecular mechanisms that remain unknown in detail. Nevertheless, we and others could show that a group of genes affect glucose-stimulated insulin secretion, a group incretin-stimulated insulin secretion (incretin sensitivity or secretion) and a group proinsulin-to-insulin conversion. The most important so far type 2 diabetes risk gene, TCF7L2, interferes with all three mechanisms. In addition to advancing knowledge in the pathophysiology of type 2 diabetes, the discovery of novel genetic determinants of diabetes susceptibility may help understanding of gene-environment, gene-therapy and gene-gene interactions. It was also hoped that it could make determination of the individual risk for type 2 diabetes feasible. However, the allelic relative risks of most genetic variants discovered so far are relatively low. Thus, at present, clinical criteria assess the risk for type 2 diabetes with greater sensitivity and specificity than the combination of all known genetic variants.
Collapse
Affiliation(s)
- Silke A Schäfer
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University of Tübingen, Germany
| | | | | | | | | |
Collapse
|
20
|
Abbasi S, Shin DM, Beaty N, Masiuk M, Chen S, Gonzalez-Garcia I, Zhao M, Goding J, Morse HC, Wang H. Characterization of monoclonal antibodies to the plasma cell alloantigen ENPP1. Hybridoma (Larchmt) 2011; 30:11-7. [PMID: 21466281 DOI: 10.1089/hyb.2010.0089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1) has documented roles in mineralization, nucleotide recycling, and insulin resistance. While ENPP1 was first identified as an alloantigen on mouse plasma cells (PCs), later studies revealed expression in many tissues. Previously described monoclonal antibodies against ENPP1 expressed at the cell surface recognized cells only from mice bearing the a allotype, ENPP1(a), precluding studies of mice bearing the alternative allele, ENPP1(b). Here, we characterize a novel anti-ENPP1 monoclonal antibody that recognizes both alleles and can be used for flow cytometry.
Collapse
Affiliation(s)
- Sadia Abbasi
- Laboratory of Immunopathology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5640 Fishers Lane, Rockville, MD 20850, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Genetic and Clinical Risk Factors of New-Onset Diabetes After Transplantation in Hispanic Kidney Transplant Recipients. Transplantation 2011; 91:1114-9. [DOI: 10.1097/tp.0b013e31821620f9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
22
|
Radha V, Kanthimathi S, Mohan V. Genetics of Type 2 diabetes in Asian Indians. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/dmt.11.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
|
23
|
Lee JE, Choi YK, Seo HA, Jeon JH, Jeong JY, Moon SS, Kim JG, Kim BW, Kim SW, Kim JY, Lee IK. Impact of ENPP1 and MMP3 gene polymorphisms on aortic calcification in patients with type 2 diabetes in a Korean population. Diabetes Res Clin Pract 2010; 88:87-96. [PMID: 20092902 DOI: 10.1016/j.diabres.2010.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/17/2009] [Accepted: 01/04/2010] [Indexed: 01/17/2023]
Abstract
AIMS We investigated whether gene polymorphisms of Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) and matrix metalloproteinase 3 (MMP3) are associated with increased vascular calcification in patients with type 2 diabetes (T2D) and evaluated whether serum MMP3 and osteoprotegerin (OPG) levels are related to calcification. METHODS This study included 464 subjects: 269 patients with T2D and 195 healthy controls in South Korea. We genotyped subjects for four single nucleotide polymorphisms (SNPs): ENPP1 K121Q, ENPP1 A/G+1044TGA, MMP3 -709A>G and MMP3 -1475G>A. The presence or absence of calcifications in the aortic arch was assessed by plain chest radiography. RESULTS The SNPs ENPP1 K121Q and MMP3 -709A>G showed significant associations with T2D (P=0.001 and P=0.004). The SNP ENPP1 K121Q showed a significant association with aortic arch calcification in T2D (P=0.036). Serum OPG levels were significantly higher in T2D patients than in the control group (P<0.001). However, serum MMP3 levels were significantly lower in T2D patients than in the control group (P<0.001). CONCLUSIONS Our study demonstrates that the ENPP1 K121Q and MMP3 -709A>G polymorphisms are associated with T2D, and that the ENPP1 Q allele is associated with increased aortic arch calcification in a Korean population.
