151
|
Ushijima K, Fukami M, Ayabe T, Narumi S, Okuno M, Nakamura A, Takahashi T, Ihara K, Ohkubo K, Tachikawa E, Nakayama S, Arai J, Kikuchi N, Kikuchi T, Kawamura T, Urakami T, Hata K, Nakabayashi K, Matsubara Y, Amemiya S, Ogata T, Yokota I, Sugihara S. Comprehensive screening for monogenic diabetes in 89 Japanese children with insulin-requiring antibody-negative type 1 diabetes. Pediatr Diabetes 2018; 19:243-250. [PMID: 28597946 DOI: 10.1111/pedi.12544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 04/03/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mutations in causative genes for neonatal diabetes or maturity-onset diabetes of the young have been identified in multiple patients with autoantibody-negative type 1 diabetes (T1D). OBJECTIVES We aimed to clarify the prevalence and phenotypic characteristics of monogenic abnormalities among 89 children with autoantibody-negative insulin-requiring T1D. METHODS Mutations in 30 genes were screened using next-generation sequencing, and copy-number alterations of 4 major causative genes were examined using multiplex-ligation-dependent probe amplification. We compared the clinical characteristics between mutation carriers and non-carriers. RESULTS We identified 11 probable pathogenic substitutions (6 in INS , 2 in HNF1A , 2 in HNF4A , and 1 in HNF1B ) in 11 cases, but no copy-number abnormalities. Only 2 mutation carriers had affected parents. De novo occurrence was confirmed for 3 mutations. The non-carrier group, but not the carrier group, was enriched with susceptible HLA alleles. Mutation carriers exhibited comparable phenotypes to those of non-carriers, except for a relatively normal body mass index (BMI) at diagnosis. CONCLUSIONS This study demonstrated significant genetic overlap between autoantibody-negative T1D and monogenic diabetes. Mutations in INS and HNF genes, but not those in GCK and other monogenic diabetes genes, likely play critical roles in children with insulin-requiring T1D. This study also suggests the relatively high de novo rates of INS and HNF mutations, and the etiological link between autoimmune abnormalities and T1D in the non-carrier group. Carriers of monogenic mutations show non-specific phenotypes among all T1D cases, although they are more likely to have a normal BMI at diagnosis than non-carriers.
Collapse
Affiliation(s)
- Kikumi Ushijima
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Tadayuki Ayabe
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, Sanaikai General Hospital, Misato, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Misako Okuno
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Akie Nakamura
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | - Kenji Ihara
- Department of Pediatrics, Oita University School of Medicine, Oita, Japan
| | - Kazuhiro Ohkubo
- Department of Pediatrics, Kyushu University School of Medicine, Fukuoka, Japan
| | - Emiko Tachikawa
- Department of Pediatrics, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Shoji Nakayama
- Department of Pediatrics, Mominoki Hospital, Kochi, Japan
| | - Junichi Arai
- Department of Pediatrics, Hosogi Hospital, Kochi, Japan
| | - Nobuyuki Kikuchi
- Department of Pediatrics, Yokohama City Minato Red Cross Hospital, Yokohama, Japan
| | - Toru Kikuchi
- Department of Pediatrics, Saitama Medical University Faculty of Medicine, Saitama, Japan
| | - Tomoyuki Kawamura
- Department of Pediatrics, Osaka City University School of Medicine, Osaka, Japan
| | - Tatsuhiko Urakami
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yoichi Matsubara
- Institute Director, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Shin Amemiya
- Department of Pediatrics, Saitama Medical University Faculty of Medicine, Saitama, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ichiro Yokota
- Department of Pediatrics, Division of Pediatric Endocrinology and Metabolism, Shikoku Medical Center for Children and Adults, Kagawa, Japan
| | - Shigetaka Sugihara
- Department of Pediatrics, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | | |
Collapse
|
152
|
Perleberg C, Kind A, Schnieke A. Genetically engineered pigs as models for human disease. Dis Model Mech 2018; 11:11/1/dmm030783. [PMID: 29419487 PMCID: PMC5818075 DOI: 10.1242/dmm.030783] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genetically modified animals are vital for gaining a proper understanding of disease mechanisms. Mice have long been the mainstay of basic research into a wide variety of diseases but are not always the most suitable means of translating basic knowledge into clinical application. The shortcomings of rodent preclinical studies are widely recognised, and regulatory agencies around the world now require preclinical trial data from nonrodent species. Pigs are well suited to biomedical research, sharing many similarities with humans, including body size, anatomical features, physiology and pathophysiology, and they already play an important role in translational studies. This role is set to increase as advanced genetic techniques simplify the generation of pigs with precisely tailored modifications designed to replicate lesions responsible for human disease. This article provides an overview of the most promising and clinically relevant genetically modified porcine models of human disease for translational biomedical research, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We briefly summarise the technologies involved and consider the future impact of recent technical advances. Summary: An overview of porcine models of human disease, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We summarise the technologies involved and potential future impact of recent technical advances.
Collapse
Affiliation(s)
- Carolin Perleberg
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
153
|
Huang X, Gong S, Ma Y, Cai X, Zhou L, Luo Y, Li M, Liu W, Zhang S, Zhang X, Ren Q, Zhu Y, Zhou X, Zhang R, Chen L, Gao X, Zhang F, Wang Y, Han X, Ji L. Lower Circulating miR-122 Level in Patients with HNF1A Variant-Induced Diabetes Compared with Type 2 Diabetes. J Diabetes Res 2018; 2018:7842064. [PMID: 30155490 PMCID: PMC6093029 DOI: 10.1155/2018/7842064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/02/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022] Open
Abstract
miR-122, the expression of which is regulated by several transcription factors, such as HNF1A, was recently reported to be associated with type 2 diabetes (T2DM) and hepatocellular carcinoma. HNF1A variants can cause diabetes and might be involved in the development of primary liver neoplasm. Differences in miR-122 expression among different types of diabetes have not been studied. This study aimed to investigate differences in serum miR-122 levels in Chinese patients with different forms of diabetes, including T2DM, type 1 diabetes (T1DM), HNF1A variant-induced diabetes (HNF1A-DM), glucokinase variant-induced diabetes (GCK-DM), and mitochondrial A3243G mutation-induced diabetes (MDM). In total, 12 HNF1A-DM patients, 24 gender-, age-, and body mass index-matched (1 : 2) T2DM patients and 24 healthy subjects were included in this study. In addition, 30 monogenic diabetes (11 GCK-DM and 19 MDM) and 17 T1DM patients were included. Fasted blood biochemistry and miR-122 were measured. The results showed that the HNF1A-DM patients had lower miR-122 levels [0.046 (0.023, 0.121)] than T2DM patients [0.165 (0.036, 0.939), P = 0.02] and healthy controls [0.249 (0.049, 1.234), P = 0.019]. The area under the curve of the receiver operating characteristic curve for miR-122 to discriminate HNF1A-DM and T2DM was 0.687 (95% CI: 0.52-0.86, P = 0.07). There was no difference in serum miR-122 among HNF1A-DM, GCK-DM, MDM, and T1DM patients. Lower serum miR-122 is a unique feature of HNF1A-DM patients and might partially explain the increased risk for liver neoplasm and abnormal lipid metabolism in HNF1A-DM patients.
Collapse
Affiliation(s)
- Xiuting Huang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Siqian Gong
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Yumin Ma
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Lingli Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Yingying Luo
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Meng Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Wei Liu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Simin Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Xiuying Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Qian Ren
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Yu Zhu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Xianghai Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Rui Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Ling Chen
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Xueying Gao
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Fang Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Yanai Wang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing, 100044, China
| |
Collapse
|
154
|
Firdous P, Nissar K, Ali S, Ganai BA, Shabir U, Hassan T, Masoodi SR. Genetic Testing of Maturity-Onset Diabetes of the Young Current Status and Future Perspectives. Front Endocrinol (Lausanne) 2018; 9:253. [PMID: 29867778 PMCID: PMC5966560 DOI: 10.3389/fendo.2018.00253] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a global epidemic problem growing exponentially in Asian countries posing a serious threat. Among diabetes, maturity-onset diabetes of the young (MODY) is a heterogeneous group of monogenic disorders that occurs due to β cell dysfunction. Genetic defects in the pancreatic β-cells result in the decrease of insulin production required for glucose utilization thereby lead to early-onset diabetes (often <25 years). It is generally considered as non-insulin dependent form of diabetes and comprises of 1-5% of total diabetes. Till date, 14 genes have been identified and mutation in them may lead to MODY. Different genetic testing methodologies like linkage analysis, restriction fragment length polymorphism, and DNA sequencing are used for the accurate and correct investigation of gene mutations associated with MODY. The next-generation sequencing has emerged as one of the most promising and effective tools to identify novel mutated genes related to MODY. Diagnosis of MODY is mainly relying on the sequential screening of the three marker genes like hepatocyte nuclear factor 1 alpha (HNF1α), hepatocyte nuclear factor 4 alpha (HNF4α), and glucokinase (GCK). Interestingly, MODY patients can be managed by diet alone for many years and may also require minimal doses of sulfonylureas. The primary objective of this article is to provide a review on current status of MODY, its prevalence, genetic testing/diagnosis, possible treatment, and future perspective.
Collapse
Affiliation(s)
- Parveena Firdous
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Kamran Nissar
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Sajad Ali
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Bashir Ahmad Ganai
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
- *Correspondence: Bashir Ahmad Ganai,
| | - Uzma Shabir
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Toyeeba Hassan
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Shariq Rashid Masoodi
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| |
Collapse
|
155
|
Brunerova L, Rahelić D, Ceriello A, Broz J. Use of oral antidiabetic drugs in the treatment of maturity-onset diabetes of the young: A mini review. Diabetes Metab Res Rev 2018; 34. [PMID: 28840639 DOI: 10.1002/dmrr.2940] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/29/2022]
Abstract
MODY (maturity-onset diabetes of the young) is a genetically linked group of clinically heterogeneous subtypes of diabetes. Roughly 5% of people with diabetes mellitus diagnosed prior to age 45 have MODY diabetes. Most of them have been erroneously diagnosed as patients with either type 1 or type 2 diabetes and, as a result, have been improperly treated. Genetic identification of MODY diabetes and its subtypes allows proper treatment and enables clinicians to switch many patients to oral antidiabetic agents, mainly sulphonylureas. However, some new classes of oral antidiabetic drugs have also been tested and found to be effective in MODY patients. We have searched for research articles and case reports written in full-text English or with an English abstract, using the following keywords: MODY and oral antidiabetic* in the databases Cochrane Library, PubMed, and Science Direct. Therapeutic options using currently standardized oral antidiabetic drugs (mainly sulphonylureas), as well as more experimental treatment with other classes of oral antidiabetic drugs in different types of MODY, are discussed, with special focus on the therapy of the most common MODY subtypes, including specific conditions such as pregnancy. This review article summarizes the currently available information about oral antidiabetic treatment of patients with MODY diabetes.
Collapse
Affiliation(s)
- Ludmila Brunerova
- Diabetes Center, 2nd Department of Medicine, Faculty Hospital Kralovske Vinohrady, Charles University 3rd Faculty of Medicine, Prague, Czech Republic
| | | | - Antonio Ceriello
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomedica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
- Department of Cardiovascular and Metabolic Diseases, IRCCS Multimedica Sesto San Giovanni (MI), Giovanni, Italy
| | - Jan Broz
- Department of Internal Medicine, Charles University 2nd Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
156
|
Abstract
Pancreatic β-cells are finely tuned to secrete insulin so that plasma glucose levels are maintained within a narrow physiological range (3.5-5.5 mmol/L). Hyperinsulinaemic hypoglycaemia (HH) is the inappropriate secretion of insulin in the presence of low plasma glucose levels and leads to severe and persistent hypoglycaemia in neonates and children. Mutations in 12 different key genes (ABCC8, KCNJ11, GLUD1, GCK, HADH, SLC16A1, UCP2, HNF4A, HNF1A, HK1, PGM1 and PMM2) that are involved in the regulation of insulin secretion from pancreatic β-cells have been described to be responsible for the underlying molecular mechanisms leading to congenital HH. In HH due to the inhibitory effect of insulin on lipolysis and ketogenesis there is suppressed ketone body formation in the presence of hypoglycaemia thus leading to increased risk of hypoglycaemic brain injury. Therefore, a prompt diagnosis and immediate management of HH is essential to avoid hypoglycaemic brain injury and long-term neurological complications in children. Advances in molecular genetics, imaging techniques (18F-DOPA positron emission tomography/computed tomography scanning), medical therapy and surgical advances (laparoscopic and open pancreatectomy) have changed the management and improved the outcome of patients with HH. This review article provides an overview to the background, clinical presentation, diagnosis, molecular genetics and therapy in children with different forms of HH.
Collapse
Affiliation(s)
- Hüseyin Demirbilek
- Hacettepe University Faculty of Medicine, Department of Paediatric Endocrinology, Ankara, Turkey
| | - Khalid Hussain
- Sidra Medical and Research Center, Clinic of Paediatric Medicine, Doha, Qatar
,* Address for Correspondence: Sidra Medical and Research Center, Clinic of Paediatric Medicine, Doha, Qatar Phone: +974-30322007 E-mail:
| |
Collapse
|
157
|
Abstract
Diabetes mellitus (DM) is a critical and long-term disorder due to the insufficient production of insulin by the pancreas or ineffective use of insulin by the body. Importantly, cardiovascular disease (CVD) has long been thought to be linked with diabetes. Despite more diabetic individuals surviving from better medications and treatments, there has been significant rise in the morbidity and mortality from CVD. Indeed, the classification of DM based on the electrocardiogram signals of the heart will be an advantageous system. Further, computer-aided classification of DM with integrated algorithms may enhance the execution of the system. In this paper, we have reviewed various studies using heart rate variability signals for automated classification of diabetes. Furthermore, the different techniques used to extract the features and the efficiency of the classification systems are discussed.
