1
|
Bhat JA, Bhat MH, Masoodi SR, Ahmad H, Ahmad PO, Wangnoo BR, Bashir MI, Wani AI, Misgar RA, Rashid I, Ganai BA, Nissar K, Shah ZA. Prevalence and Clinical Profile of Maturity Onset Diabetes of the Young among People with Diabetes Attending a Tertiary Care Centre. Indian J Endocrinol Metab 2022; 26:543-550. [PMID: 39005520 PMCID: PMC11245292 DOI: 10.4103/ijem.ijem_334_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/22/2022] [Accepted: 10/29/2022] [Indexed: 02/05/2023] Open
Abstract
Background Maturity onset diabetes of young (MODY) is considered to be the most underdiagnosed condition. The correct diagnosis of MODY has a definite bearing on the outcome and clinical course of the disease. We aim to determine the prevalence and clinical profile of MODY among young diabetic patients attending at Department of Endocrinology, a tertiary care institute in North India. Methods It was a cross-sectional study involving all consecutive consenting patients with diabetes and age of onset ≤35 years. A total of 1,094 patients were included in this study, of whom 858 were having age of onset of diabetes <25 years. All patients were screened for MODY using clinical criteria and MODY Probability calculator (available on diabetesgenes.org). Patients with high clinical probability of MODY having negative anti-GAD65 antibody and fasting serum C-peptide levels >0.6 ng/mL were subjected to the Ala98 Val polymorphism (SNP) in hepatocyte nuclear factor (HNF) 1a gene. Results The prevalence of MODY among the study cohort as per clinical criteria was found to be 7.7%. Males constituted the majority of patients (male vs female, 56% vs. 44%; P < 0.001). The patients with MODY were younger (p < 0.001), leaner (p < 0.001), had younger age at onset of diabetes mellitus (p < 0.001), and lower frequency of features of insulin resistance in the form of skin tags and acanthosis nigricans. Among the 40 patients who were subjected to Ala98Val polymorphism of HNF1α gene (MODY 3), the mutant genotype was seen in 20 (50%) patients. Conclusion We report a higher prevalence of MODY in our young diabetic patients. A high index of suspicion is required to diagnose MODY as misdiagnosis and inappropriate treatment may have a significant impact on quality-of-life (QOL) with increased cost and unnecessary treatment with insulin.
Collapse
Affiliation(s)
- Javaid Ahmad Bhat
- Department of Endocrinology, Superspeciality Hospital, GMC, Srinagar, Jammu and Kashmir, India
| | - Moomin Hussain Bhat
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Shariq Rashid Masoodi
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Hilal Ahmad
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Peerzada Ovais Ahmad
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Bisma Rashid Wangnoo
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Mir Iftikhar Bashir
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Arshad Iqbal Wani
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Raiz Ahmad Misgar
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Imtiyaz Rashid
- Department of Immunology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Bashir Ahmad Ganai
- Centre of Research for Development, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Kamran Nissar
- Centre of Research for Development, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Zafar Amin Shah
- Department of Immunology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
2
|
da Silva Santos T, Fonseca L, Santos Monteiro S, Borges Duarte D, Martins Lopes A, Couto de Carvalho A, Oliveira MJ, Borges T, Laranjeira F, Couce ML, Cardoso MH. MODY probability calculator utility in individuals' selection for genetic testing: Its accuracy and performance. Endocrinol Diabetes Metab 2022; 5:e00332. [PMID: 35822264 PMCID: PMC9471596 DOI: 10.1002/edm2.332] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction MODY probability calculator (MPC) represents an easy‐to‐use tool developed by Exeter University to help clinicians prioritize which individuals should be oriented to genetic testing. We aimed to assess the utility of MPC in a Portuguese cohort with early‐onset monogenic diabetes. Methods This single‐centre retrospective study enrolled 132 participants submitted to genetic testing between 2015 and 2020. Automatic sequencing and, in case of initial negative results, generation sequencing were performed. MODY probability was calculated using the probability calculator available online. Positive and negative predictive values (PPV and NPV, respectively), accuracy, sensitivity and specificity of the calculator were determined for this cohort. Results Seventy‐three individuals were included according to inclusion criteria: 20 glucokinase (GCK‐MODY); 16 hepatocyte nuclear factor 1A (HNF1A‐MODY); 2 hepatocyte nuclear factor 4A (HNF4A‐MODY) and 35 DM individuals with no monogenic mutations found. The median probability score of MODY was significantly higher in monogenic diabetes‐positive subgroup (75.5% vs. 24.2%, p < .001). The discriminative accuracy of the calculator, as expressed by area under the curve, was 75% (95% CI: 64%–85%). In our cohort, the best cut‐off value for the MODY calculator was found to be 36%, with a PPV of 74.4%, NPV of 73.5% and corresponding sensitivity and specificity of 76.2% and 71.4%, respectively. Conclusions In a highly pre‐selected group of probands qualified for genetic testing, the Exeter MODY probability calculator provided a useful tool in individuals' selection for genetic testing, with good discrimination ability under an optimal probability cut‐off of 36%. Further geographical and population adjustments are warranted for general use.
Collapse
Affiliation(s)
- Tiago da Silva Santos
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - Liliana Fonseca
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - Sílvia Santos Monteiro
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - Diana Borges Duarte
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - Ana Martins Lopes
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - André Couto de Carvalho
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - Maria João Oliveira
- Division of Pediatric Endocrinology Department of Pediatrics Centro Materno‐Infantil do Norte – Centro Hospitalar e Universitário do Porto Porto Portugal
| | - Teresa Borges
- Division of Pediatric Endocrinology Department of Pediatrics Centro Materno‐Infantil do Norte – Centro Hospitalar e Universitário do Porto Porto Portugal
| | | | - María Luz Couce
- University Clinical Hospital of Santiago de Compostela, IDIS CIBERER MetabERN Santiago de Compostela Spain
| | - Maria Helena Cardoso
- Division of Endocrinology, Diabetes and Metabolism Hospital de Santo António – Centro Hospitalar e Universitário do Porto Porto Portugal
| |
Collapse
|
3
|
César Ernesto LC, Álvaro EO, Yayoi SK, Juanita SS, María Teresa TL, Almeda-Valdes P. Differentiating Among Type 1, Type 2 Diabetes, and MODY: Raising Awareness About the Clinical Implementation of Genetic Testing in Latin America. AACE Clin Case Rep 2021; 7:138-140. [PMID: 34095472 PMCID: PMC8053617 DOI: 10.1016/j.aace.2020.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective To describe a case of maturity-onset diabetes of the young (MODY) to highlight the importance of a correct diabetes diagnosis. Methods We describe a Mexican family misdiagnosed with T1D and T2D. Results A 36-year-old woman with diabetes and adverse outcomes during 2 pregnancies had been diagnosed with T2D 10 years ago. Genetic testing was performed due to clinical and family history, which showed a pathogenic heterozygous variant c.544G>T (p.Val182Leu) in the GCK gene. This mutation was also confirmed in most of the family members who had been diagnosed with diabetes. Conclusion This case highlights the need for a correct diabetes classification. Reassessment of diabetes etiology is justified, especially in individuals with unclear clinical presentation or when family history is suggestive.
Collapse
Affiliation(s)
- Lam-Chung César Ernesto
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Elizondo Ochoa Álvaro
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Segura Kato Yayoi
- Molecular Biology and Genomic Medicine Unit; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Silva-Serrano Juanita
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Tusié Luna María Teresa
- Molecular Biology and Genomic Medicine Unit; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Paloma Almeda-Valdes
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
- Address correspondence and reprint requests to Dr. Paloma Almeda-Valdes, Department of Endocrinology and Metabolism,Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan 14080, México City, México.
| |
Collapse
|
4
|
Carreira NR, Gonçalves C, Wahnon A, Dâmaso S, Martins J. Late Diagnosis of Maturity-Onset Diabetes of the Young (MODY) 12 With Catastrophic Consequences. Cureus 2021; 13:e13145. [PMID: 33728157 PMCID: PMC7935225 DOI: 10.7759/cureus.13145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY) is a genetically and clinically heterogeneous group of diseases characterized by autosomal dominant monogenic non-ketogenic diabetes mellitus, usually with early-onset, with a prevalence of 1-5% of all diabetes cases. A 72-year-old female was admitted with intestinal occlusion, anorexia, vomiting, and weight loss for four months. Medical history of type 2 diabetes mellitus, chronic pancreatitis with abnormal pancreatic development, and acute obstructive jaundice due to a mass in the head of the pancreas with duodenum extension four months before. Assuming surgically unresectable pancreatic neoplasm, digestive bypass surgery was performed. The pathologic examination of surgical specimens was negative for neoplasm. Abdominal imaging showed the pancreatic mass, proximal bowel distension and ascites, which was negative for neoplastic cells. A percutaneous biopsy of the mass revealed adenocarcinoma. Palliative chemotherapy was started. Next-generation sequencing revealed the variant c.-8G>T in the 5’ untranslated region (UTR) region of the adenosine triphosphate (ATP) binding cassette subfamily C member 8 (ABCC8) gene in heterozygosity, associated with the MODY 12 subtype. We report a possible case of MODY 12 diabetes with a phenotype not previously described: a non-neoplastic pancreatic mass that appears in a previously abnormally developed pancreas, with evolution to neoplasm along with the late development of diabetes mellitus. Although this ABCC8 gene mutation could be incidental, there could be a relationship between this mutation, pancreatic malformation, chronic pancreatitis and pancreatic neoplasm. Investigation of new phenotypes is critical, including the potential role of the ABCC8 gene in oncogenesis.