Collapse
Affiliation(s)
- Jung-Eun Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Korostishevsky M, Cohen Z, Malkin I, Ermakov S, Yarenchuk O, Livshits G. Morphological and biochemical features of obesity are associated with mineralization genes' polymorphisms. Int J Obes (Lond) 2010; 34:1308-18. [PMID: 20231843 DOI: 10.1038/ijo.2010.53] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) was recently extensively studied as a candidate gene for obesity phenotypes. As the human homologue of the mouse progressive ankylosis (ANKH) and alkaline phosphatase (ALPL) are known functional partners of ENPP1 in bone mineralization, we hypothesized that these genes may also be jointly involved in determining obesity features. AIM To examine the effects of the three genes, possible gene-sex and gene-gene interactions on variability of four obesity phenotypes: the body mass index (BMI), the waist-hip ratio (WHR), the epidermal growth factor receptor (EGFR), and leptin. SUBJECTS AND METHODS In all, 962 healthy individuals from 230 families were genotyped for 45 single nucleotide polymorphisms (SNPs). The association analysis was performed using two family based association tests (family based association test and pedigree disequilibrium test). The combined P-values of the two tests were estimated by Monte-Carlo simulations. Relative magnitude of the genetic and familial effects, gene-sex and gene-gene interactions were assessed using variance component models. RESULTS Associations were observed between ENPP1 polymorphisms and BMI (P=0.0037) and leptin (P=0.0068). ALPL markers were associated with WHR (P=0.0026) and EGFR (P=0.0001). The ANKH gene was associated with all four studied obesity-related traits (P<0.0184), and its effects were modulated by sex. Gene-gene interactions were not detected. CONCLUSION The observed pattern of association signals indicates that ANKH may have a generalized effect on adipose tissue physiology, whereas ENPP1 and ALPL affect distinct obesity features. The joint analysis of related genes and integration of the results obtained by different methods used in this research should benefit other studies of similar design.
Collapse
Affiliation(s)
- M Korostishevsky
- Department of Anatomy and Anthropology, Human Population Biology Research Unit, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
25
|
Prudente S, Morini E, Trischitta V. Insulin signaling regulating genes: effect on T2DM and cardiovascular risk. Nat Rev Endocrinol 2009; 5:682-93. [PMID: 19924153 DOI: 10.1038/nrendo.2009.215] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a complex disorder that has a heterogeneous genetic and environmental background. In this Review, we discuss the role of relatively infrequent polymorphisms of genes that regulate insulin signaling (including the K121Q polymorphism of ENPP1, the G972R polymorphism of IRS1 and the Q84R polymorphism of TRIB3) in T2DM and other conditions related to insulin resistance. The biological relevance of these three polymorphisms has been very thoroughly characterized both in vitro and in vivo and the available data indicate that they all affect insulin signaling and action as well as insulin secretion. They also affect insulin-mediated regulation of endothelial cell function. In addition, several reports indicate that the effects of all three polymorphisms on the risk of T2DM and cardiovascular diseases related to insulin resistance depend on the clinical features of the individual, including their body weight and age at disease onset. Thus, these polymorphisms might be used to demonstrate how difficult it is to ascertain the contribution of relatively infrequent genetic variants with heterogeneous effects on disease susceptibility. Unraveling the role of such variants might be facilitated by improving disease definition and focusing on specific subsets of patients.