Collapse
Affiliation(s)
- MUHAMMAD ADAM
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore
| | - JEN HONG TAN
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore
| | - EDDIE Y. K. NG
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
158
|
Nault JC, Paradis V, Cherqui D, Vilgrain V, Zucman-Rossi J. Molecular classification of hepatocellular adenoma in clinical practice. J Hepatol 2017; 67:1074-1083. [PMID: 28733222 DOI: 10.1016/j.jhep.2017.07.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/29/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022]
Abstract
Hepatocellular adenomas (HCA) are rare benign liver tumors occurring in young women taking contraception. They are associated with rare complications such as bleeding or malignant transformation into hepatocellular carcinoma. A molecular classification has divided HCA in several subgroups linked with risk factors, clinical behaviour, histological features and imaging: HNF1A inactivated HCA, Inflammatory HCA, CTNNB1 mutated HCA in exon 3, CTNNB1 mutated in exon 7 and 8 HCA, sonic hedgehog HCA and unclassified HCA. CTNNB1 mutated HCA in exon 3 and sonic hedgehog HCA have been linked with a high risk of malignant transformation and bleeding respectively. Herein, we review how molecular classification has modified our understanding of the pathophysiology and risk factors of HCA development, analysing its impact on clinical care in the field of diagnosis and therapeutic stratification.
Collapse
Affiliation(s)
- Jean-Charles Nault
- Unité Mixte de Recherche 1162, Génomique fonctionnelle des tumeurs solides, Institut National de la Santé et de la Recherche Médicale, Paris, France; Liver Unit, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France; Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France.
| | - Valérie Paradis
- Pathology Department, Beaujon Hospital, APHP, Clichy 92110, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France; INSERM UMR 1149, Inflammation Research Center, Paris-Diderot University, Paris
| | - Daniel Cherqui
- Hepatobiliary Centre, Paul Brousse Hospital - Université Paris Sud, Institut National de la Santé et de la Recherche Médicale U1193, 14 Avenue Paul Vaillant Couturier, 94800 Villejuif, France
| | - Valérie Vilgrain
- University Paris Diderot, Sorbonne Paris Cité, Paris, France; Department of Radiology, University Hospitals Paris Nord Val de Seine, Beaujon, Clichy, Hauts-de-Seine, France; INSERM U1149, centre de recherche biomédicale Bichat-Beaujon, CRB3 Paris, France
| | - Jessica Zucman-Rossi
- Unité Mixte de Recherche 1162, Génomique fonctionnelle des tumeurs solides, Institut National de la Santé et de la Recherche Médicale, Paris, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Assistance Publique-Hôpitaux de Paris, Hopital Europeen Georges Pompidou, F-75015 Paris, France
| |
Collapse
|
159
|
Globa E, Zelinska N, Elblova L, Dusatkova P, Cinek O, Lebl J, Colclough K, Ellard S, Pruhova S. MODY in Ukraine: genes, clinical phenotypes and treatment. J Pediatr Endocrinol Metab 2017; 30:1095-1103. [PMID: 28862987 DOI: 10.1515/jpem-2017-0075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/24/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Maturity-onset diabetes of the young (MODY) has not been previously studied in Ukraine. We investigated the genetic etiology in a selected cohort of patients with diabetes diagnosed before 18 years of age, and in their family members. METHODS Genetic testing of the most prevalent MODY genes (GCK, HNF1A, HNF4A, HNF1B and INS) was undertaken for 36 families (39 affected individuals) by Sanger or targeted next generation sequencing. RESULTS A genetic diagnosis of MODY was made in 15/39 affected individuals from 12/36 families (33%). HNF1A and HNF4A MODY were the most common subtypes, accounting for 9/15 of MODY cases. Eight patients with HNF1A or HNF4A MODY and inadequate glycemic control were successfully transferred to sulfonylureas. Median HbA1c decreased from 67 mmol/mol (range 58-69) to 47 mmol/mol (range 43-50) (8.3% [7.5-8.5] to 6.4% [6.1-6.7]) 3 months after transfer (p=0.006). CONCLUSIONS Genetic testing identified pathogenic HNF1A and HNF4A variants as the most common cause of MODY in Ukraine. Transfer to sulfonylureas substantially improved the glycemic control of these patients.
Collapse
|
160
|
Patel KA, Kettunen J, Laakso M, Stančáková A, Laver TW, Colclough K, Johnson MB, Abramowicz M, Groop L, Miettinen PJ, Shepherd MH, Flanagan SE, Ellard S, Inagaki N, Hattersley AT, Tuomi T, Cnop M, Weedon MN. Heterozygous RFX6 protein truncating variants are associated with MODY with reduced penetrance. Nat Commun 2017; 8:888. [PMID: 29026101 PMCID: PMC5638866 DOI: 10.1038/s41467-017-00895-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/04/2017] [Indexed: 12/20/2022] Open
Abstract
Finding new causes of monogenic diabetes helps understand glycaemic regulation in humans. To find novel genetic causes of maturity-onset diabetes of the young (MODY), we sequenced MODY cases with unknown aetiology and compared variant frequencies to large public databases. From 36 European patients, we identify two probands with novel RFX6 heterozygous nonsense variants. RFX6 protein truncating variants are enriched in the MODY discovery cohort compared to the European control population within ExAC (odds ratio = 131, P = 1 × 10-4). We find similar results in non-Finnish European (n = 348, odds ratio = 43, P = 5 × 10-5) and Finnish (n = 80, odds ratio = 22, P = 1 × 10-6) replication cohorts. RFX6 heterozygotes have reduced penetrance of diabetes compared to common HNF1A and HNF4A-MODY mutations (27, 70 and 55% at 25 years of age, respectively). The hyperglycaemia results from beta-cell dysfunction and is associated with lower fasting and stimulated gastric inhibitory polypeptide (GIP) levels. Our study demonstrates that heterozygous RFX6 protein truncating variants are associated with MODY with reduced penetrance.Maturity-onset diabetes of the young (MODY) is the most common subtype of familial diabetes. Here, Patel et al. use targeted DNA sequencing of MODY patients and large-scale publically available data to show that RFX6 heterozygous protein truncating variants cause reduced penetrance MODY.
Collapse
Affiliation(s)
- Kashyap A Patel
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Jarno Kettunen
- Department of Endocrinology, Abdominal Centre, Helsinki University Hospital, Helsinki, 00029, Finland
- Folkhalsan Research Center, University of Helsinki, Helsinki, 00014, Finland
- Research Program of Diabetes and Obesity, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
| | - Markku Laakso
- Department of Medicine, Kuopio University Hospital, Kuopio, 70029, Finland
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, 70029, Finland
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, 70029, Finland
| | - Thomas W Laver
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Kevin Colclough
- Department of Molecular Genetics, Royal Devon and Exeter National Health Service Foundation Trust, Exeter, EX2 5DW, UK
| | - Matthew B Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Marc Abramowicz
- IRIBHM, Genetics Department, Erasmus Hospital, Université Libre de Bruxelles, Brussels, 1070, Belgium
| | - Leif Groop
- Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Skåne University Hospital, Malmö, SE, 20502, Sweden
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00100, Finland
| | - Päivi J Miettinen
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland
- Molecular Neurology and Biomedicum Stem Cell Centre, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Maggie H Shepherd
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Sarah E Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Sian Ellard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Tiinamaija Tuomi
- Department of Endocrinology, Abdominal Centre, Helsinki University Hospital, Helsinki, 00029, Finland
- Folkhalsan Research Center, University of Helsinki, Helsinki, 00014, Finland
- Research Program of Diabetes and Obesity, Research Programs Unit, University of Helsinki, Helsinki, 00014, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00100, Finland
| | - Miriam Cnop
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, 1070, Belgium.
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, 1070, Belgium.
| | - Michael N Weedon
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK.
| |
Collapse
|
161
|
Labriet A, De Mattia E, Cecchin E, Lévesque É, Jonker D, Couture F, Buonadonna A, D'Andrea M, Villeneuve L, Toffoli G, Guillemette C. Improved Progression-Free Survival in Irinotecan-Treated Metastatic Colorectal Cancer Patients Carrying the HNF1A Coding Variant p.I27L. Front Pharmacol 2017; 8:712. [PMID: 29066969 PMCID: PMC5641335 DOI: 10.3389/fphar.2017.00712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/22/2017] [Indexed: 01/15/2023] Open
Abstract
Hepatocyte nuclear factor 1-alpha (HNF1A) is a liver-enriched transcription factor that plays a key role in many aspects of hepatic functions including detoxification processes. We examined whether HNF1A polymorphisms are associated with clinical outcomes in two independent cohorts combining 417 European ancestry patients with metastatic colorectal cancer (mCRC) treated with irinotecan-based chemotherapy. The intronic rs2244608A>G marker was predictive of an improved progression-free survival with a trend in the Canadian cohort and reaching significance in the Italian cohort, with hazard ratios (HR) of 0.74 and 0.72, P = 0.076 and 0.038, respectively. A strong association between rs2244608A>G and improved PFS was found in the combined analysis of both cohorts (HR = 0.72; P = 0.002). Consistent with an altered HNF1A function, mCRC carriers of the rs2244608G minor allele displayed enhanced drug exposure by 45% (P = 0.032) compared to non-carriers. In Caucasians, rs2244608A>G is in strong linkage with the coding variant rs1169288c.79A>C (HNF1A p.I27L). In healthy donors, we observed an altered hepatic (ABCC1, P = 0.009, ABCC2, P = 0.048 and CYP3A5, P = 0.001; n = 89) and intestinal (TOP1, P = 0.004; n = 75) gene expression associated with the rs1169288C allele. In addition, the rs1169288C polymorphism could significantly increase the ABCC1 promoter activity by 27% (P = 0.008) and 15% (P = 0.041) in the human kidney HEK293 and the human liver HepG2 cell lines, respectively. Our findings suggest that the HNF1A rs2244608, or the tightly linked functional coding variant p.I27L, might be a potential prognostic marker with irinotecan-based regimens.
Collapse
Affiliation(s)
- Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Elena De Mattia
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Éric Lévesque
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Derek Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Félix Couture
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Angela Buonadonna
- Medical Oncology Unit, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| |
Collapse
|
162
|
Demirbilek H, Rahman SA, Buyukyilmaz GG, Hussain K. Diagnosis and treatment of hyperinsulinaemic hypoglycaemia and its implications for paediatric endocrinology. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2017; 2017:9. [PMID: 28855921 PMCID: PMC5575922 DOI: 10.1186/s13633-017-0048-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/15/2017] [Indexed: 12/14/2022]
Abstract
Glucose homeostasis requires appropriate and synchronous coordination of metabolic events and hormonal activities to keep plasma glucose concentrations in a narrow range of 3.5–5.5 mmol/L. Insulin, the only glucose lowering hormone secreted from pancreatic β-cells, plays the key role in glucose homeostasis. Insulin release from pancreatic β-cells is mainly regulated by intracellular ATP-generating metabolic pathways. Hyperinsulinaemic hypoglycaemia (HH), the most common cause of severe and persistent hypoglycaemia in neonates and children, is the inappropriate secretion of insulin which occurs despite low plasma glucose levels leading to severe and persistent hypoketotic hypoglycaemia. Mutations in 12 different key genes (ABCC8, KCNJ11, GLUD1, GCK, HADH, SLC16A1, UCP2, HNF4A, HNF1A, HK1, PGM1 and PMM2) constitute the underlying molecular mechanisms of congenital HH. Since insulin supressess ketogenesis, the alternative energy source to the brain, a prompt diagnosis and immediate management of HH is essential to avoid irreversible hypoglycaemic brain damage in children. Advances in molecular genetics, imaging methods (18F–DOPA PET-CT), medical therapy and surgical approach (laparoscopic and open pancreatectomy) have changed the management and improved the outcome of patients with HH. This up to date review article provides a background to the diagnosis, molecular genetics, recent advances and therapeutic options in the field of HH in children.
Collapse
Affiliation(s)
- Huseyin Demirbilek
- Department of Paediatric Endocrinology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Sofia A Rahman
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, 30 Guilford Street, London, WC1N 1EH UK
| | - Gonul Gulal Buyukyilmaz
- Department of Paediatric Endocrinology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Khalid Hussain
- Department of Paediatric Medicine Sidra Medical & Research Center, OPC, C6-337, PO Box 26999, Doha, Qatar
| |
Collapse
|
163
|
Abstract
The precision medicine approach of tailoring treatment to the individual characteristics of each patient or subgroup has been a great success in monogenic diabetes subtypes, MODY and neonatal diabetes. This review examines what has led to the success of a precision medicine approach in monogenic diabetes (precision diabetes) and outlines possible implications for type 2 diabetes. For monogenic diabetes, the molecular genetics can define discrete aetiological subtypes that have profound implications on diabetes treatment and can predict future development of associated clinical features, allowing early preventative or supportive treatment. In contrast, type 2 diabetes has overlapping polygenic susceptibility and underlying aetiologies, making it difficult to define discrete clinical subtypes with a dramatic implication for treatment. The implementation of precision medicine in neonatal diabetes was simple and rapid as it was based on single clinical criteria (diagnosed <6 months of age). In contrast, in MODY it was more complex and slow because of the lack of single criteria to identify patients, but it was greatly assisted by the development of a diagnostic probability calculator and associated smartphone app. Experience in monogenic diabetes suggests that successful adoption of a precision diabetes approach in type 2 diabetes will require simple, quick, easily accessible stratification that is based on a combination of routine clinical data, rather than relying on newer technologies. Analysing existing clinical data from routine clinical practice and trials may provide early success for precision medicine in type 2 diabetes.