Collapse
Affiliation(s)
- Nuno R Carreira
- Internal Medicine, Serviço de Medicina 2, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, PRT
| | - Catarina Gonçalves
- Internal Medicine, Serviço de Medicina 2, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, PRT
| | - Alexandra Wahnon
- Internal Medicine, Serviço de Medicina 2, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, PRT
| | - Sara Dâmaso
- Oncology, Serviço de Medicina 2, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, PRT
| | - Joao Martins
- Internal Medicine: Diabetes and Endocrinology, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, PRT
| |
Collapse
|
5
|
Ocular surface assessment in maturity-onset diabetes of the young. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-020-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
6
|
Baldacchino I, Pace NP, Vassallo J. Screening for monogenic diabetes in primary care. Prim Care Diabetes 2020; 14:1-11. [PMID: 31253563 DOI: 10.1016/j.pcd.2019.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 10/26/2022]
Abstract
AIMS Updates on the latest diagnostic methods and features of MODY (Maturity Onset Diabetes of the Young) and promotion of education and awareness on the subject are discussed. METHOD Previous recommendations were identified using PubMed and using combinations of terms including "MODY" "monogenic diabetes" "mature onset diabetes" "MODY case review". The diabetesgenes.org website and the US Monogenic Diabetes Registry (University of Colorado) were directly referenced. The remaining referenced papers were taken from peer-reviewed journals. The initial literature search occurred in January 2017 and the final search occurred in September 2018. RESULTS A diagnosis of MODY has implications for treatment, quality of life, management in pregnancy and research. The threshold for referral and testing varies among different ethnic groups, and depends on body mass index, family history of diabetes and associated syndromes. Novel causative genetic variations are still being discovered however testing is currently limited by low referral rates. Educational material is currently being promoted in the UK in an effort to raise awareness. CONCLUSIONS The benefits and implications of life altering treatment such as termination of insulin administration are significant but little can be done without appropriate identification and referral.
Collapse
Affiliation(s)
- Ian Baldacchino
- Specialist Training Programme in Family Medicine, Birkirkara Health Centre, Birkirkara, Malta.
| | - Nikolai Paul Pace
- Faculty of Medicine & Surgery, Biomedical Sciences Building, University of Malta, Msida, Malta.
| | - Josanne Vassallo
- Division of Diabetes and Endocrinology, University of Malta Medical School, Mater Dei Hospital, Msida, Malta.
| |
Collapse
|
7
|
Maturity-Onset Diabetes of the Young (MODY) in Portugal: Novel GCK, HNFA1 and HNFA4 Mutations. J Clin Med 2020; 9:jcm9010288. [PMID: 31968686 PMCID: PMC7019520 DOI: 10.3390/jcm9010288] [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: 12/14/2019] [Revised: 01/02/2020] [Accepted: 01/15/2020] [Indexed: 12/17/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY) is a frequently misdiagnosed type of diabetes, which is characterized by early onset, autosomal dominant inheritance, and absence of insulin dependence. The most frequent subtypes are due to mutations of the GCK (MODY 2), HNF1A (MODY 3), and HNF4A (MODY 1) genes. We undertook the first multicenter genetic study of MODY in the Portuguese population. The GCK, HNF1A, and HNF4A genes were sequenced in 46 unrelated patients that had at least two of the three classical clinical criteria for MODY (age at diagnosis, family history, and clinical presentation). The functional consequences of the mutations were predicted by bioinformatics analysis. Mutations were identified in 23 (50%) families. Twelve families had mutations in the GCK gene, eight in the HNF1A gene, and three in the HNF4A gene. These included seven novel mutations (GCK c.494T>C, GCK c.563C>G, HNF1A c.1623G>A, HNF1A c.1729C>G, HNF4A c.68delG, HNF4A c.422G>C, HNF4A c.602A>C). Mutation-positive patients were younger at the time of diagnosis when compared to mutation-negative patients (14.3 vs. 23.0 years, p = 0.011). This study further expands the spectrum of known mutations associated with MODY, and may contribute to a better understanding of this type of diabetes and a more personalized clinical management of affected individuals.
Collapse
|
8
|
Ming-Qiang Z, Yang-Li D, Ke H, Wei W, Jun-Fen F, Chao-Chun Z, Guan-Ping D. Maturity onset diabetes of the young (MODY) in Chinese children: genes and clinical phenotypes. J Pediatr Endocrinol Metab 2019; 32:759-765. [PMID: 31216263 DOI: 10.1515/jpem-2018-0446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/04/2019] [Indexed: 12/17/2022]
Abstract
Background To investigate the clinical and molecular characteristics of Chinese children with maturity onset diabetes of the young (MODY). Methods A total of 42 Chinese patients suspected MODY referred to our unit from 2014 to 2018 were enrolled. Mutational analysis of monogenic diabetes mellitus genes was performed by next-generation sequencing and confirmed by Sanger sequencing. Results There were 28 males (66.7%) and 14 females (33.3%) with a mean age of 9.49 ± 3.46 years (range, 1.4-15.3 years) and a mean birth weight of 3.38 ± 0.49 kg (range, 2.55-4.90 kg). Among these patients, 15 patients had polyuria, polydipsia or weight loss. Two patients (4.8%) were obese and six (14.3%) were overweight. Moreover, 13 patients (30.9%) had a family history of diabetes. Thirty variants were identified in 28 patients. Twenty-six variants in 25 patients were pathogenic or likely pathogenic genes (59.5%, 25/42), including 15 patients (60.0%, 15/25) with GCK mutation, four (16.0%, 4/25) with PAX4 mutation, three (12.0%, 3/25) with HNF4A mutation, one (4.0%, 1/25) with INS mutation, one (4.0%, 1/25) with NEUROD1 mutation and one (4.0%, 1/25) with HNF1A mutation. Nine mutations (36.0%, 9/25) were novel. There was no difference between mutation-suspected patients and MODY-confirmed patients except for a 2-h glucose increment in an oral glucose tolerance test (OGTT), while the GCK-MODY had lower glycated hemoglobin (HbA1c) and a significantly smaller 2-h glucose increment in an OGTT compared with transcription factor MODYs. The GCK-MODY was identified by incidental hyperglycemia without glycosuria. GCK-MODY without drug management and hepatocyte nuclear factor-1 alpha (HNF4A) or HNF1A-MODY with sulfonylurea therapy obtained good glucose controlling. Conclusions Mutation of the GCK gene is the most common in MODY patients in China followed by PAX4. The screening criteria can improve the cost-effectiveness of disease diagnosis and treatment. A precise molecular diagnosis would lead to optimal treatment of the patients.
Collapse
Affiliation(s)
- Zhu Ming-Qiang
- Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dai Yang-Li
- Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huang Ke
- Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wu Wei
- Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Fu Jun-Fen
- Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zou Chao-Chun
- Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dong Guan-Ping
- Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou 310051, China, Phone: +86-13757119832, Fax: +86-571-87033296
| |
Collapse
|
9
|
Sajadimajd S, Bahrami G, Daglia M, Nabavi SM, Naseri R, Farzaei MH. Plant-Derived Supplementary Carbohydrates, Polysaccharides and Oligosaccharides in Management of Diabetes Mellitus: A Comprehensive Review. FOOD REVIEWS INTERNATIONAL 2019. [DOI: 10.1080/87559129.2019.1584818] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Soraya Sajadimajd
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamreza Bahrami
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maria Daglia
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Pavia, Italy
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Rozita Naseri
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
10
|
Johnson SR, Ellis JJ, Leo PJ, Anderson LK, Ganti U, Harris JE, Curran JA, McInerney-Leo AM, Paramalingam N, Song X, Conwell LS, Harris M, Jones TW, Brown MA, Davis EA, Duncan EL. Comprehensive genetic screening: The prevalence of maturity-onset diabetes of the young gene variants in a population-based childhood diabetes cohort. Pediatr Diabetes 2019; 20:57-64. [PMID: 30191644 DOI: 10.1111/pedi.12766] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/05/2018] [Accepted: 08/12/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Maturity-onset diabetes of the young (MODY) is caused by autosomal dominant mutations in one of 13 confirmed genes. Estimates of MODY prevalence vary widely, as genetic screening is usually restricted based on clinical features, even in population studies. We aimed to determine prevalence of MODY variants in a large and unselected pediatric diabetes cohort. METHODS MODY variants were assessed using massively parallel sequencing in the population-based diabetes cohort (n = 1363) of the sole tertiary pediatric diabetes service for Western Australia (population 2.6 million). All individuals were screened, irrespective of clinical features. MODY variants were also assessed in a control cohort (n = 993). RESULTS DNA and signed consent were available for 821 children. Seventeen children had pathogenic/likely pathogenic variants in MODY genes, two diagnosed with type 2 diabetes, four diagnosed with antibody-negative type 1 diabetes (T1DM), three diagnosed with antibody-positive T1DM, and eight previously diagnosed with MODY. Prevalence of MODY variants in the sequenced cohort was 2.1%, compared to 0.3% of controls. CONCLUSIONS This is the first comprehensive study of MODY variants in an unselected population-based pediatric diabetes cohort. The observed prevalence, increasing access to rapid and affordable genetic screening, and significant clinical implications suggest that genetic screening for MODY could be considered for all children with diabetes, irrespective of other clinical features.