Collapse
Affiliation(s)
- Sabrina Prudente
- IRCCS Casa Sollievo della Sofferenza, Mendel Institute, Rome, Italy
| | | | | |
Collapse
|
26
|
Bacci S, Di Paola R, Menzaghi C, Di Fulvio P, Di Silvestre S, Pellegrini F, Baratta R, Marucci A, Mastroianno S, Fini G, Formoso G, Consoli A, Perticone F, Frittitta L, Pandolfi A, Trischitta V. ENPP1
Q121 Variant, Increased Pulse Pressure and Reduced Insulin Signaling, and Nitric Oxide Synthase Activity in Endothelial Cells. Arterioscler Thromb Vasc Biol 2009; 29:1678-83. [DOI: 10.1161/atvbaha.109.189191] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Insulin resistance induces increased pulse pressure (PP), endothelial dysfunction (ED), and reduced bioavailability of endothelium-derived nitric oxide (NO). The genetic background of these 3 cardiovascular risk factors might be partly common. The
ENPP1
K121Q polymorphism is associated with insulin resistance and cardiovascular risk.
Methods and Results—
We investigated whether the K121Q polymorphism is associated with increased PP in white Caucasians and with ED in vitro. In 985 individuals, (390 unrelated and 595 from 248 families), the K121Q polymorphism was associated with PP (
P
=8.0×10
−4
). In the families, the Q121 variant accounted for 0.08 of PP heritability (
P
=9.4×10
−4
). This association was formally replicated in a second sample of 475 individuals (
P
=2.6×10
−2
) but not in 2 smaller samples of 289 and 236 individuals (
P
=0.49 and 0.21, respectively). In the individual patients’ data meta-analysis, comprising 1985 individuals, PP was associated with the Q121 variant (
P
=1.2×10
−3
). Human endothelial cells carrying the KQ genotype showed, as compared to KK cells, reduced insulin-mediated insulin receptor autophosphorylation (
P
=0.03), Ser
473
-Akt phosphorylation (
P
=0.03), and NO synthase activity (
P
=0.003).
Conclusions—
Our data suggest that the
ENPP1
Q121 variant is associated with increased PP in vivo and reduced insulin signaling and ED in vitro, thus indicating a possible pathogenic mechanism for the increased cardiovascular risk observed in
ENPP1
Q121 carriers.
Collapse
Affiliation(s)
- Simonetta Bacci
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Rosa Di Paola
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Claudia Menzaghi
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Patrizia Di Fulvio
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Sara Di Silvestre
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Fabio Pellegrini
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Roberto Baratta
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Antonella Marucci
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Sandra Mastroianno
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Grazia Fini
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Gloria Formoso
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Agostino Consoli
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Francesco Perticone
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Lucia Frittitta
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Assunta Pandolfi
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| | - Vincenzo Trischitta
- From the Endocrine Unit (S.B., S.M.), the Research Unit of Diabetes and Endocrine Diseases (R.D.P., C.M., A.M., G. Fini, V.T.), and the Unit of Biostatistics (F.P.), IRCCS “Casa Sollievo della Sofferenza,” San Giovanni Rotondo, Italy; the Departments of Medicine and Aging Sciences (P.D.F., G. Formoso, A.C.) and Biomedical Sciences (S.D.S., A.P.), University “G. d’Annunzio,” Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti-Pescara, Italy; the Department of Clinical
| |
Collapse
|
27
|
Prudente S, Morini E, Trischitta V. The emerging role of TRIB3 as a gene affecting human insulin resistance and related clinical outcomes. Acta Diabetol 2009; 46:79-84. [PMID: 19139803 DOI: 10.1007/s00592-008-0087-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 11/10/2008] [Indexed: 01/18/2023]
Abstract
Type 2 diabetes is becoming epidemic. The personal and social burden imposed by diabetes will increase in the close future as its prevalence is expected to double in the next 15-20 years. Type 2 diabetes is caused by the combination of resistance to insulin action and inadequate insulin secretion. Despite the role of profound changes in individual environmental exposure is incontrovertible, several findings clearly indicate that type 2 diabetes and insulin resistance are also heritable. Among the several inhibitors of insulin signalling, which have been recently proposed as determinants of insulin resistance, is TRIB3, a mammalian tribbles homolog which affects insulin signalling at the level of Akt-2, a key modulator of insulin action in target cells. We here report data on a prevalent Q84R TRIB3 missense single nucleotide polymorphism (rs2295490) we first described few years ago. Several lines of evidences indicate that this amino-acid change is, in fact, a gain of function mutation with the potential to affect insulin signalling and thus, to increase the risk of insulin resistance and related clinical outcomes.