Collapse
Affiliation(s)
- Andrew T Hattersley
- The Institute of Biomedical and Clinical Science, University of Exeter Medical School, RILD Building, Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK.
| | - Kashyap A Patel
- The Institute of Biomedical and Clinical Science, University of Exeter Medical School, RILD Building, Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| |
Collapse
|
164
|
Yang G, Chu PL, Rump LC, Le TH, Stegbauer J. ACE2 and the Homolog Collectrin in the Modulation of Nitric Oxide and Oxidative Stress in Blood Pressure Homeostasis and Vascular Injury. Antioxid Redox Signal 2017; 26:645-659. [PMID: 27889958 DOI: 10.1089/ars.2016.6950] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Hypertension is the leading risk factor causing mortality and morbidity worldwide. Angiotensin (Ang) II, the most active metabolite of the renin-angiotensin system, plays an outstanding role in the pathogenesis of hypertension and vascular injury. Activation of angiotensin converting enzyme 2 (ACE2) has shown to attenuate devastating effects of Ang II in the cardiovascular system by reducing Ang II degradation and increasing Ang-(1-7) generation leading to Mas receptor activation. Recent Advances: Activation of the ACE2/Ang-(1-7)/Mas receptor axis reduces hypertension and improves vascular injury mainly through an increased nitric oxide (NO) bioavailability and decreased reactive oxygen species production. Recent studies reported that shedding of the enzymatically active ectodomain of ACE2 from the cell surface seems to regulate its activity and serves as an interorgan communicator in cardiovascular disease. In addition, collectrin, an ACE2 homolog with no catalytic activity, regulates blood pressure through an NO-dependent mechanism. CRITICAL ISSUES Large body of experimental data confirmed sustained beneficial effects of ACE2/Ang-(1-7)/Mas receptor axis activation on hypertension and vascular injury. Experimental studies also suggest that activation of collectrin might be beneficial in hypertension and endothelial dysfunction. Their role in clinical hypertension is unclear as selective and reliable activators of both axes are not yet available. FUTURE DIRECTIONS This review will highlight the results of recent research progress that illustrate the role of both ACE and collectrin in the modulation of NO and oxidative stress in blood pressure homeostasis and vascular injury, providing evidence for the potential therapeutic application of ACE2 and collectrin in hypertension and vascular disease. Antioxid. Redox Signal. 26, 645-659.
Collapse
Affiliation(s)
- Guang Yang
- 1 Department of Nephrology, Medical Faculty, Heinrich-Heine University Düsseldorf , Düsseldorf, Germany
| | - Pei-Lun Chu
- 2 Division of Nephrology, Department of Medicine, University of Virginia , Charlottesville, Virginia.,3 Department of Internal Medicine, Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Lars C Rump
- 1 Department of Nephrology, Medical Faculty, Heinrich-Heine University Düsseldorf , Düsseldorf, Germany
| | - Thu H Le
- 2 Division of Nephrology, Department of Medicine, University of Virginia , Charlottesville, Virginia
| | - Johannes Stegbauer
- 1 Department of Nephrology, Medical Faculty, Heinrich-Heine University Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
165
|
Santana LS, Caetano LA, Costa-Riquetto AD, Quedas EPS, Nery M, Collett-Solberg P, Boguszewski MCS, Vendramini MF, Crisostomo LG, Floh FO, Zarabia ZI, Kohara SK, Guastapaglia L, Passone CGB, Sewaybricker LE, Jorge AAL, Teles MG. Clinical application of ACMG-AMP guidelines in HNF1A and GCK variants in a cohort of MODY families. Clin Genet 2017; 92:388-396. [PMID: 28170077 DOI: 10.1111/cge.12988] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 01/05/2023]
Abstract
Maturity-onset diabetes of the young (MODY) is a form of monogenic diabetes with autosomal dominant inheritance. GCK -MODY and HNF1A -MODY are the prevalent subtypes. Currently, there is growing concern regarding the correct interpretation of molecular genetic findings. The American College of Medical Genetics and Genomics (ACMG) updated guidelines to interpret and classify molecular variants. This study aimed to determine the prevalence of MODY ( GCK / HNF1A ) in a large cohort of Brazilian families, to report variants related to phenotype, and to classify them according to ACMG guidelines. One hundred and nine probands were investigated, 45% with clinical suspicion of GCK -MODY and 55% with suspicion of HNF1A -MODY. Twenty-five different variants were identified in GCK gene (30 probands-61% of positivity), and 7 variants in HNF1A (10 probands-17% of positivity). Fourteen of them were novel (12- GCK /2- HNF1A ). ACMG guidelines were able to classify a large portion of variants as pathogenic (36%- GCK /86%- HNF1A ) and likely pathogenic (44%- GCK /14%- HNF1A ), with 16% (5/32) as uncertain significance. This allows us to determine the pathogenicity classification more efficiently, and also reinforces the suspected associations with the phenotype among novel variants.
Collapse
Affiliation(s)
- L S Santana
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Laboratory of Molecular & Cellular Endocrinology/LIM25, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - L A Caetano
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Laboratory of Molecular & Cellular Endocrinology/LIM25, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil.,Diabetes Unit, Clinics Hospital, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - A D Costa-Riquetto
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Laboratory of Molecular & Cellular Endocrinology/LIM25, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil.,Diabetes Unit, Clinics Hospital, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - E P S Quedas
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Laboratory of Molecular & Cellular Endocrinology/LIM25, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - M Nery
- Diabetes Unit, Clinics Hospital, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - P Collett-Solberg
- Department of Endocrinology, University of Rio de Janeiro State (UERJ), Rio de Janeiro, RJ, Brazil
| | - M C S Boguszewski
- Departamento de Pediatria, Universidade Federal do Paraná (UFPR), Curitiba, PR, Brazil
| | - M F Vendramini
- Serviço de Endocrinologia, Hospital do Servidor Público Estadual de São Paulo (HSPE-SP), Sao Paulo, SP, Brazil
| | - L G Crisostomo
- Serviço de Endocrinologia, Hospital Israelita Albert Eisntein, Sao Paulo, SP, Brazil.,Faculdade de Medicina, Centro Universitário São Camilo, Sao Paulo, SP, Brazil
| | - F O Floh
- Serviço de Endocrinologia, Hospital Israelita Albert Eisntein, Sao Paulo, SP, Brazil
| | - Z I Zarabia
- Serviço de Endocrinologia, Hospital Infantil Dr. Jeser Amarante Faria, Joinville, SC, Brazil
| | - S K Kohara
- Serviço de Endocrinologia, Universidade da Região de Joinville (UNIVILLE), Joinville, SC, Brazil
| | - L Guastapaglia
- Serviço de Endocrinologia, Hospital do Servidor Público Municipal de São Paulo (HSPM-SP), Sao Paulo, SP, Brazil
| | - C G B Passone
- Instituto da Criança, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), Sao Paulo, SP, Brazil
| | - L E Sewaybricker
- Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - A A L Jorge
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Laboratory of Molecular & Cellular Endocrinology/LIM25, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - M G Teles
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Laboratory of Molecular & Cellular Endocrinology/LIM25, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil.,Diabetes Unit, Clinics Hospital, School of Medicine, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| |
Collapse
|
166
|
Abstract
Hepatitis C virus (HCV) infection often causes intrahepatic diseases, such as chronic hepatitis, liver chirrohsis, and hepatocellular carcinoma (HCC). Moreover, HCV infection exhibits various extrahepatic manifestations, such as thyroiditis, glucose and lipid metabolic disorder, and iron metabolic disorder. HCV infection is often associated with type 2 diabetes, involving hepatic fibrosis and poor prognosis. Type 2 diabetes increases the risk of HCC. We have been investigating molecular mechanisms of HCV-induced glucose metabolic disorder and we reported that HCV infection promotes hepatic gluconeogenesis through forkhead box O1 (FoxO1)-dependent pathway and that HCV infection suppresses the cell surface expression of glucose transporter 2 (GLUT2), resulting in suppression of glucose uptake. We have found that HCV NS5A protein plays important roles in these two independent pathways. Here we discuss the roles of HCV NS5A in HCV-induced glucose metabolic disorder.
Collapse
|
167
|
Sato Y, Tsuyama T, Sato C, Karim MF, Yoshizawa T, Inoue M, Yamagata K. Hypoxia reduces HNF4α/MODY1 protein expression in pancreatic β-cells by activating AMP-activated protein kinase. J Biol Chem 2017; 292:8716-8728. [PMID: 28364040 DOI: 10.1074/jbc.m116.767574] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/29/2017] [Indexed: 12/12/2022] Open
Abstract
Hypoxia plays a role in the deterioration of β-cell function. Hepatocyte nuclear factor 4α (HNF4α) has an important role in pancreatic β-cells, and mutations of the human HNF4A gene cause a type of maturity-onset diabetes of the young (MODY1). However, it remains unclear whether hypoxia affects the expression of HNF4α in β-cells. Here, we report that hypoxia reduces HNF4α protein expression in β-cells. Hypoxia-inducible factor was not involved in the down-regulation of HNF4α under hypoxic conditions. The down-regulation of HNF4α was dependent on the activation of AMP-activated protein kinase (AMPK), and the reduction of HNF4α protein expression by metformin, an AMPK activator, and hypoxia was inhibited by the overexpression of a kinase-dead (KD) form of AMPKα2. In addition, hypoxia decreased the stability of the HNF4α protein, and the down-regulation of HNF4α was sensitive to proteasome inhibitors. Adenovirus-mediated overexpression of KD-AMPKα2 improved insulin secretion in metformin-treated islets, hypoxic islets, and ob/ob mouse islets. These results suggest that down-regulation of HNF4α could be of importance in β-cell dysfunction by hypoxia.
Collapse
Affiliation(s)
- Yoshifumi Sato
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Tomonori Tsuyama
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Chinami Sato
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Md Fazlul Karim
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Tatsuya Yoshizawa
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Masahiro Inoue
- the Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Kazuya Yamagata
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| |
Collapse
|
168
|
Najmi LA, Aukrust I, Flannick J, Molnes J, Burtt N, Molven A, Groop L, Altshuler D, Johansson S, Bjørkhaug L, Njølstad PR. Functional Investigations of HNF1A Identify Rare Variants as Risk Factors for Type 2 Diabetes in the General Population. Diabetes 2017; 66:335-346. [PMID: 27899486 PMCID: PMC5860263 DOI: 10.2337/db16-0460] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/18/2016] [Indexed: 12/18/2022]
Abstract
Variants in HNF1A encoding hepatocyte nuclear factor 1α (HNF-1A) are associated with maturity-onset diabetes of the young form 3 (MODY 3) and type 2 diabetes. We investigated whether functional classification of HNF1A rare coding variants can inform models of diabetes risk prediction in the general population by analyzing the effect of 27 HNF1A variants identified in well-phenotyped populations (n = 4,115). Bioinformatics tools classified 11 variants as likely pathogenic and showed no association with diabetes risk (combined minor allele frequency [MAF] 0.22%; odds ratio [OR] 2.02; 95% CI 0.73-5.60; P = 0.18). However, a different set of 11 variants that reduced HNF-1A transcriptional activity to <60% of normal (wild-type) activity was strongly associated with diabetes in the general population (combined MAF 0.22%; OR 5.04; 95% CI 1.99-12.80; P = 0.0007). Our functional investigations indicate that 0.44% of the population carry HNF1A variants that result in a substantially increased risk for developing diabetes. These results suggest that functional characterization of variants within MODY genes may overcome the limitations of bioinformatics tools for the purposes of presymptomatic diabetes risk prediction in the general population.
Collapse
Affiliation(s)
- Laeya Abdoli Najmi
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ingvild Aukrust
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Janne Molnes
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Noel Burtt
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Anders Molven
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Clinical Research Center, Lund University, Malmö, Sweden
| | - David Altshuler
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
- Departments of Genetics and Medicine, Harvard Medical School, Boston, MA
- Departments of Molecular Biology and Diabetes Unit, Massachusetts General Hospital, Boston, MA
| | - Stefan Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lise Bjørkhaug
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Biomedical Laboratory Sciences, Bergen University College, Bergen, Norway
| | - Pål Rasmus Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
169
|
Giuffrida FMA, Moises RS, Weinert LS, Calliari LE, Manna TD, Dotto RP, Franco LF, Caetano LA, Teles MG, Lima RA, Alves C, Dib SA, Silveiro SP, Dias-da-Silva MR, Reis AF. Maturity-onset diabetes of the young (MODY) in Brazil: Establishment of a national registry and appraisal of available genetic and clinical data. Diabetes Res Clin Pract 2017; 123:134-142. [PMID: 28012402 DOI: 10.1016/j.diabres.2016.10.017] [Citation(s) in RCA: 14] [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: 08/30/2016] [Revised: 10/12/2016] [Accepted: 10/19/2016] [Indexed: 01/10/2023]
Abstract
AIMS Maturity-Onset Diabetes of the Young (MODY) comprises a heterogeneous group of monogenic forms of diabetes caused by mutations in at least 14 genes, but mostly by mutations in Glucokinase (GCK) and hepatocyte nuclear factor-1 homeobox A (HNF1A). This study aims to establish a national registry of MODY cases in Brazilian patients, assessing published and unpublished data. METHODS 311 patients with clinical characteristics of MODY were analyzed, with unpublished data on 298 individuals described in 12 previous publications and 13 newly described cases in this report. RESULTS 72 individuals had GCK mutations, 9 described in Brazilian individuals for the first time. One previously unpublished novel GCK mutation, Gly178Ala, was found in one family. 31 individuals had HNF1A mutations, 2 described for the first time in Brazilian individuals. Comparisons of GCK probands vs HNF1A: age 16±11 vs 35±20years; age at diagnosis 11±8 vs 21±7years; BMI 19±6 vs 25±6kg/m2; sulfonylurea users 5 vs 83%; insulin users 5 vs 17%; presence of arterial hypertension 0 vs. 33%, all p<0.05. No differences were observed in lipids and C-peptide. CONCLUSIONS Most MODY cases in Brazil are due to GCK mutations. In agreement with other studied populations, novel mutations are common. Only 14% of patients with familial diabetes carry a HNF1A mutation. Diagnosis of other rare forms of MODY is still a challenge in Brazilian population, as well as adequate strategies to screen individuals for molecular diagnosis.