Collapse
Affiliation(s)
- Stephanie R Johnson
- Department of Endocrinology and Diabetes, Lady Cilento Children's Hospital, South Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Jonathan J Ellis
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Paul J Leo
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Lisa K Anderson
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Uma Ganti
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Perth, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Jessica E Harris
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Jacqueline A Curran
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Perth, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Aideen M McInerney-Leo
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Nirubasini Paramalingam
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Perth, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Xiaoxia Song
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Louise S Conwell
- Department of Endocrinology and Diabetes, Lady Cilento Children's Hospital, South Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark Harris
- Department of Endocrinology and Diabetes, Lady Cilento Children's Hospital, South Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Timothy W Jones
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Perth, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia.,School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Matthew A Brown
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Elizabeth A Davis
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Perth, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia.,School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Emma L Duncan
- Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology (QUT), Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Endocrinology and Diabetes, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Xie F, Chan JCN, Ma RCW. Precision medicine in diabetes prevention, classification and management. J Diabetes Investig 2018; 9:998-1015. [PMID: 29499103 PMCID: PMC6123056 DOI: 10.1111/jdi.12830] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/12/2018] [Indexed: 12/18/2022] Open
Abstract
Diabetes has become a major burden of healthcare expenditure. Diabetes management following a uniform treatment algorithm is often associated with progressive treatment failure and development of diabetic complications. Recent advances in our understanding of the genomic architecture of diabetes and its complications have provided the framework for development of precision medicine to personalize diabetes prevention and management. In the present review, we summarized recent advances in the understanding of the genetic basis of diabetes and its complications. From a clinician's perspective, we attempted to provide a balanced perspective on the utility of genomic medicine in the field of diabetes. Using genetic information to guide management of monogenic forms of diabetes represents the best-known examples of genomic medicine for diabetes. Although major strides have been made in genetic research for diabetes, its complications and pharmacogenetics, ongoing efforts are required to translate these findings into practice by incorporating genetic information into a risk prediction model for prioritization of treatment strategies, as well as using multi-omic analyses to discover novel drug targets with companion diagnostics. Further research is also required to ensure the appropriate use of this information to empower individuals and healthcare professionals to make personalized decisions for achieving the optimal outcome.
Collapse
Affiliation(s)
- Fangying Xie
- Department of Medicine and TherapeuticsPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
| | - Juliana CN Chan
- Department of Medicine and TherapeuticsPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
- Hong Kong Institute of Diabetes and ObesityPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
- Li Ka Shing Institute of Health SciencesPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
- CUHK‐SJTU Joint Research Centre in Diabetes Genomics and Precision MedicinePrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
| | - Ronald CW Ma
- Department of Medicine and TherapeuticsPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
- Hong Kong Institute of Diabetes and ObesityPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
- Li Ka Shing Institute of Health SciencesPrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
- CUHK‐SJTU Joint Research Centre in Diabetes Genomics and Precision MedicinePrince of Wales HospitalThe Chinese University of Hong KongShatinHong Kong
| |
Collapse
|
12
|
Analysis of the GCK gene in 79 MODY type 2 patients: A multicenter Turkish study, mutation profile and description of twenty novel mutations. Gene 2018; 641:186-189. [DOI: 10.1016/j.gene.2017.10.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 08/01/2017] [Accepted: 10/18/2017] [Indexed: 11/18/2022]
|
13
|
Davis TM, Makepeace AE, Ellard S, Colclough K, Peters K, Hattersley A, Davis WA. The prevalence of monogenic diabetes in Australia: the Fremantle Diabetes Study Phase II. Med J Aust 2017; 207:344-347. [PMID: 29020906 DOI: 10.5694/mja16.01201] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 01/06/2017] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To determine the prevalence of monogenic diabetes in an Australian community. DESIGN Longitudinal observational study of a cohort recruited between 2008 and 2011. SETTING Urban population of 157 000 people (Fremantle, Western Australia). PARTICIPANTS 1668 (of 4639 people with diabetes) who consented to participation (36.0% participation). MAIN OUTCOME MEASURES Prevalence of maturity-onset diabetes of the young (MODY) and permanent neonatal diabetes in patients under 35 years of age, from European and non-European ethnic backgrounds, who were at risk of MODY according to United Kingdom risk prediction models, and who were then genotyped for relevant mutations. RESULTS Twelve of 148 young participants with European ethnic backgrounds (8%) were identified by the risk prediction model as likely to have MODY; four had a glucokinase gene mutation. Thirteen of 45 with non-European ethnic backgrounds (28%) were identified as likely to have MODY, but none had a relevant mutation (DNA unavailable for one patient). Two patients with European ethnic backgrounds (one likely to have MODY) had neonatal diabetes. The estimated MODY prevalence among participants with diagnosed diabetes was 0.24% (95% confidence interval [CI], 0.08-0.66%), an overall population prevalence of 89 cases per million; the prevalence of permanent neonatal diabetes was 0.12% (95% CI, 0.02-0.48%) and the population prevalence 45 cases per million. CONCLUSIONS One in 280 Australians diagnosed with diabetes have a monogenic form; most are of European ethnicity. Diagnosing MODY and neonatal diabetes is important because their management (including family screening) and prognosis can differ significantly from those for types 1 and 2 diabetes.
Collapse
Affiliation(s)
| | | | - Sian Ellard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, United Kingdom
| | - Kevin Colclough
- Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | | | - Andrew Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, United Kingdom
| | | |
Collapse
|
14
|
Kim SH. Maturity-Onset Diabetes of the Young: What Do Clinicians Need to Know? Diabetes Metab J 2015; 39:468-77. [PMID: 26706916 PMCID: PMC4696982 DOI: 10.4093/dmj.2015.39.6.468] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/25/2015] [Indexed: 11/15/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY) is a monogenic form of diabetes that is characterized by an early onset, autosomal dominant mode of inheritance and a primary defect in pancreatic β-cell function. MODY represents less than 2% of all diabetes cases and is commonly misdiagnosed as type 1 or type 2 diabetes mellitus. At least 13 MODY subtypes with distinct genetic etiologies have been identified to date. A correct genetic diagnosis is important as it often leads to personalized treatment for those with diabetes and enables predictive genetic testing for their asymptomatic relatives. Next-generation sequencing may provide an efficient method for screening mutations in this form of diabetes as well as identifying new MODY genes. In this review, I discuss a current update on MODY in the literatures and cover the studies that have been performed in Korea.
Collapse
Affiliation(s)
- Sung Hoon Kim
- Division of Endocrinology & Metabolism, Department of Medicine, Cheil General Hospital & Women's Healthcare Center, Dankook University College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Chen YT, Lin WD, Liao WL, Lin YJ, Chang JG, Tsai FJ. PTPRD silencing by DNA hypermethylation decreases insulin receptor signaling and leads to type 2 diabetes. Oncotarget 2015; 6:12997-3005. [PMID: 26079428 PMCID: PMC4536994 DOI: 10.18632/oncotarget.4092] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/11/2015] [Indexed: 11/28/2022] Open
Abstract
Genome-wide association study (GWAS) data showed that the protein tyrosine phosphatase receptor type delta (PTPRD) is associated with increased susceptibility to type 2 diabetes (T2D) in Han Chinese. A replication study indicated that PTPRD is involved in the insulin signaling pathway; however, the underlying mechanism remains unclear. We evaluated PTPRD expression in patients with T2D and controls. PTPRD expression levels were lower in patients and were correlated with the duration of the disease. Overexpression of the human insulin receptor PPARγ2 in HepG2 cells induced overexpression of PTPRD and the insulin receptor. PTPRD knockdown, using a shRNA, resulted in down-regulation of the insulin receptor. These results indicate that PTPRD activates PPARγ2 in the insulin signaling pathway. Similar results for PTPRD expression were found using a T2D mouse model. Silencing of PTPRD was caused by DNA methylation in T2D mice and patients, and correlated with DNMT1 expression. Furthermore, we showed that a DNMT1 SNP (rs78789647) was correlated with susceptibility to T2D. This study shows for the first time that DNMT1 caused PTPRD DNA hypermethylation and induced insulin signaling silencing in T2D patients. Our findings contribute to a better understanding of the crucial roles of these regulatory elements in human T2D.