Collapse
Affiliation(s)
- Sabrina Prudente
- CSS-Mendel Institute, Viale Regina Margherita 261, 00198, Rome, Italy.
| | | | | |
Collapse
|
28
|
Umar A, Kang H, Timmermans AM, Look MP, Meijer-van Gelder ME, den Bakker MA, Jaitly N, Martens JWM, Luider TM, Foekens JA, Pasa-Tolić L. Identification of a putative protein profile associated with tamoxifen therapy resistance in breast cancer. Mol Cell Proteomics 2009; 8:1278-94. [PMID: 19329653 PMCID: PMC2690491 DOI: 10.1074/mcp.m800493-mcp200] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Tamoxifen resistance is a major cause of death in patients with recurrent breast cancer. Current clinical factors can correctly predict therapy response in only half of the treated patients. Identification of proteins that are associated with tamoxifen resistance is a first step toward better response prediction and tailored treatment of patients. In the present study we intended to identify putative protein biomarkers indicative of tamoxifen therapy resistance in breast cancer using nano-LC coupled with FTICR MS. Comparative proteome analysis was performed on ∼5,500 pooled tumor cells (corresponding to ∼550 ng of protein lysate/analysis) obtained through laser capture microdissection (LCM) from two independently processed data sets (n = 24 and n = 27) containing both tamoxifen therapy-sensitive and therapy-resistant tumors. Peptides and proteins were identified by matching mass and elution time of newly acquired LC-MS features to information in previously generated accurate mass and time tag reference databases. A total of 17,263 unique peptides were identified that corresponded to 2,556 non-redundant proteins identified with ≥2 peptides. 1,713 overlapping proteins between the two data sets were used for further analysis. Comparative proteome analysis revealed 100 putatively differentially abundant proteins between tamoxifen-sensitive and tamoxifen-resistant tumors. The presence and relative abundance for 47 differentially abundant proteins were verified by targeted nano-LC-MS/MS in a selection of unpooled, non-microdissected discovery set tumor tissue extracts. ENPP1, EIF3E, and GNB4 were significantly associated with progression-free survival upon tamoxifen treatment for recurrent disease. Differential abundance of our top discriminating protein, extracellular matrix metalloproteinase inducer, was validated by tissue microarray in an independent patient cohort (n = 156). Extracellular matrix metalloproteinase inducer levels were higher in therapy-resistant tumors and significantly associated with an earlier tumor progression following first line tamoxifen treatment (hazard ratio, 1.87; 95% confidence interval, 1.25–2.80; p = 0.002). In summary, comparative proteomics performed on laser capture microdissection-derived breast tumor cells using nano-LC-FTICR MS technology revealed a set of putative biomarkers associated with tamoxifen therapy resistance in recurrent breast cancer.
Collapse
Affiliation(s)
- Arzu Umar
- Erasmus Medical Center Rotterdam, Josephine Nefkens Inst., Dept. of Medical Oncology, Laboratory of Genomics and Proteomics of Breast Cancer, Dr. Molewaterplein 50, Be 430c, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Prudente S, Scarpelli D, Chandalia M, Zhang YY, Morini E, Del Guerra S, Perticone F, Li R, Powers C, Andreozzi F, Marchetti P, Dallapiccola B, Abate N, Doria A, Sesti G, Trischitta V. The TRIB3 Q84R polymorphism and risk of early-onset type 2 diabetes. J Clin Endocrinol Metab 2009; 94:190-6. [PMID: 18984671 PMCID: PMC2630876 DOI: 10.1210/jc.2008-1365] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT The prevalence of type 2 diabetes (T2D), particularly among young adults, has been rising steadily during the past 2 decades. T2D, especially in its early-onset subtype, is under genetic control. TRIB3 inhibits insulin-stimulated Akt