Collapse
Affiliation(s)
- Fernando M A Giuffrida
- Universidade do Estado da Bahia (UNEB), Salvador, Brazil; Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| | - Regina S Moises
- Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Leticia S Weinert
- Endocrinology Unit - Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luis E Calliari
- Faculdade de Medicina da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Thais Della Manna
- Instituto da Criança, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Renata P Dotto
- Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Luciana F Franco
- Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Lilian A Caetano
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Diabetes Unit, University of Sao Paulo (USP) Medical School, Sao Paulo, Brazil
| | - Milena G Teles
- Monogenic Diabetes Group, Genetic Endocrinology Unit and Diabetes Unit, University of Sao Paulo (USP) Medical School, Sao Paulo, Brazil
| | - Renata Andrade Lima
- Pediatric Endocrinology Unit, University Hospital Prof. Edgard Santos, Faculty of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Crésio Alves
- Pediatric Endocrinology Unit, University Hospital Prof. Edgard Santos, Faculty of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Sergio A Dib
- Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Sandra P Silveiro
- Endocrinology Unit - Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Magnus R Dias-da-Silva
- Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Andre F Reis
- Disciplina de Endocrinologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
170
|
Kleinberger JW, Maloney KA, Pollin TI. The Genetic Architecture of Diabetes in Pregnancy: Implications for Clinical Practice. Am J Perinatol 2016; 33:1319-1326. [PMID: 27571483 PMCID: PMC5507691 DOI: 10.1055/s-0036-1592078] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The genetic architecture of diabetes mellitus in general and in pregnancy is complex, owing to the multiple types of diabetes that comprise both complex/polygenic forms and monogenic (largely caused by a mutation in a single gene) forms such as maturity-onset diabetes of the young (MODY). Type 1 diabetes (T1D) and type 2 diabetes (T2D) have complex genetic etiologies, with over 40 and 90 genes/loci, respectively, implicated that interact with environmental/lifestyle factors. The genetic etiology of gestational diabetes mellitus has largely been found to overlap that of T2D. Genetic testing for complex forms of diabetes is not currently useful clinically, but genetic testing for monogenic forms, particularly MODY, has important utility for determining treatment, managing risk in family members, and pregnancy management. In particular, diagnosing MODY2, caused by GCK mutations, indicates that insulin should not be used, including during pregnancy, with the possible exception of an unaffected pregnancy during the third trimester to prevent macrosomia. A relatively simple method for identifying women with MODY2 has been piloted. MODY1, caused by HNF4A mutations, can paradoxically cause neonatal hyperinsulinemic hypoglycemia and macrosomia, indicating that detecting these cases is also clinically important. Diagnosing all MODY types provides opportunities for diagnosing other family members.
Collapse
Affiliation(s)
| | | | - Toni I. Pollin
- To Whom Correspondence May Be Addressed: Toni I. Pollin, MS, PhD, 660 West Redwood Street, Room 445C, Baltimore, MD 21201, 410-706-1630,
| |
Collapse
|
171
|
Timsit J, Saint-Martin C, Dubois-Laforgue D, Bellanné-Chantelot C. Searching for Maturity-Onset Diabetes of the Young (MODY): When and What for? Can J Diabetes 2016; 40:455-461. [DOI: 10.1016/j.jcjd.2015.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/02/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022]
|
172
|
Freudenthal B, Logan J, Croucher PI, Williams GR, Bassett JHD. Rapid phenotyping of knockout mice to identify genetic determinants of bone strength. J Endocrinol 2016; 231:R31-46. [PMID: 27535945 PMCID: PMC5064764 DOI: 10.1530/joe-16-0258] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/17/2016] [Indexed: 12/27/2022]
Abstract
The genetic determinants of osteoporosis remain poorly understood, and there is a large unmet need for new treatments in our ageing society. Thus, new approaches for gene discovery in skeletal disease are required to complement the current genome-wide association studies in human populations. The International Knockout Mouse Consortium (IKMC) and the International Mouse Phenotyping Consortium (IMPC) provide such an opportunity. The IKMC generates knockout mice representing each of the known protein-coding genes in C57BL/6 mice and, as part of the IMPC initiative, the Origins of Bone and Cartilage Disease project identifies mutants with significant outlier skeletal phenotypes. This initiative will add value to data from large human cohorts and provide a new understanding of bone and cartilage pathophysiology, ultimately leading to the identification of novel drug targets for the treatment of skeletal disease.
Collapse
Affiliation(s)
- Bernard Freudenthal
- Molecular Endocrinology LaboratoryDepartment of Medicine, Imperial College London, London, UK
| | - John Logan
- Molecular Endocrinology LaboratoryDepartment of Medicine, Imperial College London, London, UK
| | - Peter I Croucher
- Garvan Institute of Medical ResearchSydney, New South Wales, Australia
| | - Graham R Williams
- Molecular Endocrinology LaboratoryDepartment of Medicine, Imperial College London, London, UK
| | - J H Duncan Bassett
- Molecular Endocrinology LaboratoryDepartment of Medicine, Imperial College London, London, UK
| |
Collapse
|
173
|
Abstract
Maturity Onset Diabetes of the Young type 3 (MODY3), linked to mutations in the transcription factor HNF1A, is the most prevalent form of monogenic diabetes mellitus. HNF1alpha-deficiency leads to defective insulin secretion via a molecular mechanism that is still not completely understood. Moreover, in MODY3 patients the severity of insulin secretion can be extremely variable even in the same kindred, indicating that modifier genes may control the onset of the disease. With the use of a mouse model for HNF1alpha-deficiency, we show here that specific genetic backgrounds (C3H and CBA) carry a powerful genetic suppressor of diabetes. A genome scan analysis led to the identification of a major suppressor locus on chromosome 3 (Moda1). Moda1 locus contains 11 genes with non-synonymous SNPs that significantly interacts with other loci on chromosomes 4, 11 and 18. Mechanistically, the absence of HNF1alpha in diabetic-prone (sensitive) strains leads to postnatal defective islets growth that is remarkably restored in resistant strains. Our findings are relevant to human genetics since Moda1 is syntenic with a human locus identified by genome wide association studies of fasting glycemia in patients. Most importantly, our results show that a single genetic locus can completely suppress diabetes in Hnf1a-deficiency.
Collapse
|
174
|
Zheng B, Chen L, Gonzalez FJ. ISN Forefronts Symposium 2015: Nuclear Receptors and Diabetic Nephropathy. Kidney Int Rep 2016; 1:177-188. [PMID: 28932823 PMCID: PMC5601313 DOI: 10.1016/j.ekir.2016.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/22/2016] [Accepted: 07/27/2016] [Indexed: 01/19/2023] Open
Abstract
Diabetic nephropathy (DN) is the major reason for end stage renal disease in the western world. Patients with DN developed more severe cardiovascular complications with worse prognosis. In spite of tight blood pressure and glucose control through applying angiotensin II receptor antagonism, angiotensin receptor inhibitors and even direct renin inhibitors, the progression and development of DN has continued to accelerate. Nuclear receptors are, with few exceptions, ligand-depended transcription factors some of which modulate genes involved in the transportation and metabolism of carbohydrate or lipid, and inflammation. Considering the diverse biological functions of nuclear receptors, efforts have been made to explore their contributions to the pathogenesis of DN and potential therapeutic strategies. This review is mainly focused on the association between various nuclear receptors and the pathogenesis of DN, the potential beneficial effects of targeting these receptors for preventing the progress of DN, and the important role that nuclear receptors may play in future therapeutic strategies for DN.
Collapse
Affiliation(s)
- Bo Zheng
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
- National Center for Liver Cancer, Shanghai, China
| | - Lei Chen
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China
- National Center for Liver Cancer, Shanghai, China
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
175
|
Moritani M, Yokota I, Horikawa R, Urakami T, Nishii A, Kawamura T, Kikuchi N, Kikuchi T, Ogata T, Sugihara S, Amemiya S. Identification of monogenic gene mutations in Japanese subjects diagnosed with type 1B diabetes between >5 and 15.1 years of age. J Pediatr Endocrinol Metab 2016; 29:1047-54. [PMID: 27398945 DOI: 10.1515/jpem-2016-0030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Monogenic mutations, such as those in the potassium inwardly-rectifying channel, subfamily J, member 11 (KCNJ11) and insulin (INS) genes, are identified in young patients with type 1B diabetes (non-autoimmune-mediated). We recently reported the results of a test for monogenic forms of diabetes in Japanese children who were diagnosed with type 1B diabetes at <5 years of age. In this study, we tested for monogenic forms of diabetes in Japanese children aged >5 to ≤15.1 years at the diagnosis of type 1B diabetes. METHODS Thirty-two Japanese children (eight males, 24 females) with type 1 diabetes negative for glutamate decarboxylase (GAD) 65 and/or IA-2A autoantibodies and who were aged >5 to 15.1 years at diagnosis were recruited from 16 independent hospitals participating in the Japanese Study Group of Insulin Therapy for Childhood and Adolescent Diabetes (JSGIT). We performed mutational analyses of genes with a high frequency of mutation [INS, KCNJ11, hepatocyte nuclear factor 1 alpha (HNF1α) and hepatocyte nuclear factor 4 alpha (HNF4α)]. RESULTS We identified one missense mutation (G32S) in the INS gene and two mutations (R131Q and R203S) in the HNF1α gene that could be associated with diabetes. No missense change was found in the KCNJ11 gene. CONCLUSIONS Our results suggest that although mutations in the INS gene can be detected in Japanese patients aged >5 years at diagnosis, the frequency of mutations decrease in older age groups. Conversely, the frequency of the mutation in the HNF1α gene increased in patients diagnosed at age 5 or older. Clinicians should consider the possibility of maturity onset diabetes of the young (MODY) in children diagnosed with type 1B diabetes.
Collapse
|
176
|
Javorhazy A, Farkas N, Beothe T, Pusztai C, Szanto A, Kovacs G. Lack of TMEM27 expression is associated with postoperative progression of clinically localized conventional renal cell carcinoma. J Cancer Res Clin Oncol 2016; 142:1947-53. [PMID: 27417314 DOI: 10.1007/s00432-016-2207-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/08/2016] [Indexed: 11/25/2022]
Abstract
PURPOSE In spite of early detection of conventional renal cell carcinoma (RCC) by widespread use of abdominal imaging, approximately 10-15 % of patients will die due to disease. The aim of this study was to identify new biomarkers predicting the postoperative progression of conventional RCC. METHODS Tissue multiarrays (TMA) of conventional RCC from a cohort of 486 patients were analysed by immunohistochemistry for expression of the transmembrane protein TMEM27, which was identified as a candidate biomarker by Affymetrix U133 Plus 2.0 array. Univariate and multivariate Cox regression models were addressed to assess cancer-specific survival in association with clinicopathological variables and TMEM27 expression. Cancer-specific survival time was estimated with Kaplan-Meier analysis, and the comparison of survival curves was made with the log-rank test. RESULTS The Kaplan-Meier survival analysis indicated a poor disease-specific survival rates for tumours without TMEM27 staining. Univariate analysis revealed an association of patient survival with T stadium, grade, stage and size of tumour and TMEM27 expression in all cases as well as in the cohort of patients with postoperative tumour progression. In multivariate analysis, only T stadium and TMEM27 staining showed a significant association with postoperative cancer-specific death (p < 0.001). CONCLUSIONS Lack of expression of the TMEM27 in conventional RCC defines a group of patients at high risk for cancer-related death.
Collapse
Affiliation(s)
- Andras Javorhazy
- Department of Urology, Medical School, University of Pecs, Munkacsy M. u. 2, Pecs, 7621, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pecs, Pecs, Hungary
| | - Tamas Beothe
- Department of Urology, Medical School, University of Pecs, Munkacsy M. u. 2, Pecs, 7621, Hungary
| | - Csaba Pusztai
- Department of Urology, Medical School, University of Pecs, Munkacsy M. u. 2, Pecs, 7621, Hungary
| | - Arpad Szanto
- Department of Urology, Medical School, University of Pecs, Munkacsy M. u. 2, Pecs, 7621, Hungary
| | - Gyula Kovacs
- Department of Urology, Medical School, University of Pecs, Munkacsy M. u. 2, Pecs, 7621, Hungary.
| |
Collapse
|
177
|
Docena MK, Faiman C, Stanley CM, Pantalone KM. Mody-3: novel HNF1A mutation and the utility of glucagon-like peptide (GLP)-1 receptor agonist therapy. Endocr Pract 2016; 20:107-11. [PMID: 24014008 DOI: 10.4158/ep13254.or] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE An estimated 1 to 2% of cases of diabetes mellitus have a monogenic basis; however, delayed diagnosis and misdiagnosis as type 1 and 2 diabetes are common. Correctly identifying the molecular basis of an individual's diabetes may significantly alter the management approach to both the patient and his or her relatives. We describe a case of mature onset diabetes of the young (MODY) with sufficient evidence to support the classification of a novel HNF1A (hepatocyte nuclear factor-1-α) mutation as a cause of MODY-3. METHODS A 21-year-old Caucasian female presented to our office with a diagnosis of noninsulin-dependent diabetes mellitus (NIDDM) at age 10; glycemia was initially managed with oral antidiabetic (OAD) agents and insulin detemir. The patient reported a strong family history of early-onset NIDDM in both her mother and maternal grandmother, both of whom eventually required insulin therapy to control glycemia. The patient's medical and family history were highly suggestive of maturity-onset diabetes of the young (MODY), and genetic testing was performed. RESULTS Genetic screening detected a mutation p. Arg200Trp in the HNF1A gene in the patient, her mother, and maternal grandmother, suggesting a diagnosis of MODY-3. This finding resulted in a change of antidiabetic therapy in all 3 patients, including the addition of once-daily liraglutide therapy, which helped improve their glycemic control. CONCLUSION Our case report supports the classification of the p. Arg200Trp mutation as a cause of MODY-3. The findings also suggest that glucagon-like peptide-1 (GLP-1) receptor agonist therapy may be of value in managing glycemia in patients with MODY-3.