Collapse
Affiliation(s)
- Yng-Tay Chen
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-De Lin
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Lin Liao
- Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Ying-Ju Lin
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of China Medical Science, China Medical University, Taichung, Taiwan
| | - Jan-Gowth Chang
- Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
16
|
Gandica RG, Chung WK, Deng L, Goland R, Gallagher MP. Identifying monogenic diabetes in a pediatric cohort with presumed type 1 diabetes. Pediatr Diabetes 2015; 16:227-33. [PMID: 25082184 PMCID: PMC4767163 DOI: 10.1111/pedi.12150] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Monogenic diabetes (MD) is rare and can often be confused with type 1 diabetes (T1D) in a pediatric cohort. We sought to determine clinical criteria that could optimally identify candidates for genetic testing of two common forms of MD that alter therapy: glucokinase (GCK) and hepatocyte nuclear factor 1 alpha (HNF1α). RESEARCH DESIGN AND METHODS We performed a retrospective chart review of 939 patients with a presumed diagnosis of T1D, 6 months-20 yr of age, and identified four clinical criteria that were unusual for T1D and could warrant further evaluation for MD: (i) negative pancreatic autoantibodies, (ii) evidence of prolonged endogenous insulin production, or (iii) strong family history of diabetes in multiple generations. One hundred and twenty-one patients were identified as having one or more of these high-risk clinical criteria and were offered screening for mutations in GCK and HNF1α; 58 consented for genetic testing. RESULTS Of 58 patients with presumed T1D who underwent genetic testing, four were found to have GCK and one had HNF1α. No patients with only one high-risk feature were found to have MD. Of 10 patients who had two or more high risk criteria, five had MD (50%). CONCLUSION A high frequency of MD from mutations in GCK/HNF1α may be identified among pediatric diabetic patients originally considered to have T1D by performing genetic testing on those patients with multiple clinical risk factors for MD.
Collapse
Affiliation(s)
- Rachelle G. Gandica
- Division of Pediatric Endocrinology, Diabetes, and Metabolism, Columbia University Medical Center, New York, NY 10032, USA
| | - Wendy K. Chung
- Division of Molecular Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Liyong Deng
- Division of Molecular Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Robin Goland
- Division of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Mary Pat Gallagher
- Division of Pediatric Endocrinology, Diabetes, and Metabolism, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
17
|
Joint identification of genetic variants for physical activity in Korean population. Int J Mol Sci 2014; 15:12407-21. [PMID: 25026172 PMCID: PMC4139850 DOI: 10.3390/ijms150712407] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/11/2014] [Indexed: 01/30/2023] Open
Abstract
There has been limited research on genome-wide association with physical activity (PA). This study ascertained genetic associations between PA and 344,893 single nucleotide polymorphism (SNP) markers in 8842 Korean samples. PA data were obtained from a validated questionnaire that included information on PA intensity and duration. Metabolic equivalent of tasks were calculated to estimate the total daily PA level for each individual. In addition to single- and multiple-SNP association tests, a pathway enrichment analysis was performed to identify the biological significance of SNP markers. Although no significant SNP was found at genome-wide significance level via single-SNP association tests, 59 genetic variants mapped to 76 genes were identified via a multiple SNP approach using a bootstrap selection stability measure. Pathway analysis for these 59 variants showed that maturity onset diabetes of the young (MODY) was enriched. Joint identification of SNPs could enable the identification of multiple SNPs with good predictive power for PA and a pathway enriched for PA.
Collapse
|
18
|
Estrada K, Aukrust I, Bjørkhaug L, Burtt NP, Mercader JM, García-Ortiz H, Huerta-Chagoya A, Moreno-Macías H, Walford G, Flannick J, Williams AL, Gómez-Vázquez MJ, Fernandez-Lopez JC, Martínez-Hernández A, Jiménez-Morales S, Centeno-Cruz F, Mendoza-Caamal E, Revilla-Monsalve C, Islas-Andrade S, Córdova EJ, Soberón X, González-Villalpando ME, Henderson E, Wilkens LR, Le Marchand L, Arellano-Campos O, Ordóñez-Sánchez ML, Rodríguez-Torres M, Rodríguez-Guillén R, Riba L, Najmi LA, Jacobs SBR, Fennell T, Gabriel S, Fontanillas P, Hanis CL, Lehman DM, Jenkinson CP, Abboud HE, Bell GI, Cortes ML, Boehnke M, González-Villalpando C, Orozco L, Haiman CA, Tusié-Luna T, Aguilar-Salinas CA, Altshuler D, Njølstad PR, Florez JC, MacArthur DG. Association of a low-frequency variant in HNF1A with type 2 diabetes in a Latino population. JAMA 2014; 311:2305-14. [PMID: 24915262 PMCID: PMC4425850 DOI: 10.1001/jama.2014.6511] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE Latino populations have one of the highest prevalences of type 2 diabetes worldwide. OBJECTIVES To investigate the association between rare protein-coding genetic variants and prevalence of type 2 diabetes in a large Latino population and to explore potential molecular and physiological mechanisms for the observed relationships. DESIGN, SETTING, AND PARTICIPANTS Whole-exome sequencing was performed on DNA samples from 3756 Mexican and US Latino individuals (1794 with type 2 diabetes and 1962 without diabetes) recruited from 1993 to 2013. One variant was further tested for allele frequency and association with type 2 diabetes in large multiethnic data sets of 14,276 participants and characterized in experimental assays. MAIN OUTCOME AND MEASURES Prevalence of type 2 diabetes. Secondary outcomes included age of onset, body mass index, and effect on protein function. RESULTS A single rare missense variant (c.1522G>A [p.E508K]) was associated with type 2 diabetes prevalence (odds ratio [OR], 5.48; 95% CI, 2.83-10.61; P = 4.4 × 10(-7)) in hepatocyte nuclear factor 1-α (HNF1A), the gene responsible for maturity onset diabetes of the young type 3 (MODY3). This variant was observed in 0.36% of participants without type 2 diabetes and 2.1% of participants with it. In multiethnic replication data sets, the p.E508K variant was seen only in Latino patients (n = 1443 with type 2 diabetes and 1673 without it) and was associated with type 2 diabetes (OR, 4.16; 95% CI, 1.75-9.92; P = .0013). In experimental assays, HNF-1A protein encoding the p.E508K mutant demonstrated reduced transactivation activity of its target promoter compared with a wild-type protein. In our data, carriers and noncarriers of the p.E508K mutation with type 2 diabetes had no significant differences in compared clinical characteristics, including age at onset. The mean (SD) age for carriers was 45.3 years (11.2) vs 47.5 years (11.5) for noncarriers (P = .49) and the mean (SD) BMI for carriers was 28.2 (5.5) vs 29.3 (5.3) for noncarriers (P = .19). CONCLUSIONS AND RELEVANCE Using whole-exome sequencing, we identified a single low-frequency variant in the MODY3-causing gene HNF1A that is associated with type 2 diabetes in Latino populations and may affect protein function. This finding may have implications for screening and therapeutic modification in this population, but additional studies are required.