phosphorylation and subsequent insulin action. A TRIB3 gain-of-function polymorphism, Q84R (rs2295490), impairs insulin signaling. OBJECTIVE The objective of the study was to verify the association of TRIB3 Q84R with: 1) T2D, either subtyped or not according to age at diagnosis (early-onset, <45 yr, or >or= 45 yr); 2) insulin secretion and sensitivity in nondiabetic individuals; or 3) in vitro insulin secretion from isolated human islets. DESIGN Four different case-control samples comprising a total of 5,469 whites were examined. Insulinogenic and insulin sensitivity indexes and their interplay (disposition index) were assessed in 645 nondiabetic individuals at oral glucose tolerance test, glucose (16.7 mmol/liter)-induced in vitro insulin secretion was assessed in islets isolated from 54 nondiabetic donors. RESULTS In the whole sample, the R84 variant was nominally associated with T2D (odds ratio 1.17, 95% confidence interval 1.00-1.36, P = 0.04). When stratifying according to age of diabetes onset, R84 carriers had an increased risk of early-onset T2D (odds ratio 1.32, 95% confidence interval 1.10-1.58, P = 0.002). Among 645 nondiabetic subjects, R84 carriers had higher glucose levels (P = 0.005) and lower insulinogenic (P = 0.03) and disposition index (P = 0.02) during the oral glucose tolerance test. R84 islets were more likely to display relatively low glucose-stimulated insulin release (P = 0.04). CONCLUSIONS The TRIB3 R84 variant is associated with early-onset T2D in whites. Alteration in the insulin secretion/insulin sensitivity interplay appears to underlie this association.
Collapse
Affiliation(s)
- Sabrina Prudente
- CSS-Mendel Institute, Viale Regina Margherita 261, 00198 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Baratta R, Rossetti P, Prudente S, Barbetti F, Sudano D, Nigro A, Farina MG, Pellegrini F, Trischitta V, Frittitta L. Role of the ENPP1 K121Q polymorphism in glucose homeostasis. Diabetes 2008; 57:3360-4. [PMID: 18776139 PMCID: PMC2584144 DOI: 10.2337/db07-1830] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Accepted: 08/26/2008] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To study the role of the ENPP1 Q121 variant on glucose homeostasis in whites from Italy. RESEARCH DESIGN AND METHODS We conducted case-control studies in 764 adults (from two independent samples of 289 nonobese and 485 obese individuals) and 240 overweight/obese children undergoing oral glucose tolerance testing (OGTT). Early-phase insulin secretion and insulin sensitivity (the insulinogenic index and the insulin sensitivity index) and their interplay (the disposition index) were calculated. RESULTS In adult subjects, glucose profiles during OGTT were significantly (P = 2 x 10(-2)) different across K121Q genotype groups and higher in QQ than KK individuals (P = 5 x 10(-2)). The insulinogenic index was significantly reduced in QQ (18.5 +/- 3.4) compared with both KK (31.6 +/- 1.0; P = 2.2 x 10(-7)) and KQ (30.5 +/- 1.5; P = 3.2 x 10(-6)) individuals. KQ individuals also showed a reduced insulin sensitivity index compared with KK subjects (P = 3.6 x 10(-2)). The disposition index was lower in QQ carriers than in KQ and KK individuals (P = 8 x 10(-3) and 4 x 10(-4), respectively) and lower in KQ than in KK individuals (P = 3 x 10(-2)). Data obtained in overweight/obese children were very similar to those observed in adults, with QQ individuals showing (compared with KQ and KK subjects) a reduced insulinogenic index (P = 7 x 10(-3) and 2 x 10(-2), respectively) and disposition index (P = 2 x 10(-2) and 7 x 10(-3), respectively). CONCLUSIONS Homozygous carriers of the ENPP1 Q121 variant are characterized by an altered glucose homeostasis. Reduced early-phase insulin secretion and inefficient interplay between insulin secretion and sensitivity, which occur at early ages, are major determinants of this defect.