Collapse
Affiliation(s)
| | - Charles Faiman
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Kevin M Pantalone
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
178
|
Beer NL, Gloyn AL. Genome-edited human stem cell-derived beta cells: a powerful tool for drilling down on type 2 diabetes GWAS biology. F1000Res 2016; 5:F1000 Faculty Rev-1711. [PMID: 27508066 PMCID: PMC4955023 DOI: 10.12688/f1000research.8682.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2016] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes (T2D) is a disease of pandemic proportions, one defined by a complex aetiological mix of genetic, epigenetic, environmental, and lifestyle risk factors. Whilst the last decade of T2D genetic research has identified more than 100 loci showing strong statistical association with disease susceptibility, our inability to capitalise upon these signals reflects, in part, a lack of appropriate human cell models for study. This review discusses the impact of two complementary, state-of-the-art technologies on T2D genetic research: the generation of stem cell-derived, endocrine pancreas-lineage cells and the editing of their genomes. Such models facilitate investigation of diabetes-associated genomic perturbations in a physiologically representative cell context and allow the role of both developmental and adult islet dysfunction in T2D pathogenesis to be investigated. Accordingly, we interrogate the role that patient-derived induced pluripotent stem cell models are playing in understanding cellular dysfunction in monogenic diabetes, and how site-specific nucleases such as the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system are helping to confirm genes crucial to human endocrine pancreas development. We also highlight the novel biology gleaned in the absence of patient lines, including an ability to model the whole phenotypic spectrum of diabetes phenotypes occurring both in utero and in adult cells, interrogating the non-coding 'islet regulome' for disease-causing perturbations, and understanding the role of other islet cell types in aberrant glycaemia. This article aims to reinforce the importance of investigating T2D signals in cell models reflecting appropriate species, genomic context, developmental time point, and tissue type.
Collapse
Affiliation(s)
- Nicola L. Beer
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Oxford, UK,
| | - Anna L. Gloyn
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Oxford, UK,Wellcome Trust Centre for Human Genetics, Oxford, UK,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
179
|
Lerner J, Bagattin A, Verdeguer F, Makinistoglu MP, Garbay S, Felix T, Heidet L, Pontoglio M. Human mutations affect the epigenetic/bookmarking function of HNF1B. Nucleic Acids Res 2016; 44:8097-111. [PMID: 27229139 PMCID: PMC5041451 DOI: 10.1093/nar/gkw467] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/13/2016] [Indexed: 12/19/2022] Open
Abstract
Bookmarking factors are transcriptional regulators involved in the mitotic transmission of epigenetic information via their ability to remain associated with mitotic chromatin. The mechanisms through which bookmarking factors bind to mitotic chromatin remain poorly understood. HNF1β is a bookmarking transcription factor that is frequently mutated in patients suffering from renal multicystic dysplasia and diabetes. Here, we show that HNF1β bookmarking activity is impaired by naturally occurring mutations found in patients. Interestingly, this defect in HNF1β mitotic chromatin association is rescued by an abrupt decrease in temperature. The rapid relocalization to mitotic chromatin is reversible and driven by a specific switch in DNA-binding ability of HNF1β mutants. Furthermore, we demonstrate that importin-β is involved in the maintenance of the mitotic retention of HNF1β, suggesting a functional link between the nuclear import system and the mitotic localization/translocation of bookmarking factors. Altogether, our studies have disclosed novel aspects on the mechanisms and the genetic programs that account for the mitotic association of HNF1β, a bookmarking factor that plays crucial roles in the epigenetic transmission of information through the cell cycle.
Collapse
Affiliation(s)
- Jonathan Lerner
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| | - Alessia Bagattin
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| | - Francisco Verdeguer
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| | - Munevver P Makinistoglu
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| | - Serge Garbay
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| | - Tristan Felix
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| | - Laurence Heidet
- Department of Pediatric Nephrology, Assistance Publique des Hôpitaux de Paris, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hospital Necker-Enfants Malades, Paris 75015, France
| | - Marco Pontoglio
- Department of Development, Reproduction and Cancer, Institut Cochin, INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Paris 75014, France
| |
Collapse
|
180
|
Yang J, Jiang F, Guo H, Soniya T, Yan CX, Tian ZF, Shi BY. Studies of genetic variability of the hepatocyte nuclear factor-1α gene in an Indian maturity-onset diabetes of the young family. Cell Biosci 2016; 6:29. [PMID: 27148439 PMCID: PMC4855895 DOI: 10.1186/s13578-016-0095-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/19/2016] [Indexed: 11/28/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY), one of the specific types of diabetes mellitus, is a monogenetic disorder characterized by an autosomal dominant (AD) inheritance and β-cell dysfunction. To study an Indian family with clinical diagnosis of MODY and detect the genetic mutations in the aspect of molecular mechanism, seven blood samples were obtained from the diabetic patients of this pedigree and genomic DNA was extracted from peripheral leukocytes. The exon1, exon2 and exon4 of hepatocyte nuclear factor-1α (HNF-1α) gene were amplified by polymerase chain reaction. Then the products were sequenced and compared with standard sequences on gene bank. As a result, two mutations were detected in exon1. That was CTC → CTG (Leu → Leu) in codon17 and ATC → CTC (Ile → Leu) in codon27. I27L was speculated to have a close relationship with the glycometabolism and the pathogenesis of diabetes mellitus together with the putative novel mutation existed in this Indian pedigree. Meanwhile, one mutation of GGG → GGC (Gly → Gly) in codon288 of exon4 was detected in the proband. No mutations were found in exon2 but a G → T base substitution in the intron4 region among all seven samples was detected. It may have some potential effects on the onset of diabetes in this family, but we do not have any evidence right now. Although it requires further investigation on the function of mutations found in the intron region, our research may provide some clue for this issue and it deserves more attention.
Collapse
Affiliation(s)
- Jing Yang
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Feng Jiang
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Hui Guo
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Thadimacca Soniya
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Chun-xia Yan
- />Department of Forensic Medicine, Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Zhu-fang Tian
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Bing-yin Shi
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| |
Collapse
|
181
|
Epidemiology and Inherited Predisposition for Sporadic Pancreatic Adenocarcinoma. Hematol Oncol Clin North Am 2016; 29:619-40. [PMID: 26226901 DOI: 10.1016/j.hoc.2015.04.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Given the changing demographics of Western populations, the numbers of pancreatic cancer cases are projected to increase during the next decade. Diabetes, recent cigarette smoking, and excess body weight are the cancer's most consistent risk factors. The search for common and rare germline variants that influence risk of pancreatic cancer through genome-wide association studies and high-throughput-sequencing-based studies is underway and holds the promise of increasing the knowledge of variants and genes that play a role in inherited susceptibility of this disease. Research reported in this review has advanced the understanding of pancreatic cancer.
Collapse
|
182
|
Bockenhauer D, Jaureguiberry G. HNF1B-associated clinical phenotypes: the kidney and beyond. Pediatr Nephrol 2016; 31:707-14. [PMID: 26160100 DOI: 10.1007/s00467-015-3142-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 05/19/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
Abstract
Mutations in HNF1B, the gene encoding hepatocyte nuclear factor 1β are the most commonly identified genetic cause of renal malformations. HNF1B was first identified as a disease gene for diabetes (MODY5) in 1997, and its involvement in renal disease was subsequently noted through clinical observations in pedigrees affected by MODY5. Since then, a whole spectrum of associated phenotypes have been reported, including genital malformations, autism, epilepsy, gout, hypomagnesaemia, primary hyperparathyroidism, liver and intestinal abnormalities and a rare form of kidney cancer. The most commonly identified mutation, in approximately 50 % of patients, is an entire gene deletion occurring in the context of a 17q12 chromosomal microdeletion that also includes several other genes. Some of the associated phenotypes, especially the neurologic ones, appear to occur only in the context of this microdeletion and thus may not be directly linked to HNF1B. Here we review the spectrum of associated phenotypes and discuss potential implications for clinical management.
Collapse
Affiliation(s)
- Detlef Bockenhauer
- UCL Institute of Child Health, 30 Guilford Street, London, WC1N 3EH, UK. .,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| | | |
Collapse
|
183
|
Sujjitjoon J, Kooptiwut S, Chongjaroen N, Tangjittipokin W, Plengvidhya N, Yenchitsomanus PT. Aberrant mRNA splicing of paired box 4 (PAX4) IVS7-1G>A mutation causing maturity-onset diabetes of the young, type 9. Acta Diabetol 2016; 53:205-16. [PMID: 25951767 DOI: 10.1007/s00592-015-0760-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 04/12/2015] [Indexed: 01/24/2023]
Abstract
AIMS Paired box 4 (PAX4) mutations cause maturity-onset diabetes of the young, type 9 (MODY9). The molecular defect and alteration of PAX4 function associated with the mutation PAX4 IVS7-1G>A in a family with MODY9 and severe diabetic complications were studied. METHODS We investigated the functional consequences of PAX4 IVS7-1G>A on mRNA splicing using minigene assays. Wild-type and mutant PAX4 were expressed in mouse pancreatic β- and α-cell lines, and protein levels and translocation of PAX4 into the nucleus were determined. We also examined transcriptional repression of PAX4 target-gene promoters and β-cell viability under diabetic-like (high-glucose) conditions. RESULTS PAX4 IVS7-1G>A disrupts an acceptor splice site, causing an adjacent cryptic splice site within exon 8 to be used, resulting in a three-nucleotide deletion and glutamine deletion at position 250 (p.Q250del). Wild-type and PAX4 Q250del proteins were expressed at similar levels and could translocate normally into the nucleus in βTC3 and αTC1.9 cells. However, the repressor functions of PAX4 Q250del on human insulin and glucagon promoters in INS-1 832/13 and αTC1.9 cells were significantly decreased, compared with that of wild-type PAX4. Moreover, the rate of apoptosis was increased in INS-1 cells over-expressing PAX4 Q250del when cultured in high-glucose conditions. CONCLUSIONS PAX4 IVS7-1G>A caused aberrant mRNA splicing and PAX4 Q250 deletion. The mutation impaired PAX4 repressor functions on target-gene promoters and increased susceptibility to apoptosis upon high glucose exposure. Thus, PAX4 IVS7-1G>A contributes to the pathogenesis of diabetes in this MODY9 family through β-cell dysfunction.
Collapse
Affiliation(s)
- Jatuporn Sujjitjoon
- Division of Molecular Medicine, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Nalinee Chongjaroen
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Watip Tangjittipokin
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Nattachet Plengvidhya
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
184
|
Balamurugan K, Bjørkhaug L, Mahajan S, Kanthimathi S, Njølstad PR, Srinivasan N, Mohan V, Radha V. Structure-function studies of HNF1A (MODY3) gene mutations in South Indian patients with monogenic diabetes. Clin Genet 2016; 90:486-495. [PMID: 26853433 DOI: 10.1111/cge.12757] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 12/30/2022]
Abstract
Maturity-onset diabetes of the young (MODY) is a genetically heterogeneous monogenic form of diabetes characterized by onset of diabetes below 25 years of age, autosomal dominant mode of inheritance and primary defect in insulin secretion. Mutations in the gene (HNF1A) encoding transcription factor hepatocyte nuclear factor 1A (HNF-1A) results in one of the most common forms of MODY (MODY3). HNF-1A is mainly enriched in pancreatic β-cells and hepatocytes and important for organ development and normal pancreatic function. We here report on the functional interrogation of eight missense HNF1A mutations associated with MODY3 in South Indian subjects, and the contributing effect of common variant (S487N) within HNF1A. Of the eight mutations, three mutations (p.R171G, p.G245R and p.R263H), in particular, affected HNF-1A function in transfected HeLa cells by reducing both transcriptional activity and nuclear localization, possibly due to disruption of the integrity of the three dimensional structure. The common variant p.S487N contributed further to the loss-of-function of p.R271Q (p.R271Q+p.S487N double mutant), in vitro, on both activity and localization. Our data on the first functional study of HNF1A mutations in South India subjects confers that the defect of the HNF-1A mutant proteins are responsible for MODY3 diabetes in these patients.
Collapse
Affiliation(s)
- K Balamurugan
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - L Bjørkhaug
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - S Mahajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - S Kanthimathi
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - P R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - N Srinivasan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - V Mohan
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - V Radha
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| |
Collapse
|
185
|
Yang Y, Zhou TC, Liu YY, Li X, Wang WX, Irwin DM, Zhang YP. Identification of HNF4A Mutation p.T130I and HNF1A Mutations p.I27L and p.S487N in a Han Chinese Family with Early-Onset Maternally Inherited Type 2 Diabetes. J Diabetes Res 2016; 2016:3582616. [PMID: 26981542 PMCID: PMC4766352 DOI: 10.1155/2016/3582616] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/17/2015] [Accepted: 01/14/2016] [Indexed: 12/03/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY) is characterized by the onset of diabetes before the age of 25 years, positive family history, high genetic predisposition, monogenic mutations, and an autosomal dominant mode of inheritance. Here, we aimed to investigate the mutations and to characterize the phenotypes of a Han Chinese family with early-onset maternally inherited type 2 diabetes. Detailed clinical assessments and genetic screening for mutations in the HNF4α, GCK, HNF-1α, IPF-1, HNF1β, and NEUROD1 genes were carried out in this family. One HNF4A mutation (p.T130I) and two HNF1A polymorphisms (p.I27L and p.S487N) were identified. Mutation p.T130I was associated with both early-onset and late-onset diabetes and caused downregulated HNF4A expression, whereas HNF1A polymorphisms p.I27L and p.S487N were associated with the age of diagnosis of diabetes. We demonstrated that mutation p.T130I in HNF4A was pathogenic as were the predicted polymorphisms p.I27L and p.S487N in HNF1A by genetic and functional analysis. Our results show that mutations in HNF4A and HNF1A genes might account for this early-onset inherited type 2 diabetes.