Collapse
Affiliation(s)
| | - Karol Estrada
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts2Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston3Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ingvild Aukrust
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway6Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Lise Bjørkhaug
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway5Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Noël P Burtt
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Josep M Mercader
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts7Center for Human Genetic Research and Diabetes Research Center (Diabetes Unit), Massachusetts General Hospital, Boston8Joint BSC-CRG-IRB Research Prog
| | | | - Alicia Huerta-Chagoya
- Instituto de Investigaciones Biomédicas, UNAM Unidad de Biología Molecular y Medicina Genómica, UNAM/INCMNSZ, Coyoacán, Mexico City, Mexico
| | | | - Geoffrey Walford
- Department of Medicine, Harvard Medical School, Boston, Massachusetts7Center for Human Genetic Research and Diabetes Research Center (Diabetes Unit), Massachusetts General Hospital, Boston
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts13Department of Molecular Biology, Harvard Medical School, Boston, Massachusetts
| | - Amy L Williams
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts14Department of Biological Sciences, Columbia University, New York, New York
| | - María J Gómez-Vázquez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | | | | | | | | | | | - Cristina Revilla-Monsalve
- Unidad de Investigación Médica en Enfermedades Metabólicas, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City
| | - Sergio Islas-Andrade
- Unidad de Investigación Médica en Enfermedades Metabólicas, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City
| | - Emilio J Córdova
- Instituto Nacional de Medicina Genómica, Tlalpan, Mexico City, Mexico
| | - Xavier Soberón
- Instituto Nacional de Medicina Genómica, Tlalpan, Mexico City, Mexico
| | - María E González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Publica, Mexico City, Mexico
| | - E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu
| | - Olimpia Arellano-Campos
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | - Maria L Ordóñez-Sánchez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | - Maribel Rodríguez-Torres
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | - Rosario Rodríguez-Guillén
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | - Laura Riba
- Instituto de Investigaciones Biomédicas, UNAM Unidad de Biología Molecular y Medicina Genómica, UNAM/INCMNSZ, Coyoacán, Mexico City, Mexico
| | - Laeya A Najmi
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway23Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Suzanne B R Jacobs
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Timothy Fennell
- The Genomics Platform, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Stacey Gabriel
- The Genomics Platform, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Pierre Fontanillas
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Craig L Hanis
- Human Genetics Center, University of Texas Health Science Center at Houston
| | - Donna M Lehman
- Department of Medicine, University of Texas Health Science Center at San Antonio
| | | | - Hanna E Abboud
- Department of Medicine, University of Texas Health Science Center at San Antonio
| | - Graeme I Bell
- Department of Human Genetics, University of Chicago, Chicago, Illinois28Department of Medicine, University of Chicago, Chicago, Illinois
| | - Maria L Cortes
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor
| | - Clicerio González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Publica, Mexico City, Mexico
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica, Tlalpan, Mexico City, Mexico
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles
| | - Teresa Tusié-Luna
- Instituto de Investigaciones Biomédicas, UNAM Unidad de Biología Molecular y Medicina Genómica, UNAM/INCMNSZ, Coyoacán, Mexico City, Mexico17Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | - Carlos A Aguilar-Salinas
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Sección XVI, Tlalpan, Mexico City, Mexico
| | - David Altshuler
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts3Department of Medicine, Harvard Medical School, Boston, Massachusetts7Center for Human Genetic Research and Diabetes Research Center (Diabetes Unit)
| | - Pål R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway5Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jose C Florez
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts3Department of Medicine, Harvard Medical School, Boston, Massachusetts7Center for Human Genetic Research and Diabetes Research Center (Diabetes Unit)
| | - Daniel G MacArthur
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts3Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Abstract
Most diabetes is polygenic in etiology, with (type 1 diabetes, T1DM) or without (type 2 diabetes, T2DM) an autoimmune basis. Genetic counseling for diabetes generally focuses on providing empiric risk information based on family history and/or the effects of maternal hyperglycemia on pregnancy outcome. An estimated one to five percent of diabetes is monogenic in nature, e.g., maturity onset diabetes of the young (MODY), with molecular testing and etiology-based treatment available. However, recent studies show that most monogenic diabetes is misdiagnosed as T1DM or T2DM. While efforts are underway to increase the rate of diagnosis in the diabetes clinic, genetic counselors and clinical geneticists are in a prime position to identify monogenic cases through targeted questions during a family history combined with working in conjunction with diabetes professionals to diagnose and assure proper treatment and familial risk assessment for individuals with monogenic diabetes.
Collapse
Affiliation(s)
- Stephanie A Stein
- Department of Medicine, Division of Endocrinology, Diabetes & Nutrition, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kristin L Maloney
- Department of Medicine, Division of Endocrinology, Diabetes & Nutrition, University of Maryland School of Medicine, Baltimore, Maryland ; Program in Genetics and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Toni I Pollin
- Department of Medicine, Division of Endocrinology, Diabetes & Nutrition, University of Maryland School of Medicine, Baltimore, Maryland ; Program in Genetics and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland ; Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
20
|
|
21
|
Pinelli M, Acquaviva F, Barbetti F, Caredda E, Cocozza S, Delvecchio M, Mozzillo E, Pirozzi D, Prisco F, Rabbone I, Sacchetti L, Tinto N, Toni S, Zucchini S, Iafusco D. Identification of candidate children for maturity-onset diabetes of the young type 2 (MODY2) gene testing: a seven-item clinical flowchart (7-iF). PLoS One 2013; 8:e79933. [PMID: 24244580 PMCID: PMC3823596 DOI: 10.1371/journal.pone.0079933] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 10/04/2013] [Indexed: 12/30/2022] Open
Abstract
MODY2 is the most prevalent monogenic form of diabetes in Italy with an estimated prevalence of about 0.5–1.5%. MODY2 is potentially indistinguishable from other forms of diabetes, however, its identification impacts on patients' quality of life and healthcare resources. Unfortunately, DNA direct sequencing as diagnostic test is not readily accessible and expensive. In addition current guidelines, aiming to establish when the test should be performed, proved a poor detection rate. Aim of this study is to propose a reliable and easy-to-use tool to identify candidate patients for MODY2 genetic testing. We designed and validated a diagnostic flowchart in the attempt to improve the detection rate and to increase the number of properly requested tests. The flowchart, called 7-iF, consists of 7 binary “yes or no” questions and its unequivocal output is an indication for whether testing or not. We tested the 7-iF to estimate its clinical utility in comparison to the clinical suspicion alone. The 7-iF, in a prospective 2-year study (921 diabetic children) showed a precision of about the 76%. Using retrospective data, the 7-iF showed a precision in identifying MODY2 patients of about 80% compared to the 40% of the clinical suspicion. On the other hand, despite a relatively high number of missing MODY2 patients, the 7-iF would not suggest the test for 90% of the non-MODY2 patients, demonstrating that a wide application of this method might 1) help less experienced clinicians in suspecting MODY2 patients and 2) reducing the number of unnecessary tests. With the 7-iF, a clinician can feel confident of identifying a potential case of MODY2 and suggest the molecular test without fear of wasting time and money. A Qaly-type analysis estimated an increase in the patients' quality of life and savings for the health care system of about 9 million euros per year.
Collapse
Affiliation(s)
- Michele Pinelli
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Fabio Acquaviva
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Fabrizio Barbetti
- Laboratory of Molecular Endocrinology and Metabolism, Bambino Gesù Children's Hospital, Scientific Institute - Laboratory of Mendelian Diabetes, Rome, Italy
| | | | - Sergio Cocozza
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Maurizio Delvecchio
- Pediatrics Unit, IRCCS Casa Sollievo della Sofferenza Hospital, S. Giovanni Rotondo (FG), Italy
| | - Enza Mozzillo
- Department of Pediatrics, University of Naples “Federico II”, Naples, Italy
| | - Daniele Pirozzi
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE – Advanced Biotechnologies S. C. a R. L., Naples, Italy
| | - Francesco Prisco
- Department of Pediatrics, Second University of Naples, Naples, Italy
| | - Ivana Rabbone
- Department of Pediatrics, University of Turin, Turin, Italy
| | - Lucia Sacchetti
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE – Advanced Biotechnologies S. C. a R. L., Naples, Italy
| | - Nadia Tinto
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE – Advanced Biotechnologies S. C. a R. L., Naples, Italy
| | - Sonia Toni
- Pediatric Diabetologic Unit, Meyer Children Hospital, Florence, Italy
| | | | - Dario Iafusco
- Department of Pediatrics, Second University of Naples, Naples, Italy
- * E-mail:
| | | |
Collapse
|
22
|
Thanabalasingham G, Kaur K, Talbot F, Colclough K, Mathews A, Taylor J, Ellard S, Owen KR. Atypical phenotype associated with reported GCK exon 10 deletions: Clinical judgement is needed alongside appropriate genetic investigations. Diabet Med 2013; 30:e233-8. [PMID: 23586928 DOI: 10.1111/dme.12210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/10/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Maturity-onset diabetes of the young (MODY) caused by heterozygous mutations in the glucokinase (GCK) gene typically presents with lifelong, stable, mild fasting hyperglycaemia. With the exception of pregnancy, patients with GCK-MODY usually do not require pharmacological therapy. We report two unrelated patients whose initial genetic test results indicated a deletion of GCK exon 10, but whose clinical phenotypes were not typical of GCK-MODY. CASE REPORTS In case 1, the patient was hyperglycaemic at diagnosis (glucose > 30 mmol/l) and elevated glucose levels > 10 mmol/l persisted after withdrawal of insulin therapy. The patient in case 2 was also hyperglycaemic at diagnosis [HbA1c > 86 mmol/mol (10%)], which improved with the introduction of oral hypoglycaemic agents. These clinical features were not consistent with GCK-MODY. Both patients had a single nucleotide variant that prevented multiplex ligation-dependent probe analysis, which generated a false positive result of a GCK exon 10 deletion. CONCLUSION False positive genetic results in these two unrelated cases were attributable to the presence of a rare single nucleotide variant that prevented ligation of the probe in the multiplex ligation-dependent probe analysis kit used and falsely indicated deletion of exon 10 within GCK. Both cases had clinical features that did not tally with the typical GCK-MODY phenotype. These cases emphasize the need to interpret the results of definitive genetic tests within the specific clinical context. Increased medical sequencing is likely to lead to more reports of novel mutations of uncertain significance. If genetic investigations do not agree with the clinical picture, clinicians should exercise caution when making therapeutic changes based on these results.