Collapse
Affiliation(s)
- Roberto Baratta
- Unit of Endocrinology, Department of Internal and Specialistic Medicine, University of Catania Medical School, Garibaldi Hospital, Catania, Italy
| | - Paola Rossetti
- Unit of Endocrinology, Department of Internal and Specialistic Medicine, University of Catania Medical School, Garibaldi Hospital, Catania, Italy
| | - Sabrina Prudente
- Research Unit of Diabetes and Endocrine Diseases, Casa Sollievo della Sofferenza (CSS) Scientific Institute, San Giovanni Rotondo, Italy
- CSS-Mendel Institute, Rome, Italy
| | - Fabrizio Barbetti
- Bambino Gesù Pediatric Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Dora Sudano
- Unit of Endocrinology, Department of Internal and Specialistic Medicine, University of Catania Medical School, Garibaldi Hospital, Catania, Italy
| | - Angela Nigro
- Unit of Endocrinology, Department of Internal and Specialistic Medicine, University of Catania Medical School, Garibaldi Hospital, Catania, Italy
| | - Maria Grazia Farina
- Unit of Endocrinology, Department of Internal and Specialistic Medicine, University of Catania Medical School, Garibaldi Hospital, Catania, Italy
| | - Fabio Pellegrini
- Research Unit of Diabetes and Endocrine Diseases, Casa Sollievo della Sofferenza (CSS) Scientific Institute, San Giovanni Rotondo, Italy
- Unit of Biostatistics, Department of Clinical Pharmacology and Epidemiology, Consorzio Mario Negri Sud, S. Maria Imbaro, Chieti, Italy
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine Diseases, Casa Sollievo della Sofferenza (CSS) Scientific Institute, San Giovanni Rotondo, Italy
- CSS-Mendel Institute, Rome, Italy
- Unit of Endocrinology, CSS Scientific Institute, San Giovanni Rotondo, Italy
- Department of Clinical Sciences, Sapienza University, Rome, Italy
| | - Lucia Frittitta
- Unit of Endocrinology, Department of Internal and Specialistic Medicine, University of Catania Medical School, Garibaldi Hospital, Catania, Italy
| |
Collapse
|
31
|
Stolerman ES, Manning AK, McAteer JB, Dupuis J, Fox CS, Cupples LA, Meigs JB, Florez JC. Haplotype structure of the ENPP1 Gene and Nominal Association of the K121Q missense single nucleotide polymorphism with glycemic traits in the Framingham Heart Study. Diabetes 2008; 57:1971-7. [PMID: 18426862 PMCID: PMC2453609 DOI: 10.2337/db08-0266] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Accepted: 04/16/2008] [Indexed: 12/03/2022]
Abstract
OBJECTIVE A recent meta-analysis demonstrated a nominal association of the ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1) K-->Q missense single nucleotide polymorphism (SNP) at position 121 with type 2 diabetes. We set out to confirm the association of ENPP1 K121Q with hyperglycemia, expand this association to insulin resistance traits, and determine whether the association stems from K121Q or another variant in linkage disequilibrium with it. RESEARCH DESIGN AND METHODS We characterized the haplotype structure of ENPP1 and selected 39 tag SNPs that captured 96% of common variation in the region (minor allele frequency > or =5%) with an r(2) value > or =0.80. We genotyped the SNPs in 2,511 Framingham Heart Study participants and used age- and sex-adjusted linear mixed effects (LME) models to test for association with quantitative metabolic traits. We also examined whether interaction between K121Q and BMI affected glycemic trait levels. RESULTS The Q allele of K121Q (rs1044498) was associated with increased fasting plasma glucose (FPG), A1C, fasting insulin, and insulin resistance by homeostasis model assessment (HOMA-IR; all P = 0.01-0.006). Two noncoding SNPs (rs7775386 and rs7773477) demonstrated similar associations, but LME models indicated that their effects were not independent from K121Q. We found no association of K121Q with obesity, but interaction models suggested that the effect of the Q allele on FPG and HOMA-IR was stronger in those with a higher BMI (P = 0.008 and 0.01 for interaction, respectively). CONCLUSIONS The Q allele of ENPP1 K121Q is associated with hyperglycemia and insulin resistance in whites. We found an adiposity-SNP interaction, with a stronger association of K121Q with diabetes-related quantitative traits in people with a higher BMI.