Collapse
Affiliation(s)
- Ying Yang
- Department of Endocrinology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan 650021, China
- Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan 650091, China
| | - Tai-Cheng Zhou
- Department of Endocrinology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan 650021, China
- Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan 650091, China
- The Central Laboratory of the Second People's Hospital of Yunnan Province, Kunming, Yunnan 650021, China
| | - Yong-Ying Liu
- Department of Endocrinology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan 650021, China
| | - Xiao Li
- The Central Laboratory of the Second People's Hospital of Yunnan Province, Kunming, Yunnan 650021, China
| | - Wen-Xue Wang
- Laboratory of Biochemistry and Molecular Biology, Yunnan University, Kunming, Yunnan 650091, China
| | - David M. Irwin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada M5S 2E8
| | - Ya-Ping Zhang
- Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan 650091, China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| |
Collapse
|
186
|
Althari S, Gloyn AL. When is it MODY? Challenges in the Interpretation of Sequence Variants in MODY Genes. Rev Diabet Stud 2016; 12:330-48. [PMID: 27111119 DOI: 10.1900/rds.2015.12.330] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The genomics revolution has raised more questions than it has provided answers. Big data from large population-scale resequencing studies are increasingly deconstructing classic notions of Mendelian disease genetics, which support a simplistic correlation between mutational severity and phenotypic outcome. The boundaries are being blurred as the body of evidence showing monogenic disease-causing alleles in healthy genomes, and in the genomes of individu-als with increased common complex disease risk, continues to grow. In this review, we focus on the newly emerging challenges which pertain to the interpretation of sequence variants in genes implicated in the pathogenesis of maturity-onset diabetes of the young (MODY), a presumed mono-genic form of diabetes characterized by Mendelian inheritance. These challenges highlight the complexities surrounding the assignments of pathogenicity, in particular to rare protein-alerting variants, and bring to the forefront some profound clinical diagnostic implications. As MODY is both genetically and clinically heterogeneous, an accurate molecular diagnosis and cautious extrapolation of sequence data are critical to effective disease management and treatment. The biological and translational value of sequence information can only be attained by adopting a multitude of confirmatory analyses, which interrogate variant implication in disease from every possible angle. Indeed, studies which have effectively detected rare damaging variants in known MODY genes in normoglycemic individuals question the existence of a sin-gle gene mutation scenario: does monogenic diabetes exist when the genetic culprits of MODY have been systematical-ly identified in individuals without MODY?
Collapse
Affiliation(s)
- Sara Althari
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, UK
| |
Collapse
|
187
|
Hepatocyte nuclear factor 1 coordinates multiple processes in a model of intestinal epithelial cell function. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:591-8. [PMID: 26855178 DOI: 10.1016/j.bbagrm.2016.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 12/26/2022]
Abstract
Mutations in hepatocyte nuclear factor 1 transcription factors (HNF1α/β) are associated with diabetes. These factors are well studied in the liver, pancreas and kidney, where they direct tissue-specific gene regulation. However, they also have an important role in the biology of many other tissues, including the intestine. We investigated the transcriptional network governed by HNF1 in an intestinal epithelial cell line (Caco2). We used chromatin immunoprecipitation followed by direct sequencing (ChIP-seq) to identify HNF1 binding sites genome-wide. Direct targets of HNF1 were validated using conventional ChIP assays and confirmed by siRNA-mediated depletion of HNF1, followed by RT-qPCR. Gene ontology process enrichment analysis of the HNF1 targets identified multiple processes with a role in intestinal epithelial cell function, including properties of the cell membrane, cellular response to hormones, and regulation of biosynthetic processes. Approximately 50% of HNF1 binding sites were also occupied by other members of the intestinal transcriptional network, including hepatocyte nuclear factor 4A (HNF4A), caudal type homeobox 2 (CDX2), and forkhead box A2 (FOXA2). Depletion of HNF1 in Caco2 cells increases FOXA2 abundance and decreases levels of CDX2, illustrating the coordinated activities of the network. These data suggest that HNF1 plays an important role in regulating intestinal epithelial cell function, both directly and through interactions with other intestinal transcription factors.
Collapse
|
188
|
A candidate-gene association study of topiramate-induced weight loss in obese patients with and without type 2 diabetes mellitus. Pharmacogenet Genomics 2016; 26:53-65. [DOI: 10.1097/fpc.0000000000000185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
189
|
Abstract
Although relatively rare, pancreatic tumors are highly lethal [1]. In the United States, an estimated 48,960 individuals will be diagnosed with pancreatic cancer and 40,560 will die from this disease in 2015 [1]. Globally, 337,872 new pancreatic cancer cases and 330,391 deaths were estimated in 2012 [2]. In contrast to most other cancers, mortality rates for pancreatic cancer are not improving; in the US, it is predicted to become the second leading cause of cancer related deaths by 2030 [3, 4]. The vast majority of tumors arise in the exocrine pancreas, with pancreatic ductal adenocarcinoma (PDAC) accounting for approximately 95% of tumors. Tumors arising in the endocrine pancreas (pancreatic neuroendocrine tumors) represent less than 5% of all pancreatic tumors [5]. Smoking, type 2 diabetes mellitus (T2D), obesity and pancreatitis are the most consistent epidemiological risk factors for pancreatic cancer [5]. Family history is also a risk factor for developing pancreatic cancer with odds ratios (OR) ranging from 1.7-2.3 for first-degree relatives in most studies, indicating that shared genetic factors may play a role in the etiology of this disease [6-9]. This review summarizes the current knowledge of germline pancreatic cancer risk variants with a special emphasis on common susceptibility alleles identified through Genome Wide Association Studies (GWAS).
Collapse
Affiliation(s)
- Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
190
|
Kofent J, Spagnoli FM. Xenopus as a model system for studying pancreatic development and diabetes. Semin Cell Dev Biol 2016; 51:106-16. [PMID: 26806634 DOI: 10.1016/j.semcdb.2016.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/06/2016] [Indexed: 02/07/2023]
Abstract
Diabetes is a chronic disease caused by the loss or dysfunction of the insulin-producing β-cells in the pancreas. To date, much of our knowledge about β-cells in humans comes from studying rare monogenic forms of diabetes. Importantly, the majority of mutations so far associated to monogenic diabetes are in genes that exert a regulatory role in pancreatic development and/or β-cell function. Thus, the identification and study of novel mutations open an unprecedented window into human pancreatic development. In this review, we summarize major advances in the genetic dissection of different types of monogenic diabetes and the insights gained from a developmental perspective. We highlight future challenges to bridge the gap between the fast accumulation of genetic data through next-generation sequencing and the need of functional insights into disease mechanisms. Lastly, we discuss the relevance and advantages of studying candidate gene variants in vivo using the Xenopus as model system.
Collapse
Affiliation(s)
- Julia Kofent
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany
| | - Francesca M Spagnoli
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany.
| |
Collapse
|
191
|
Kim YI, Lee J, Choi YJ, Seo J, Park J, Lee SY, Cho JY. Proteogenomic Study beyond Chromosome 9: New Insight into Expressed Variant Proteome and Transcriptome in Human Lung Adenocarcinoma Tissues. J Proteome Res 2015; 14:5007-5016. [PMID: 26584007 DOI: 10.1021/acs.jproteome.5b00544] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This is a report of a human proteome project (HPP) related to chromosome 9 (Chr 9). To reveal missing proteins and undiscovered features in proteogenomes, both LC-MS/MS analysis and next-generation RNA sequencing (RNA-seq)-based identification and characterization were conducted on five pairs of lung adenocarcinoma tumors and adjacent nontumor tissues. Before our previous Chromosome-Centric Human Proteome Project (C-HPP) special issue, there were 170 remaining missing proteins on Chr 9 (neXtProt 2013.09.26 rel.); 133 remain at present (neXtProt 2015.04.28 rel.). In the proteomics study, we found two missing protein candidates that require follow-up work and one unrevealed protein across all chromosomes. RNA-seq analysis detected RNA expression for four nonsynonymous (NS) single nucleotide polymorphisms (SNPs) (in CDH17, HIST1H1T, SAPCD2, and ZNF695) and three synonymous SNPs (in CDH17, CST1, and HNF1A) in all five tumor tissues but not in any of the adjacent normal tissues. By constructing a cancer patient sample-specific protein database based on individual RNA-seq data and by searching the proteomics data from the same sample, we identified four missense mutations in four genes (LTF, HDLBP, TF, and HBD). Two of these mutations were found in tumor samples but not in paired normal tissues. In summary, our proteogenomic study of human primary lung tumor tissues detected additional and revealed novel missense mutations and synonymous SNP signatures, some of which are specific to lung cancers. Data from mass spectrometry have been deposited in the ProteomeXchange with the identifier PXD002523.
Collapse
Affiliation(s)
- Yong-In Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University , Seoul 08826, South Korea
| | - Jongan Lee
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul 06351, South Korea
| | - Young-Jin Choi
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University , Seoul 08826, South Korea
- ProtAnBio , Seoul 08826, South Korea
| | - Jawon Seo
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University , Seoul 08826, South Korea
| | - Jisook Park
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul 06351, South Korea
| | - Soo-Youn Lee
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul 06351, South Korea
- Department of Clinical Pharmacology & Therapeutics, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul 06351, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University , Seoul 08826, South Korea
| |
Collapse
|
192
|
Abstract
Crigler-Najjar syndrome presents as severe unconjugated hyperbilirubinemia and is characteristically caused by a mutation in the UGT1A1 gene, encoding the enzyme responsible for bilirubin glucuronidation. Here we present a patient with Crigler-Najjar syndrome with a completely normal UGT1A1 coding region. Instead, a homozygous 3 nucleotide insertion in the UGT1A1 promoter was identified that interrupts the HNF1α binding site. This mutation results in almost complete abolishment of UGT1A1 promoter activity and prevents the induction of UGT1A1 expression by the liver nuclear receptors CAR and PXR, explaining the lack of a phenobarbital response in this patient. Although animal studies have revealed the importance of HNF1α for normal liver function, this case provides the first clinical proof that mutations in its binding site indeed result in severe liver pathology stressing the importance of promoter sequence analysis.
Collapse
|
193
|
The role of BH3-only protein Bmf in the pathogenesis of dominant negative hepatocyte nuclear factor-1 –induced mature-onset diabetes of the young in transgenic mice. BMC Proc 2015. [PMCID: PMC4625193 DOI: 10.1186/1753-6561-9-s7-a25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
194
|
Abstract
Maturity-onset diabetes of the young (MODY) is a monogenic form of diabetes that accounts for at least 1 % of all cases of diabetes mellitus. MODY classically presents as non-insulin-requiring diabetes in lean individuals typically younger than 25 with evidence of autosomal dominant inheritance, but these criteria do not capture all cases and can also overlap with other diabetes types. Genetic diagnosis of MODY is important for selecting the right treatment, yet ~95 % of MODY cases in the USA are misdiagnosed. MODY prevalence and characteristics have been well-studied in some populations, such as the UK and Norway, while other ethnicities, like African and Latino, need much more study. Emerging next-generation sequencing methods are making more widespread study and clinical diagnosis increasingly feasible; at the same time, they are detecting other mutations in the same genes of unknown clinical significance. This review will cover the current epidemiological studies of MODY and barriers and opportunities for moving toward a goal of access to an appropriate diagnosis for all affected individuals.
Collapse
Affiliation(s)
- Jeffrey W Kleinberger
- Division of Endocrinology, Diabetes, and Nutrition and Program in Personalized and Genomic Medicine, Department of Medicine, University of Maryland School of Medicine, 660 West Redwood Street, Room 445C, Baltimore, MD, 21201, USA.
| | - Toni I Pollin
- Division of Endocrinology, Diabetes, and Nutrition and Program in Personalized and Genomic Medicine, Department of Medicine, University of Maryland School of Medicine, 660 West Redwood Street, Room 445C, Baltimore, MD, 21201, USA.