Collapse
Affiliation(s)
- G Thanabalasingham
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Iismaa SE, Aplin M, Holman S, Yiu TW, Jackson K, Burchfield JG, Mitchell CJ, O’Reilly L, Davenport A, Cantley J, Schmitz-Peiffer C, Biden TJ, Cooney GJ, Graham RM. Glucose homeostasis in mice is transglutaminase 2 independent. PLoS One 2013; 8:e63346. [PMID: 23717413 PMCID: PMC3661676 DOI: 10.1371/journal.pone.0063346] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/29/2013] [Indexed: 11/18/2022] Open
Abstract
Transglutaminase type 2 (TG2) has been reported to be a candidate gene for maturity onset diabetes of the young (MODY) because three different mutations that impair TG2 transamidase activity have been found in 3 families with MODY. TG2 null (TG2−/−) mice have been reported to be glucose intolerant and have impaired glucose-stimulated insulin secretion (GSIS). Here we rigorously evaluated the role of TG2 in glucose metabolism using independently generated murine models of genetic TG2 disruption, which show no compensatory enhanced expression of other TGs in pancreatic islets or other tissues. First, we subjected chow- or fat-fed congenic SV129 or C57BL/6 wild type (WT) and TG2−/− littermates, to oral glucose gavage. Blood glucose and serum insulin levels were similar for both genotypes. Pancreatic islets isolated from these animals and analysed in vitro for GSIS and cholinergic potentiation of GSIS, showed no significant difference between genotypes. Results from intraperitoneal glucose tolerance tests (GTTs) and insulin tolerance tests (ITTs) were similar for both genotypes. Second, we directly investigated the role of TG2 transamidase activity in insulin secretion using a coisogenic model that expresses a mutant form of TG2 (TG2R579A), which is constitutively active for transamidase activity. Intraperitoneal GTTs and ITTs revealed no significant differences between WT and TG2R579A/R579A mice. Given that neither deletion nor constitutive activation of TG2 transamidase activity altered basal responses, or responses to a glucose or insulin challenge, our data indicate that glucose homeostasis in mice is TG2 independent, and question a link between TG2 and diabetes.
Collapse
Affiliation(s)
- Siiri E. Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
- * E-mail: (SEI); (RMG)
| | - Mark Aplin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Sara Holman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Ting W. Yiu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
| | - Kristy Jackson
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - James G. Burchfield
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Christopher J. Mitchell
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Liam O’Reilly
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Aimee Davenport
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - James Cantley
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Carsten Schmitz-Peiffer
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Trevor J. Biden
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Gregory J. Cooney
- Diabetes and Obesity Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Robert M. Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
- * E-mail: (SEI); (RMG)
| |
Collapse
|
24
|
Shields BM, McDonald TJ, Ellard S, Campbell MJ, Hyde C, Hattersley AT. The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia 2012; 55:1265-72. [PMID: 22218698 PMCID: PMC3328676 DOI: 10.1007/s00125-011-2418-8] [Citation(s) in RCA: 209] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 11/24/2011] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Diagnosing MODY is difficult. To date, selection for molecular genetic testing for MODY has used discrete cut-offs of limited clinical characteristics with varying sensitivity and specificity. We aimed to use multiple, weighted, clinical criteria to determine an individual's probability of having MODY, as a crucial tool for rational genetic testing. METHODS We developed prediction models using logistic regression on data from 1,191 patients with MODY (n = 594), type 1 diabetes (n = 278) and type 2 diabetes (n = 319). Model performance was assessed by receiver operating characteristic (ROC) curves, cross-validation and validation in a further 350 patients. RESULTS The models defined an overall probability of MODY using a weighted combination of the most discriminative characteristics. For MODY, compared with type 1 diabetes, these were: lower HbA(1c), parent with diabetes, female sex and older age at diagnosis. MODY was discriminated from type 2 diabetes by: lower BMI, younger age at diagnosis, female sex, lower HbA(1c), parent with diabetes, and not being treated with oral hypoglycaemic agents or insulin. Both models showed excellent discrimination (c-statistic = 0.95 and 0.98, respectively), low rates of cross-validated misclassification (9.2% and 5.3%), and good performance on the external test dataset (c-statistic = 0.95 and 0.94). Using the optimal cut-offs, the probability models improved the sensitivity (91% vs 72%) and specificity (94% vs 91%) for identifying MODY compared with standard criteria of diagnosis <25 years and an affected parent. The models are now available online at www.diabetesgenes.org . CONCLUSIONS/INTERPRETATION We have developed clinical prediction models that calculate an individual's probability of having MODY. This allows an improved and more rational approach to determine who should have molecular genetic testing.
Collapse
Affiliation(s)
- B. M. Shields
- Peninsula NIHR Clinical Research Facility, Peninsula Medical School, University of Exeter, Barrack Road, Exeter, EX2 5DW UK
| | | | - S. Ellard
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - M. J. Campbell
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - C. Hyde
- Peninsula Technology Assessment Group, Peninsula Medical School, University of Exeter, Salmon Pool Lane, Exeter, UK
| | - A. T. Hattersley
- Peninsula NIHR Clinical Research Facility, Peninsula Medical School, University of Exeter, Barrack Road, Exeter, EX2 5DW UK
| |
Collapse
|
25
|
Kropff J, Selwood MP, McCarthy MI, Farmer AJ, Owen KR. Prevalence of monogenic diabetes in young adults: a community-based, cross-sectional study in Oxfordshire, UK. Diabetologia 2011; 54:1261-3. [PMID: 21350841 DOI: 10.1007/s00125-011-2090-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 02/01/2011] [Indexed: 12/16/2022]
|
26
|
Shields BM, Hicks S, Shepherd MH, Colclough K, Hattersley AT, Ellard S. Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia 2010; 53:2504-8. [PMID: 20499044 DOI: 10.1007/s00125-010-1799-4] [Citation(s) in RCA: 464] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 04/26/2010] [Indexed: 11/27/2022]
Abstract
AIMS/HYPOTHESIS Maturity-onset diabetes of the young is frequently misdiagnosed as type 1 or type 2 diabetes. A correct diagnosis of MODY is important for determining treatment, but can only be confirmed by molecular genetic testing. We aimed to compare the regional distribution of confirmed MODY cases in the UK and to estimate the minimum prevalence. METHODS UK referrals for genetic testing in 2,072 probands and 1,280 relatives between 1996 and 2009 were examined by region, country and test result. Referral rate and prevalence were calculated using UK Census 2001 figures. RESULTS MODY was confirmed in 1,177 (35%) patients, with HNF1A (52%) and GCK mutations (32%) being most frequent in probands confirmed with MODY. There was considerable regional variation in proband referral rates (from <20 per million in Wales and Northern Ireland to >50 per million for South West England and Scotland) and patients diagnosed with MODY (5.3 per million in Northern Ireland, 48.9 per million in South West England). Referral rates and confirmed cases were highly correlated (r = 0.96, p < 0.0001). The minimum prevalence of MODY was estimated to be 108 cases per million. CONCLUSIONS/INTERPRETATION Assuming this minimal prevalence throughout the UK then >80% of MODY is not diagnosed by molecular testing. The marked regional variation in the prevalence of confirmed MODY directly results from differences in referral rates. This could reflect variation in awareness of MODY or unequal access to genetic testing. Increased referral for diagnostic testing is required if the majority of MODY patients are to have the genetic diagnosis necessary for optimal treatment.
Collapse
Affiliation(s)
- B M Shields
- Peninsula Medical School, University of Exeter, Exeter, UK
| | | | | | | | | | | |
Collapse
|
27
|
Amed S, Dean HJ, Panagiotopoulos C, Sellers EAC, Hadjiyannakis S, Laubscher TA, Dannenbaum D, Shah BR, Booth GL, Hamilton JK. Type 2 diabetes, medication-induced diabetes, and monogenic diabetes in Canadian children: a prospective national surveillance study. Diabetes Care 2010; 33:786-91. [PMID: 20067956 PMCID: PMC2845028 DOI: 10.2337/dc09-1013] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 01/06/2010] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine in Canadian children aged <18 years the 1) incidence of type 2 diabetes, medication-induced diabetes, and monogenic diabetes; 2) clinical features of type 2 diabetes; and 3) coexisting morbidity associated with type 2 diabetes at diagnosis. RESEARCH DESIGN AND METHODS This Canadian prospective national surveillance study involved a network of pediatricians, pediatric endocrinologists, family physicians, and adult endocrinologists. Incidence rates were calculated using Canadian Census population data. Descriptive statistics were used to illustrate demographic and clinical features. RESULTS From a population of 7.3 million children, 345 cases of non-type 1 diabetes were reported. The observed minimum incidence rates of type 2, medication-induced, and monogenic diabetes were 1.54, 0.4, and 0.2 cases per 100,000 children aged <18 years per year, respectively. On average, children with type 2 diabetes were aged 13.7 years and 8% (19 of 227) presented before 10 years. Ethnic minorities were overrepresented, but 25% (57 of 227) of children with type 2 diabetes were Caucasian. Of children with type 2 diabetes, 95% (206 of 216) were obese and 37% (43 of 115) had at least one comorbidity at diagnosis. CONCLUSIONS This is the first prospective national surveillance study in Canada to report the incidence of type 2 diabetes in children and also the first in the world to report the incidence of medication-induced and monogenic diabetes. Rates of type 2 diabetes were higher than expected with important regional variation. These results support recommendations that screening for comorbidity should occur at diagnosis of type 2 diabetes.