Collapse
Affiliation(s)
- Elliot S. Stolerman
- Center for Human Genetic Research and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alisa K. Manning
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Jarred B. McAteer
- Center for Human Genetic Research and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Caroline S. Fox
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - James B. Meigs
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- General Medicine Division, Massachusetts General Hospital, Boston, Massachusetts
| | - Jose C. Florez
- Center for Human Genetic Research and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Florez JC. Newly identified loci highlight beta cell dysfunction as a key cause of type 2 diabetes: where are the insulin resistance genes? Diabetologia 2008; 51:1100-10. [PMID: 18504548 DOI: 10.1007/s00125-008-1025-9] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Accepted: 04/04/2008] [Indexed: 12/13/2022]
Abstract
Although type 2 diabetes has been traditionally understood as a metabolic disorder initiated by insulin resistance, it has recently become apparent that an impairment in insulin secretion contributes to its manifestation and may play a prominent role in its early pathophysiology. The genetic dissection of Mendelian and, more recently, polygenic types of diabetes confirms the notion that primary defects in insulin synthesis, processing and/or secretion often give rise to the common form of this disorder. This concept, first advanced with the discovery and physiological characterisation of various genetic subtypes of MODY, has been extended to other forms of monogenic diabetes (e.g. neonatal diabetes). It has also led to the identification of common risk variants via candidate gene approaches (e.g. the E23K polymorphism in KCNJ11 or common variants in the MODY genes), and it has been validated by the description of the robust physiological effects conferred by polymorphisms in the TCF7L2 gene. More recently, the completion and integration of genome-wide association scans for this disease has uncovered a number of heretofore unsuspected variants, several of which also affect insulin secretion. This review provides an up-to-date account of genetic loci that influence risk of common type 2 diabetes via impairment of beta cell function, outlines their presumed mechanisms of action, and places them in the context of gene-gene and/or gene-environment interactions. Finally, a strategy for the analogous discovery of insulin resistance genes is proposed.
Collapse
Affiliation(s)
- J C Florez
- Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
33
|
McAteer JB, Prudente S, Bacci S, Lyon HN, Hirschhorn JN, Trischitta V, Florez JC. The ENPP1 K121Q polymorphism is associated with type 2 diabetes in European populations: evidence from an updated meta-analysis in 42,042 subjects. Diabetes 2008; 57:1125-30. [PMID: 18071025 DOI: 10.2337/db07-1336] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Functional studies suggest that the nonsynonymous K121Q polymorphism in the ectoenzyme nucleotide pyrophosphate phosphodiesterase 1 (ENPP1) may confer susceptibility to insulin resistance; genetic evidence on its effect on type 2 diabetes, however, has been conflicting. We therefore conducted a new meta-analysis that includes novel unpublished data from the ENPP1 Consortium and recent negative findings from large association studies to address the contribution of K121Q to type 2 diabetes. RESEARCH DESIGN AND METHODS After a systematic review of the literature, we evaluated the effect of ENPP1 K121Q on diabetes risk under three genetic models using a random-effects approach. Our primary analysis consisted of 30 studies comprising 15,801 case and 26,241 control subjects. Due to considerable heterogeneity and large differences in allele frequencies across populations, we limited our meta-analysis to those of self-reported European descent and, when available, included BMI as a covariate. RESULTS We found a modest increase in risk of type 2 diabetes for QQ homozygotes in white populations (combined odds ratio [OR] 1.38 [95% CI 1.10-1.74], P = 0.005). There was no evidence of publication bias, but we noted significant residual heterogeneity among studies (P = 0.02). On meta-regression, 16% of the effect was accounted for by the mean BMI of control subjects. This association was stronger in studies in which control subjects were leaner but disappeared after adjustment for mean control BMI (combined OR 0.93 [95% CI 0.75-1.15], P = 0.50). CONCLUSIONS The ENPP1 Q121 variant increases risk of type 2 diabetes under a recessive model of inheritance in whites, an effect that appears to be modulated by BMI.
Collapse
Affiliation(s)
- Jarred B McAteer
- Diabetes Unit/Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|