- University of Maryland School of Medicine, 660 West Redwood Street, Room 464, Baltimore, MD, 21201, USA.
| |
Collapse
|
195
|
Gaulton KJ, Ferreira T, Lee Y, Raimondo A, Mägi R, Reschen ME, Mahajan A, Locke A, Rayner NW, Robertson N, Scott RA, Prokopenko I, Scott LJ, Green T, Sparso T, Thuillier D, Yengo L, Grallert H, Wahl S, Frånberg M, Strawbridge RJ, Kestler H, Chheda H, Eisele L, Gustafsson S, Steinthorsdottir V, Thorleifsson G, Qi L, Karssen LC, van Leeuwen EM, Willems SM, Li M, Chen H, Fuchsberger C, Kwan P, Ma C, Linderman M, Lu Y, Thomsen SK, Rundle JK, Beer NL, van de Bunt M, Chalisey A, Kang HM, Voight BF, Abecasis GR, Almgren P, Baldassarre D, Balkau B, Benediktsson R, Blüher M, Boeing H, Bonnycastle LL, Bottinger EP, Burtt NP, Carey J, Charpentier G, Chines PS, Cornelis MC, Couper DJ, Crenshaw AT, van Dam RM, Doney ASF, Dorkhan M, Edkins S, Eriksson JG, Esko T, Eury E, Fadista J, Flannick J, Fontanillas P, Fox C, Franks PW, Gertow K, Gieger C, Gigante B, Gottesman O, Grant GB, Grarup N, Groves CJ, Hassinen M, Have CT, Herder C, Holmen OL, Hreidarsson AB, Humphries SE, Hunter DJ, Jackson AU, Jonsson A, Jørgensen ME, Jørgensen T, Kao WHL, Kerrison ND, Kinnunen L, Klopp N, Kong A, Kovacs P, Kraft P, Kravic J, Langford C, et alGaulton KJ, Ferreira T, Lee Y, Raimondo A, Mägi R, Reschen ME, Mahajan A, Locke A, Rayner NW, Robertson N, Scott RA, Prokopenko I, Scott LJ, Green T, Sparso T, Thuillier D, Yengo L, Grallert H, Wahl S, Frånberg M, Strawbridge RJ, Kestler H, Chheda H, Eisele L, Gustafsson S, Steinthorsdottir V, Thorleifsson G, Qi L, Karssen LC, van Leeuwen EM, Willems SM, Li M, Chen H, Fuchsberger C, Kwan P, Ma C, Linderman M, Lu Y, Thomsen SK, Rundle JK, Beer NL, van de Bunt M, Chalisey A, Kang HM, Voight BF, Abecasis GR, Almgren P, Baldassarre D, Balkau B, Benediktsson R, Blüher M, Boeing H, Bonnycastle LL, Bottinger EP, Burtt NP, Carey J, Charpentier G, Chines PS, Cornelis MC, Couper DJ, Crenshaw AT, van Dam RM, Doney ASF, Dorkhan M, Edkins S, Eriksson JG, Esko T, Eury E, Fadista J, Flannick J, Fontanillas P, Fox C, Franks PW, Gertow K, Gieger C, Gigante B, Gottesman O, Grant GB, Grarup N, Groves CJ, Hassinen M, Have CT, Herder C, Holmen OL, Hreidarsson AB, Humphries SE, Hunter DJ, Jackson AU, Jonsson A, Jørgensen ME, Jørgensen T, Kao WHL, Kerrison ND, Kinnunen L, Klopp N, Kong A, Kovacs P, Kraft P, Kravic J, Langford C, Leander K, Liang L, Lichtner P, Lindgren CM, Lindholm E, Linneberg A, Liu CT, Lobbens S, Luan J, Lyssenko V, Männistö S, McLeod O, Meyer J, Mihailov E, Mirza G, Mühleisen TW, Müller-Nurasyid M, Navarro C, Nöthen MM, Oskolkov NN, Owen KR, Palli D, Pechlivanis S, Peltonen L, Perry JRB, Platou CGP, Roden M, Ruderfer D, Rybin D, van der Schouw YT, Sennblad B, Sigurðsson G, Stančáková A, Steinbach G, Storm P, Strauch K, Stringham HM, Sun Q, Thorand B, Tikkanen E, Tonjes A, Trakalo J, Tremoli E, Tuomi T, Wennauer R, Wiltshire S, Wood AR, Zeggini E, Dunham I, Birney E, Pasquali L, Ferrer J, Loos RJF, Dupuis J, Florez JC, Boerwinkle E, Pankow JS, van Duijn C, Sijbrands E, Meigs JB, Hu FB, Thorsteinsdottir U, Stefansson K, Lakka TA, Rauramaa R, Stumvoll M, Pedersen NL, Lind L, Keinanen-Kiukaanniemi SM, Korpi-Hyövälti E, Saaristo TE, Saltevo J, Kuusisto J, Laakso M, Metspalu A, Erbel R, Jöcke KH, Moebus S, Ripatti S, Salomaa V, Ingelsson E, Boehm BO, Bergman RN, Collins FS, Mohlke KL, Koistinen H, Tuomilehto J, Hveem K, Njølstad I, Deloukas P, Donnelly PJ, Frayling TM, Hattersley AT, de Faire U, Hamsten A, Illig T, Peters A, Cauchi S, Sladek R, Froguel P, Hansen T, Pedersen O, Morris AD, Palmer CNA, Kathiresan S, Melander O, Nilsson PM, Groop LC, Barroso I, Langenberg C, Wareham NJ, O'Callaghan CA, Gloyn AL, Altshuler D, Boehnke M, Teslovich TM, McCarthy MI, Morris AP, DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) Consortium. Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci. Nat Genet 2015; 47:1415-25. [PMID: 26551672 PMCID: PMC4666734 DOI: 10.1038/ng.3437] [Show More Authors] [Citation(s) in RCA: 310] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 10/07/2015] [Indexed: 12/15/2022]
Abstract
We performed fine mapping of 39 established type 2 diabetes (T2D) loci in 27,206 cases and 57,574 controls of European ancestry. We identified 49 distinct association signals at these loci, including five mapping in or near KCNQ1. 'Credible sets' of the variants most likely to drive each distinct signal mapped predominantly to noncoding sequence, implying that association with T2D is mediated through gene regulation. Credible set variants were enriched for overlap with FOXA2 chromatin immunoprecipitation binding sites in human islet and liver cells, including at MTNR1B, where fine mapping implicated rs10830963 as driving T2D association. We confirmed that the T2D risk allele for this SNP increases FOXA2-bound enhancer activity in islet- and liver-derived cells. We observed allele-specific differences in NEUROD1 binding in islet-derived cells, consistent with evidence that the T2D risk allele increases islet MTNR1B expression. Our study demonstrates how integration of genetic and genomic information can define molecular mechanisms through which variants underlying association signals exert their effects on disease.
Collapse
Affiliation(s)
- Kyle J Gaulton
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Yeji Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Anne Raimondo
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Michael E Reschen
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Adam Locke
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - N William Rayner
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Wellcome Trust Sanger Institute, Hinxton, UK
| | - Neil Robertson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Robert A Scott
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Inga Prokopenko
- Genomics of Common Disease, Imperial College London, London, UK
| | - Laura J Scott
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Todd Green
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Thomas Sparso
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dorothee Thuillier
- Lille Institute of Biology, European Genomics Institute of Diabetes, Lille, France
| | - Loic Yengo
- Lille Institute of Biology, European Genomics Institute of Diabetes, Lille, France
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Mattias Frånberg
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
- Department for Numerical Analysis and Computer Science, Stockholm University, Stockholm, Sweden
| | - Rona J Strawbridge
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Hans Kestler
- Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany
- Medical Systems Biology, Ulm University, Ulm, Germany
| | - Himanshu Chheda
- Finnish Institute for Molecular Medicine (FIMM), Helsinki, Finland
| | - Lewin Eisele
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, Essen, Germany
| | - Stefan Gustafsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | - Lu Qi
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Lennart C Karssen
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Sara M Willems
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Han Chen
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | | | - Phoenix Kwan
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Clement Ma
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Linderman
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yingchang Lu
- Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Soren K Thomsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Jana K Rundle
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Nicola L Beer
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Martijn van de Bunt
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Anil Chalisey
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hyun Min Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin F Voight
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gonçalo R Abecasis
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter Almgren
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Damiano Baldassarre
- Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
| | - Beverley Balkau
- INSERM Centre de Recherche Epidémiologie et Santé des Populations (CESP) U1018, Villejuif, France
- University Paris Sud 11, UMRS 1018, Villejuif, France
| | - Rafn Benediktsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Landspitali University Hospital, Reykjavik, Iceland
| | - Matthias Blüher
- Integrated Treatment and Research (IFB) Center for Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Heiner Boeing
- German Institute of Human Nutrition, Potsdam-Rehbruecke, Germany
| | - Lori L Bonnycastle
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Erwin P Bottinger
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Noël P Burtt
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Jason Carey
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | | | - Peter S Chines
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Marilyn C Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - David J Couper
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Rob M van Dam
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Alex S F Doney
- Diabetes Research Centre, Biomedical Research Institute, University of Dundee, Ninewells Hospital, Dundee, UK
- Pharmacogenomics Centre, Biomedical Research Institute, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Mozhgan Dorkhan
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Novo Nordisk Scandinavia, Malmo, Sweden
| | | | - Johan G Eriksson
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Unit of General Practice, Helsinki University General Hospital, Helsinki, Finland
- Folkhalsan Research Center, Helsinki, Finland
| | - Tonu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Division of Endocrinology, Children's Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Elodie Eury
- CNRS UMR 8199, Institute of Biology and Lille 2 University, Pasteur Institute, Lille, France
| | - João Fadista
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Jason Flannick
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | | | - Caroline Fox
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, USA
- Division of Endocrinology and Metabolism, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Paul W Franks
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
- Department of Clinical Sciences, Lund University, Malmo, Sweden
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| | - Karl Gertow
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Bruna Gigante
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Omri Gottesman
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - George B Grant
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christopher J Groves
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Christian T Have
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Dusseldorf, Dusseldorf, Germany
- German Center for Diabetes Research, partner site Dusseldorf, Dusseldorf, Germany
| | - Oddgeir L Holmen
- Nord-Trøndelag Health Study (HUNT) Research Center, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway
| | | | - Steve E Humphries
- Cardiovascular Genetics, British Heart Foundation (BHF) Laboratories, Institute of Cardiovascular Sciences, University College London, London, UK
| | - David J Hunter
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Anne U Jackson
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Anna Jonsson
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | | | - Torben Jørgensen
- Research Centre for Prevention and Health, Capital Region of Denmark, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg, Aalborg, Denmark
| | - Wen-Hong L Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Nicola D Kerrison
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Leena Kinnunen
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Norman Klopp
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | | | - Peter Kovacs
- Integrated Treatment and Research (IFB) Center for Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Jasmina Kravic
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | | | - Karin Leander
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Liming Liang
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Eero Lindholm
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Allan Linneberg
- Research Centre for Prevention and Health, Capital Region of Denmark, Copenhagen, Denmark
- Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Stéphane Lobbens
- CNRS UMR 8199, Institute of Biology and Lille 2 University, Pasteur Institute, Lille, France
| | - Jian'an Luan
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Valeriya Lyssenko
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
- Steno Diabetes Center, Gentofte, Denmark
| | - Satu Männistö
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Olga McLeod
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Julia Meyer
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Ghazala Mirza
- Biomedical Research Centre Genomics Core Facility, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, Guy's &St Thomas' Hospital, London, UK
| | - Thomas W Mühleisen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig Maximilians Universität, Munich, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig Maximilians Universität, Neuherberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Carmen Navarro
- Department of Epidemiology, Murcia Regional Health Council, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Health and Social Sciences, Universidad de Murcia, Murcia, Spain
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Nikolay N Oskolkov
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Katharine R Owen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Domenico Palli
- Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Sonali Pechlivanis
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, Essen, Germany
| | - Leena Peltonen
- Wellcome Trust Sanger Institute, Hinxton, UK
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Finnish Institute for Molecular Medicine (FIMM), Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - John R B Perry
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Carl G P Platou
- Nord-Trøndelag Health Study (HUNT) Research Center, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway
- Department of Internal Medicine, Levanger Hospital, Nord-Trondelag Health Trust, Levanger, Norway
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Dusseldorf, Dusseldorf, Germany
- German Center for Diabetes Research, partner site Dusseldorf, Dusseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Douglas Ruderfer
- Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Denis Rybin
- Boston University Data Coordinating Center, Boston, Massachusetts, USA
| | | | - Bengt Sennblad
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
| | - Gunnar Sigurðsson
- Landspitali University Hospital, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Gerald Steinbach
- Department of Clinical Chemistry and Central Laboratory, University of Ulm, Ulm, Germany
| | - Petter Storm
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig Maximilians Universität, Neuherberg, Germany
| | - Heather M Stringham
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Qi Sun
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Emmi Tikkanen
- Finnish Institute for Molecular Medicine (FIMM), Helsinki, Finland
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
| | - Anke Tonjes
- Integrated Treatment and Research (IFB) Center for Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Joseph Trakalo
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elena Tremoli
- Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
| | - Tiinamaija Tuomi
- Finnish Institute for Molecular Medicine (FIMM), Helsinki, Finland
- Folkhalsan Research Center, Helsinki, Finland
- Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
- Research Program for Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Roman Wennauer
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Steven Wiltshire
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | | | - Ian Dunham
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Lorenzo Pasquali