Collapse
Affiliation(s)
- Shazhan Amed
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tsai FJ, Yang CF, Chen CC, Chuang LM, Lu CH, Chang CT, Wang TY, Chen RH, Shiu CF, Liu YM, Chang CC, Chen P, Chen CH, Fann CSJ, Chen YT, Wu JY. A genome-wide association study identifies susceptibility variants for type 2 diabetes in Han Chinese. PLoS Genet 2010; 6:e1000847. [PMID: 20174558 PMCID: PMC2824763 DOI: 10.1371/journal.pgen.1000847] [Citation(s) in RCA: 263] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 01/18/2010] [Indexed: 12/16/2022] Open
Abstract
To investigate the underlying mechanisms of T2D pathogenesis, we looked for diabetes susceptibility genes that increase the risk of type 2 diabetes (T2D) in a Han Chinese population. A two-stage genome-wide association (GWA) study was conducted, in which 995 patients and 894 controls were genotyped using the Illumina HumanHap550-Duo BeadChip for the first genome scan stage. This was further replicated in 1,803 patients and 1,473 controls in stage 2. We found two loci not previously associated with diabetes susceptibility in and around the genes protein tyrosine phosphatase receptor type D (PTPRD) (P = 8.54×10−10; odds ratio [OR] = 1.57; 95% confidence interval [CI] = 1.36–1.82), and serine racemase (SRR) (P = 3.06×10−9; OR = 1.28; 95% CI = 1.18–1.39). We also confirmed that variants in KCNQ1 were associated with T2D risk, with the strongest signal at rs2237895 (P = 9.65×10−10; OR = 1.29, 95% CI = 1.19–1.40). By identifying two novel genetic susceptibility loci in a Han Chinese population and confirming the involvement of KCNQ1, which was previously reported to be associated with T2D in Japanese and European descent populations, our results may lead to a better understanding of differences in the molecular pathogenesis of T2D among various populations. Type 2 diabetes (T2D) is a complex disease that involves many genes and environmental factors. Genome-wide and candidate-gene association studies have thus far identified at least 19 regions containing genes that may confer a risk for T2D. However, most of these studies were conducted with patients of European descent. We studied Chinese patients with T2D and identified two genes, PTPRD and SRR, that were not previously known to be involved in diabetes and are involved in biological pathways different from those implicated in T2D by previous association reports. PTPRD is a protein tyrosine phosphatase and may affect insulin signaling on its target cells. SRR encodes a serine racemase that synthesizes D-serine from L-serine. Both D-serine (coagonist) and the neurotransmitter glutamate bind to NMDA receptors and trigger excitatory neurotransmission in the brain. Glutamate signaling also regulates insulin and glucagon secretion in pancreatic islets. Thus, SRR and D-serine, in addition to regulating insulin and glucagon secretion, may play a role in the etiology of T2D. Our study suggests that, in different patient populations, different genes may confer risks for diabetes. Our findings may lead to a better understanding of the molecular pathogenesis of T2D.
Collapse
Affiliation(s)
- Fuu-Jen Tsai
- School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Genetics, Pediatrics and Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Chi-Fan Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- National Genotyping Center, Academia Sinica, Taipei, Taiwan
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chieh-Hsiang Lu
- Department of Internal Medicine, Endocrinology and Metabolism, Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Chwen-Tzuei Chang
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Tzu-Yuan Wang
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Rong-Hsing Chen
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chiung-Fang Shiu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Min Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Chun Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Pei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- National Genotyping Center, Academia Sinica, Taipei, Taiwan
| | - Cathy S. J. Fann
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- National Genotyping Center, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (Y-TC); (J-YW)
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- National Genotyping Center, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- * E-mail: (Y-TC); (J-YW)
| |
Collapse
|
29
|
Weintrob N, Stern E, Klipper-Aurbach Y, Phillip M, Gat-Yablonski G. Childhood obesity complicating the differential diagnosis of maturity-onset diabetes of the young and type 2 diabetes. Pediatr Diabetes 2008; 9:60-4. [PMID: 18036134 DOI: 10.1111/j.1399-5448.2007.00259.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To describe a proband with features of type 2 diabetes who was found to have concomitant maturity-onset diabetes of the young (MODY) and the consequent multigeneration genetic analysis. DESIGN Familial genetic analysis. SETTING Tertiary university medical center. PARTICIPANTS The proband was a 13.5-yr-old boy with marked non-ketotic hyperglycemia, obesity, systolic hypertension, and insulin resistance. His mother, maternal aunt, grandmother, and great grandmother had diabetes; his father was obese and had early ischemic heart disease. INTERVENTIONS Clinical examination, laboratory work-up, and DNA study. OUTCOME MEASURES Mutation in hepatocyte nuclear factor-1alpha gene, the most common cause of MODY. RESULTS The proband showed elevated C-peptide level and was negative for beta-cell antibodies. On genetic analysis for MODY, the 291fsinsC mutation was identified in all affected family members. A younger sister who was obese but had no signs of impaired glucose tolerance was also tested on the basis of these findings and was found to have the same mutation. CONCLUSIONS The patient, who presented with apparent type 2 diabetes, had concomitant MODY 3, inherited from his mother's side, and some features of type 2 diabetes secondary to marked obesity. This combination probably caused an earlier and more severe presentation of the disease and had significant implications for medical management. A search for MODY mutations should be considered in patients with a history of diabetes in three generations of one side of the family, even those in whom the clinical picture resembles type 2 diabetes.
Collapse
Affiliation(s)
- Naomi Weintrob
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tiqva, Israel
| | | | | | | | | |
Collapse
|
30
|
Moore AF, Florez JC. Genetic Susceptibility to Type 2 Diabetes and Implications for Antidiabetic Therapy. Annu Rev Med 2008; 59:95-111. [DOI: 10.1146/annurev.med.59.090706.135315] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Allan F. Moore
- Diabetes Unit (Department of Medicine) and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts; the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and the Department of Medicine, Harvard Medical School, Boston, Massachusetts;
| | - Jose C. Florez
- Diabetes Unit (Department of Medicine) and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts; the Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and the Department of Medicine, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
31
|
Laakso M. Challenges in studies of the genetic basis of Type 2 diabetes. Expert Rev Endocrinol Metab 2007; 2:451-459. [PMID: 30290420 DOI: 10.1586/17446651.2.4.451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The prevalence of diabetes is increasing worldwide in epidemic proportions. This increase is mainly due to increased incidence and prevalence of Type 2 diabetes, which accounts for 80-90% of all cases of diabetes. The susceptibility to develop Type 2 diabetes is determined by genetic and environmental factors. Major genes responsible for Type 2 diabetes have not yet been identified. The most replicated susceptibility gene for Type 2 diabetes is TCF7L2, recently published by investigators from Iceland. The second most widely replicated association between a genetic variation and the risk of Type 2 diabetes is that of the Pro12Ala polymorphism in the peroxisome proliferator-activated receptor γ2 gene. Furthermore, the common E23K polymorphism in the KCJN11 gene, encoding the ATP-sensitive potassium-channel subunit Kir6.2, and variants in the calpain-10 gene have been associated with increased susceptibility to Type 2 diabetes in meta-analyses. Several studies have investigated the possibility that rare, highly penetrant mutations in the maturity-onset diabetes of the young genes lead to monogenic diabetes, while common polymorphisms increase the susceptibility to Type 2 diabetes. Indeed, there is increasing evidence that single nucleotide polymorphisms in hepatic nuclear factor-4α are significantly associated with the risk of Type 2 diabetes. In this review, different approaches to identify susceptibility genes for Type 2 diabetes are discussed. In particular, the importance of prospective population-based cohort studies and prospective intervention studies are emphasized. Finally, genome-wide association studies using single nucleotide polymorphisms randomly spaced across the entire genome may be useful in the identification of susceptibility genes for Type 2 diabetes.
Collapse
Affiliation(s)
- Markku Laakso
- a Academy Professor, Department of Medicine, University of Kuopio and Kuopio University Hospital, 70210 Kuopio, Finland.
| |
Collapse
|
32
|
Abstract
Diabetes mellitus comprises a heterogeneous group of disorders characterized by chronic hyperglycemia. Type 1 and type 2 diabetes result from alterations of various genes, each having partial and additive effects. Thus, the inheritance pattern is rather complex, and environmental factors play an important role in the manifestation and clinical course of the disease. There is no genetic test to diagnose diabetes mellitus type 1 or type 2. However, certain susceptibility genes and genetic variations can be examined for specific scientific questions. Furthermore, defined genetic defects exist of pancreatic beta-cell function (maturity-onset diabetes of the young, mitochondrial diabetes) and insulin action (e.g. insulin resistance syndromes and lipodystrophy syndromes) resembling monogenic disorders. In these cases, genetic tests are crucial for the correct classification of the type of diabetes, genetic counseling, and initiation of the appropriate therapy regimen.
Collapse
Affiliation(s)
- U C Broedl
- Medizinische Klinik II, Klinikum Grosshadern der Universität München, Marchioninistrasse 15, 81377 Munich
| | | |
Collapse
|
33
|
Abstract
Evidence for a genetic basis for type 2 diabetes and the metabolic syndrome has been derived from studies of families, twins and populations with genetic admixture. Identification of genes associated with disease pathogenesis is now underway using techniques such as genome scanning by positional cloning and the candidate gene approach. Genome scanning in several different ethnic groups has identified chromosome regions harbouring type 2 diabetes susceptibility genes such as the novel gene, calpain 10 (CAPN10). The hepatic nuclear factor 4alpha (HNF4alpha) gene partly explains the linkage peak on chromosome 20, while the upstream transcription factor (USF1) is associated with familial combined hyperlipidaemia (FCHL) and maps close to the type 2 diabetes associated 1q peak. Peroxisome proliferator-activated receptor gamma (PPARgamma) was identified as a candidate gene based on its biology. A Pro12Ala variant of this gene has been associated with an increased risk of type 2 diabetes. Many genes accounting for monogenic forms of diabetes have been identified--such as maturity onset diabetes of the young (MODY); glucokinase (GCK) and HNF1alpha mutations being the most common causes of MODY. GCK variants result in 'mild' diabetes or impaired glucose tolerance (IGT) and relatively few cardiovascular complications, while HNF1alpha-associated MODY is more typical of type 2 diabetes, frequently being treated with sulphonylureas or insulin and resulting in microvascular complications. Testing for single gene disorders associated with type 2 diabetes and obesity may determine cause, prognosis and appropriate treatment; however, for the more common polygenic diseases this is not the case. In type 2 diabetes, molecular genetics has the potential to enhance understanding of disease pathogenesis, and help formulate preventative and treatment strategies.
Collapse
Affiliation(s)
- G A Hitman
- Centre for Diabetes and Metabolic Medicine, Institute of Cell and Molecular Science, Barts and the London Queen Mary's School of Medicine and Dentistry, London, UK.
| | | |
Collapse
|
34
|
Abstract
Conventional genetic analysis focuses on the genes that account for specific phenotypes, while traditional epidemiology is more concerned with the environmental causes and risk factors related to traits. Genetic epidemiology is an alliance of the 2 fields that focuses on both genetics, including allelic variants in different populations, and environment, in order to explain exactly how genes convey effects in different environmental contexts and to arrive at a more complete comprehension of the etiology of complex traits. In this review, we discuss the epidemiology of diabetes and the current understanding of the genetic bases of obesity and diabetes and provide suggestions for accelerated accumulation of clinically useful genetic information.
Collapse
Affiliation(s)
- M Alan Permutt
- Department of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri 63110-1010, USA.
| | | | | |
Collapse
|
35
|
Abstract
Type 2 diabetes (T2D) has become a health-care problem worldwide, with the rise in disease prevalence being all the more worrying as it not only affects the developed world but also developing nations with fewer resources to cope with yet another major disease burden. Furthermore, the problem is no longer restricted to the ageing population, as young adults and children are also being diagnosed with T2D. In recent years, there has been a surge in the number of genetic studies of T2D in attempts to identify some of the underlying risk factors. In this review, I highlight the main genes known to cause uncommon monogenic forms of diabetes (e.g. maturity-onset diabetes of the young--MODY--and insulin resistance syndromes), as well as describe some of the main approaches used to identify genes involved in the more common forms of T2D that result from the interaction between environmental risk factors and predisposing genotypes. Linkage and candidate gene studies have been highly successful in the identification of genes that cause the monogenic variants of diabetes and, although progress in the more common forms of T2D has been slow, a number of genes have now been reproducibly associated with T2D risk in multiple studies. These are discussed, as well as the main implications that the diabetes gene discoveries will have in diabetes treatment and prevention.
Collapse
Affiliation(s)
- I Barroso
- Metabolic Disease Group, The Wellcome Trust Sanger Institute, Cambridge, UK.
| |
Collapse
|
36
|
Avella J, Wetli CV, Wilson JC, Katz M, Hahn T. Fatal olanzapine-induced hyperglycemic ketoacidosis. Am J Forensic Med Pathol 2004; 25:172-5. [PMID: 15166774 DOI: 10.1097/01.paf.0000127391.85007.f4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Olanzapine is an antipsychotic medication linked to the development, or exacerbation of, type 2 diabetes mellitus. This report describes 3 patients being treated with olanzapine who died suddenly and unexpectedly with hyperglycemic ketoacidosis. All had olanzapine concentrations within the therapeutic range. Vitreous glucose concentrations ranged from 640 mg/dL to 833 mg/dL, and blood acetone concentrations from 25.6 mg/dL to 57.6 mg/dL. Beta-hydroxybutyrate concentrations in blood were from 55.2 mg/dL to 110 mg/dL. Low levels of isopropanol were also detected. None had a history or family history of diabetes mellitus. Glycolated (A1C) hemoglobin in 2 cases was 14.3% and 14.7%. No predisposing factors to olanzapine-induced diabetes were identified. It is recommended that chemical testing of patients dying suddenly while being treated with antipsychotic drugs include vitreous glucose and blood acetone determinations to elucidate the cause and mechanism of death in these patients. Warnings concerning this potentially fatal complication of olanzapine therapy should be included in standard pharmaceutical and prescription references.
Collapse
Affiliation(s)
- Joseph Avella
- Department of Health Services, Division of Medical-Legal and Forensic Investigations, Suffolk County, NY, USA
| | | | | | | | | |
Collapse
|
37
|
Cheung C, Akiyama TE, Kudo G, Gonzalez FJ. Hepatic expression of cytochrome P450s in hepatocyte nuclear factor 1-alpha (HNF1alpha)-deficient mice. Biochem Pharmacol 2003; 66:2011-20. [PMID: 14599559 DOI: 10.1016/s0006-2952(03)00586-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Hepatocyte nuclear factor 1 alpha (HNF1alpha) is a liver enriched homeodomain-containing transcription factor that has been shown to transactivate the promoters of several cytochrome P450 (CYP) genes, including CYP2E1, CYP1A2, CYP7A1, and CYP27, in vitro. In humans, mutations in HNF1alpha are linked to the occurrence of maturity onset diabetes of the young type 3, an autosomal dominant form of non-insulin-dependent diabetes mellitus in which afflicted subjects generally develop hyperglycemia before 25 years of age. Mice lacking HNF1alpha also develop similar phenotypes reminiscent of non-insulin-dependent diabetes mellitus. To investigate a potential role for HNF1alpha in the regulation of CYPs in vivo, the expression of major CYP genes from each family was examined in the livers of mice lacking HNF1alpha. Analysis of CYP gene expression revealed marked reductions in expression of Cyp1a2, Cyp2c29 and Cyp2e1, and a moderate reduction of Cyp3a11. In contrast Cyp2a5, Cyp2b10 and Cyp2d9 expression were elevated. There are also significant changes in the expression of genes encoding CYPs involved in fatty acid and bile acid metabolism characterized by a reduction in the expression of Cyp7b1, and Cyp27 as well as elevations in Cyp4a1/3, Cyp7a1, Cyp8b1, and Cyp39a1 expression. These results point to a critical role for HNF1alpha in the regulation of CYPs in vivo and suggest that this transcription factor may have an important influence on drug metabolism as well as lipid and bile acid homeostasis in maturity onset diabetes of the young type 3 diabetics.
Collapse
Affiliation(s)
- Connie Cheung
- Laboratory of Metabolism, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
38
|
Akiyama TE, Gonzalez FJ. Regulation of P450 genes by liver-enriched transcription factors and nuclear receptors. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1619:223-34. [PMID: 12573481 DOI: 10.1016/s0304-4165(02)00480-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cytochrome P450s (P450s) constitute a superfamily of heme-proteins that play an important role in the activation of chemical carcinogens, detoxification of numerous xenobiotics as well as in the oxidative metabolism of endogenous compounds such as steroids, fatty acids, prostaglandins, and leukotrienes. In addition, some P450s have important roles in physiological processes, such as steroidogenesis and the maintenance of bile acid and cholesterol homeostasis. Given their importance, the molecular mechanisms of P450 gene regulation have been intensely studied. Direct interactions between transcription factors, including nuclear receptors, with the promoters of P450 genes represent one of the primary means by which the expression of these genes is controlled. In this review, several liver-enriched transcription factors that play a role in the tissue-specific, developmental, and temporal regulation of P450s are discussed. In addition, the nuclear receptors that play a role in the fine control of cholesterol and bile acid homeostasis, in part, through their modulation of specific P450s, are discussed.
Collapse
Affiliation(s)
- Taro E Akiyama
- Laboratory of Metabolism, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | |
Collapse
|
39
|
Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med 2001; 345:971-80. [PMID: 11575290 DOI: 10.1056/nejmra002168] [Citation(s) in RCA: 667] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- S S Fajans
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, USA.
| | | | | |
Collapse
|