- Division of Endocrinology, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Jorge Ferrer
- Department of Medicine, Imperial College London, London, UK
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centre Esther Koplowitz, Barcelona, Spain
| | - Ruth J F Loos
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
- Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, USA
| | - Jose C Florez
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Diabetes Research Center, Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center at Baylor College of Medicine, Houston, Texas, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Netherlands Genomics Initiative, Netherlands Consortium for Healthy Ageing and Center for Medical Systems Biology, Rotterdam, the Netherlands
| | - Eric Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- General Medicine Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Frank B Hu
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Timo A Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Michael Stumvoll
- Integrated Treatment and Research (IFB) Center for Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Sirkka M Keinanen-Kiukaanniemi
- Faculty of Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
- Unit of General Practice, Oulu University Hospital, Oulu, Finland
| | | | - Timo E Saaristo
- Finnish Diabetes Association, Tampere, Finland
- Pirkanmaa District Hospital, Tampere, Finland
| | - Juha Saltevo
- Department of Medicine, Central Finland Central Hospital, Jyvasklya, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Raimund Erbel
- Clinic of Cardiology, West German Heart Centre, University Hospital of Essen, University Duisdurg-Essen, Essen, Germany
| | - Karl-Heinz Jöcke
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio, Finland
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, Essen, Germany
| | - Samuli Ripatti
- Wellcome Trust Sanger Institute, Hinxton, UK
- Finnish Institute for Molecular Medicine (FIMM), Helsinki, Finland
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
- Public Health Genomics Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Erik Ingelsson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Bernhard O Boehm
- Division of Endocrinology and Diabetes, Department of Internal Medicine, University Medical Centre Ulm, Ulm, Germany
- Lee Kong Chian School of Medicine, Imperial College London and Nanyang Technological University, Singapore
| | - Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Francis S Collins
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Heikki Koistinen
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Division of Endocrinology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
- Centre for Vascular Prevention, Danube University Krems, Krems, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kristian Hveem
- Nord-Trøndelag Health Study (HUNT) Research Center, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway
| | - Inger Njølstad
- Department of Community Medicine, Faculty of Health Sciences, University of Tromso, Tromso, Norway
| | - Panagiotis Deloukas
- Wellcome Trust Sanger Institute, Hinxton, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Peter J Donnelly
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Ulf de Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anders Hamsten
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Stephane Cauchi
- Lille Institute of Biology, European Genomics Institute of Diabetes, Lille, France
| | - Rob Sladek
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- McGill University and Génome Québec Innovation Centre, Montreal, Quebec, Canada
| | - Philippe Froguel
- Genomics of Common Disease, Imperial College London, London, UK
- Lille Institute of Biology, European Genomics Institute of Diabetes, Lille, France
- CNRS UMR 8199, Institute of Biology and Lille 2 University, Pasteur Institute, Lille, France
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrew D Morris
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Collin N A Palmer
- Diabetes Research Centre, Biomedical Research Institute, University of Dundee, Ninewells Hospital, Dundee, UK
- Pharmacogenomics Centre, Biomedical Research Institute, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Sekar Kathiresan
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Olle Melander
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Peter M Nilsson
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Leif C Groop
- Finnish Institute for Molecular Medicine (FIMM), Helsinki, Finland
- Lund University Diabetes Centre, Department of Clinical Science Malmo, Scania University Hospital, Lund University, Malmo, Sweden
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton, UK
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Christopher A O'Callaghan
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Anna L Gloyn
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - David Altshuler
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Diabetes Research Center, Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Molecular Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Boehnke
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Tanya M Teslovich
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | | |
Collapse
Collaborators
Goncalo R Abecasis, Peter Almgren, Mustafa Atalay, Damiano Baldassarre, Beverley Balkau, Inês Barroso, Nicola L Beer, John Beilby, Rafn Benediktsson, Richard N Bergman, Ewan Birney, Matthias Blüher, Bernhard O Boehm, Heiner Boeing, Eric Boerwinkle, Lori L Bonnycastle, Erwin P Borringer, Martijn van de Bunt, Noël P Burtt, Harry Campbell, Jason Carey, Stephane Cauchi, Guillaume Charpentier, Han Chen, Himanshu Chheda, Peter S Chines, Francis S Collins, Marilyn C Cornelis, David J Couper, Andrew T Crenshaw, Rob M van Dam, John Danesh, Ulf de Faire, George Dedoussis, Panagiotis Deloukas, Antigone S Dimas, Christian Dina, Alex S F Doney, Peter J Donnelly, Mozhgan Dorkhan, Cornelia van Duijn, Ian Dunham, Josée Dupuis, Sarah Edkins, Lewin Eisele, Valur Emilsson, Raimund Erbel, Johan G Eriksson, Tonu Esko, Elodie Eury, João Fadista, Teresa Ferreira, Jorge Ferrer, Jason Flannick, Jose C Florez, Pierre Fontanillas, Nita G Forouhi, Caroline Fox, Mattias Frånberg, Paul W Franks, Timothy M Frayling, Philippe Froguel, Christian Fuchsberger, Kyle J Gaulton, Karl Gertow, Christian Gieger, Bruna Gigante, Anna L Gloyn, Omri Gottesman, Harald Grallert, George B Grant, Niels Grarup, Todd Green, Leif C Groop, Christopher J Groves, Stefan Gustafsson, Anders Hamsten, Torben Hansen, Maija Hassinen, Andrew T Hattersley, Christian T Have, Caroline Hayward, Christian Herder, Albert Hofman, Oddgeir L Holmen, Momoko Horikoshi, Kees Hovingh, Astradur B Hreidarsson, Frank B Hu, Jennie Hui, Steve E Humphries, Sarah E Hunt, David J Hunter, Kristian Hveem, Thomas Illig, Erik Ingelsson, Anne U Jackson, Alan James, Karl-Heinz Jockel, Andrew D Johnson, Anna Jonsson, Marit E Jørgensen, Torben Jørgensen, Hyun Min Kang, Stavroula Kanoni, Wen Hong L Kao, Lennart C Karssen, Sekar Kathiresan, Sirkka M Keinanen-Kiukaanniemi, Nicola D Kerrison, Hans Kestler, Hassan Khan, Kay-Tee Khaw, Leena Kinnunen, Norman Klopp, Heikki Koistinen, Pirjo Komulainen, Augustine Kong, Eeva Korpi-Hyövӓlti, Peter Kovacs, Peter Kraft, Jasmina Kravic, Ashish Kumar, Johanna Kuusisto, Phoenix Kwan, Markku Laakso, Vasiliki Lagou, Timo A Lakka, Claudia Langenberg, Cordelia Langford, Karin Leander, Yeji Lee, Elisabeth M van Leeuwen, Man Li, Liming Liang, Peter Lichtner, Lars Lind, Michael Linderman, Cecilia M Lindgren, Eero Lindholm, Allan Linneberg, Ching-Ti Liu, Stéphane Lobbens, Adam Locke, Ruth J F Loos, Yingchang Lu, Jian'an Luan, Valeriya Lyssenko, Clement Ma, Reedik Mägi, Anubha Mahajan, Olga McLeod, James B Meigs, Olle Melander, Andres Metspalu, Julia Meyer, Evelin Mihailov, Ghazala Mirza, Susanne Moebus, Karen L Mohlke, Andrew D Morris, Andrew P Morris, Thomas W Mühleisen, Martina Müller-Nurasyid, Bill Musk, Satu Mӓnnistö, Carmen Navarro, Pau Navarro, Peter M Nilsson, Inger Njølstad, Markus M Nöthen, Christopher A O'Callaghan, Nikolay N Oskolkov, Katharine R Owen, Domenico Palli, Collin N A Palmer, James S Pankow, Lorenzo Pasquali, Sonali Pechlivanis, Nancy L Pedersen, Oluf Pedersen, Leena Peltonen, John R B Perry, Annette Peters, Carl G P Platou, Simon Potter, Jackie F Price, Inga Prokopenko, Lu Qi, Anne Raimondo, Loukianos Rallidis, Wolfgang Rathmann, Rainer Rauramaa, Soumya Raychaudhuri, N William Rayner, Emil Rehnberg, Michael E Reschen, Samuli Ripatti, Neil Robertson, Michael Roden, Elizabeth J Rossin, Igor Rudan, Douglas Ruderfer, Jana K Rundle, Denis Rybin, Timo E Saaristo, Danish Salehen, Veikko Salomaa, Juha Saltevo, Jouko Saramies, Yvonne T van der Schouw, Laura J Scott, Robert A Scott, Ayellet V Segre, Bengt Sennblad, Sonia Shah, Alan R Shuldiner, Gunnar Sigurðsson, Eric Sijbrands, Angela Silveira, Suthesh Sivapalaratnam, Rob Sladek, Thomas Sparso, Alena Stančáková, Kari Stefansson, Gerald Steinbach, Valgerdur Steinthorsdottir, Kathleen Stirrups, Petter Storm, Konstantin Strauch, Rona J Strawbridge, Heather M Stringham, Michael Stumvoll, Qi Sun, Ann-Christine Syvanen, Tanya M Teslovich, Soren K Thomsen, Barbara Thorand, Gudmar Thorleifsson, Unnur Thorsteinsdottir, Dorothee Thuillier, Emmi Tikkanen, Anke Tonjes, Joseph Trakalo, Elena Tremoli, Mieke D Trip, Tiinamaija Tuomi, Jaakko Tuomilehto, Andre G Uitterlinden, Sailaja Vedantam, Fabrizio Veglia, Benjamin F Voight, Simone Wahl, Nicholas J Wareham, Roman Wennauer, Sara M Willems, Tom Wilsgaard, James F Wilson, Steven Wiltshire, Andrew R Wood, Loic Yengo, Delilah Zabaneh, Eleftheria Zeggini, David Altshuler, Michael Boehnke, Mark I McCarthy,
Collapse
|
196
|
Han EH, Gorman AA, Singh P, Chi YI. Repression of HNF1α-mediated transcription by amino-terminal enhancer of split (AES). Biochem Biophys Res Commun 2015; 468:14-20. [PMID: 26549228 DOI: 10.1016/j.bbrc.2015.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/02/2015] [Indexed: 12/23/2022]
Abstract
HNF1α (Hepatocyte Nuclear Factor 1α) is one of the master regulators in pancreatic beta-cell development and function, and the mutations in Hnf1α are the most common monogenic causes of diabetes mellitus. As a member of the POU transcription factor family, HNF1α exerts its gene regulatory function through various molecular interactions; however, there is a paucity of knowledge in their functional complex formation. In this study, we identified the Groucho protein AES (Amino-terminal Enhancer of Split) as a HNF1α-specific physical binding partner and functional repressor of HNF1α-mediated transcription, which has a direct link to glucose-stimulated insulin secretion in beta-cells that is impaired in the HNF1α mutation-driven diabetes.
Collapse
Affiliation(s)
- Eun Hee Han
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Amanda A Gorman
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Puja Singh
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Young-In Chi
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN 55912, USA.
| |
Collapse
|
197
|
Shoemaker R, Yiannikouris F, Thatcher S, Cassis L. ACE2 deficiency reduces β-cell mass and impairs β-cell proliferation in obese C57BL/6 mice. Am J Physiol Endocrinol Metab 2015; 309:E621-31. [PMID: 26389599 PMCID: PMC4593774 DOI: 10.1152/ajpendo.00054.2015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/31/2015] [Indexed: 12/13/2022]
Abstract
Drugs that inhibit the renin-angiotensin system (RAS) decrease the onset of type 2 diabetes (T2D). Pancreatic islets express RAS components, including angiotensin-converting enzyme 2 (ACE2), which cleaves angiotensin II (Ang II) to angiotensin-(1-7) [Ang-(1-7)]. Overexpression of ACE2 in pancreas of diabetic mice improved glucose homeostasis. The purpose of this study was to determine if deficiency of endogenous ACE2 contributes to islet dysfunction and T2D. We hypothesized that ACE2 deficiency potentiates the decline in β-cell function and augments the development of diet-induced T2D. Male Ace2(+/y) or Ace2(-/y) mice were fed a low-fat (LF) or high-fat (HF) diet for 1 or 4 mo. A subset of 1-mo HF-fed mice were infused with Sal (Sal), losartan (Los), or Ang-(1-7). At 4 mo, while both genotypes of HF-fed mice developed a similar level of insulin resistance, adaptive hyperinsulinemia was reduced in Ace2(-/y) vs. Ace2(+/y) mice. Similarly, in vivo glucose-stimulated insulin secretion (GSIS) was reduced in 1-mo HF-fed Ace2(-/y) compared with Ace2(+/y) mice, resulting in augmented hyperglycemia. The average islet area was significantly smaller in both LF- and HF-fed Ace2(-/y) vs. Ace2(+/y) mice. Additionally, β-cell mass and proliferation were reduced significantly in HF-fed Ace2(-/y) vs. Ace2(+/y) mice. Neither infusion of Los nor Ang-(1-7) was able to correct impaired in vivo GSIS of HF-fed ACE2-deficient mice. These results demonstrate a critical role for endogenous ACE2 in the adaptive β-cell hyperinsulinemic response to HF feeding through regulation of β-cell proliferation and growth.
Collapse
Affiliation(s)
- Robin Shoemaker
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Frederique Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Sean Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Lisa Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
198
|
Affiliation(s)
- Leif Groop
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden; Finnish Institute for Molecular Medicine, Helsinki University, 20502 Helsinki, Finland.
| |
Collapse
|
199
|
Current Proceedings in the Molecular Dissection of Hepatocellular Adenomas: Review and Hands-on Guide for Diagnosis. Int J Mol Sci 2015; 16:20994-1007. [PMID: 26404250 PMCID: PMC4613237 DOI: 10.3390/ijms160920994] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 08/10/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023] Open
Abstract
Molecular dissection of hepatocellular adenomas has brought forward a diversity of well-defined entities. Their distinction is important for routine practice, since prognosis is tightly related to the individual subgroup. Very recent activity has generated new details on the molecular background of hepatocellular adenoma, which this article aims to integrate into the current concepts of taxonomy.
Collapse
|
200
|
Brial F, Lussier CR, Belleville K, Sarret P, Boudreau F. Ghrelin Inhibition Restores Glucose Homeostasis in Hepatocyte Nuclear Factor-1α (MODY3)-Deficient Mice. Diabetes 2015; 64:3314-20. [PMID: 25979074 DOI: 10.2337/db15-0124] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 05/11/2015] [Indexed: 11/13/2022]
Abstract
Hepatocyte nuclear factor-1α (HNF1α) is a transcription factor expressed in tissues of endoderm origin. Mutations in HNF1A are associated with maturity-onset diabetes of the young 3 (MODY3). Mice deficient for Hnf1α are hyperglycemic, with their pancreatic β-cells being defective in glucose-sensing insulin secretion. The specific mechanisms involved in this defect are unclear. Gut hormones control glucose homeostasis. Our objective was to explore whether changes in these hormones play a role in glucose homeostasis in the absence of Hnf1α. An increase in ghrelin gene transcript and a decrease in glucose-dependent insulinotropic polypeptide (GIP) gene transcripts were observed in the gut of Hnf1α-null mice. These changes correlated with an increase of ghrelin and a decrease of GIP-labeled cells. Ghrelin serological levels were significantly induced in Hnf1α-null mice. Paradoxically, GIP levels were also induced in these mice. Treatment of Hnf1α-null mice with a ghrelin antagonist led to a recovery of the diabetic symptoms. We conclude that upregulation of ghrelin in the absence of Hnf1α impairs insulin secretion and can be reversed by pharmacological inhibition of ghrelin/GHS-R interaction. These observations open up on future strategies to counteract ghrelin action in a program that could become beneficial in controlling non-insulin-dependent diabetes.
Collapse
Affiliation(s)
- François Brial
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Quebec, Canada
| | - Carine R Lussier
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Quebec, Canada
| | - Karine Belleville
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - Philippe Sarret
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - François Boudreau
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Quebec, Canada
| |
Collapse
|