51
|
Abstract
PURPOSE OF REVIEW Genetic obesity is responsible for up to 7% of severe childhood obesity. Although current Pediatric Endocrine Society guidelines recommend assessment of children with early-onset morbid obesity and hyperphagia for underlying genetic disorders, a vast majority of patients are not being appropriately screened for genetic obesity syndromes. RECENT FINDINGS With advances in genetic testing, more genetic causes of obesity are being identified. Treatments are likely to be individualized, depending on the cause of the obesity, and must be targeted at addressing the underlying cause. Investigational therapies include melanocortin-4 receptor antagonists, oxytocin and medications targeting the endocannabinoid system. SUMMARY Improved identification of patients with genetic obesity syndromes will lead to development of new treatments and personalized management of these diseases.
Collapse
|
52
|
Aristidou C, Theodosiou A, Ketoni A, Bak M, Mehrjouy MM, Tommerup N, Sismani C. Cryptic breakpoint identified by whole-genome mate-pair sequencing in a rare paternally inherited complex chromosomal rearrangement. Mol Cytogenet 2018; 11:34. [PMID: 29930709 PMCID: PMC5991433 DOI: 10.1186/s13039-018-0384-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/15/2018] [Indexed: 11/21/2022] Open
Abstract
Background Precise characterization of apparently balanced complex chromosomal rearrangements in non-affected individuals is crucial as they may result in reproductive failure, recurrent miscarriages or affected offspring. Case presentation We present a family, where the non-affected father and daughter were found, using FISH and karyotyping, to be carriers of a three-way complex chromosomal rearrangement [t(6;7;10)(q16.2;q34;q26.1), de novo in the father]. The family suffered from two stillbirths, one miscarriage, and has a son with severe intellectual disability. In the present study, the family was revisited using whole-genome mate-pair sequencing. Interestingly, whole-genome mate-pair sequencing revealed a cryptic breakpoint on derivative (der) chromosome 6 rendering the rearrangement even more complex. FISH using a chromosome (chr) 6 custom-designed probe and a chr10 control probe confirmed that the interstitial chr6 segment, created by the two chr6 breakpoints, was translocated onto der(10). Breakpoints were successfully validated with Sanger sequencing, and small imbalances as well as microhomology were identified. Finally, the complex chromosomal rearrangement breakpoints disrupted the SIM1, GRIK2, CNTNAP2, and PTPRE genes without causing any phenotype development. Conclusions In contrast to the majority of maternally transmitted complex chromosomal rearrangement cases, our study investigated a rare case where a complex chromosomal rearrangement, which most probably resulted from a Type IV hexavalent during the pachytene stage of meiosis I, was stably transmitted from a fertile father to his non-affected daughter. Whole-genome mate-pair sequencing proved highly successful in identifying cryptic complexity, which consequently provided further insight into the meiotic segregation of chromosomes and the increased reproductive risk in individuals carrying the specific complex chromosomal rearrangement. We propose that such complex rearrangements should be characterized in detail using a combination of conventional cytogenetic and NGS-based approaches to aid in better prenatal preimplantation genetic diagnosis and counseling in couples with reproductive problems.
Collapse
Affiliation(s)
- Constantia Aristidou
- 1Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,2The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Athina Theodosiou
- 1Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Andria Ketoni
- 1Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Mads Bak
- 3Wilhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mana M Mehrjouy
- 3Wilhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Niels Tommerup
- 3Wilhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Carolina Sismani
- 1Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,2The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
53
|
Geets E, Meuwissen MEC, Van Hul W. Clinical, molecular genetics and therapeutic aspects of syndromic obesity. Clin Genet 2018; 95:23-40. [PMID: 29700824 DOI: 10.1111/cge.13367] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 12/13/2022]
Abstract
Obesity has become a major health problem worldwide. To date, more than 25 different syndromic forms of obesity are known in which one (monogenic) or multiple (polygenic) genes are involved. This review gives an overview of these forms and focuses more in detail on 6 syndromes: Prader Willi Syndrome and Prader Willi like phenotype, Bardet Biedl Syndrome, Alström Syndrome, Wilms tumor, Aniridia, Genitourinary malformations and mental Retardation syndrome and 16p11.2 (micro)deletions. Years of research provided plenty of information on the molecular genetics of these disorders and the obesity phenotype leading to a more individualized treatment of the symptoms, however, many questions still remain unanswered. As these obesity syndromes have different signs and symptoms in common, it makes it difficult to accurately diagnose patients which may result in inappropriate treatment of the disease. Therefore, the big challenge for clinicians and scientists is to more clearly differentiate all syndromic forms of obesity to provide conclusive genetic explanations and eventually deliver accurate genetic counseling and treatment. In addition, further delineation of the (functions of the) underlying genes with the use of array- or next-generation sequencing-based technology will be helpful to unravel the mechanisms of energy metabolism in the general population.
Collapse
Affiliation(s)
- E Geets
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - M E C Meuwissen
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - W Van Hul
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
54
|
Candelo E, Feinstein MM, Ramirez-Montaño D, Gomez JF, Pachajoa H. First Case Report of Prader-Willi-Like Syndrome in Colombia. Front Genet 2018; 9:98. [PMID: 29619043 PMCID: PMC5871659 DOI: 10.3389/fgene.2018.00098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/08/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Prader-Willi-like syndrome (PWLS) is believed to be caused by a variety of disruptions in genetic pathways both inside and outside of the genetic region implicated in PWS. By definition, PWLS does not demonstrate mutations in the 15q11-q13 region itself. It is a rare disorder whose clinical hallmarks include hypotonia, obesity, short extremities, and delayed development. This syndrome has been described in patients with 1p, 2p, 3p, 6q, and 9q chromosome abnormalities and in cases with maternal uniparental disomy of chromosome 14 and fragile X syndrome. Case presentation: In the present report, we describe a 9-year-old Colombian patient who demonstrated features of PWS and was ultimately diagnosed with PWLS after genetic analysis revealed a 14.97 Mb deletion of 6q16.1-q21. Conclusions: This is the first reported case of PWLS in Colombia and represents one of the largest documented 6q21 deletions.
Collapse
Affiliation(s)
| | - Max M Feinstein
- Health School, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | | | - Juan F Gomez
- Paediatric Neurology, Fundación Valle Del Lili, Cali, Colombia
| | - Harry Pachajoa
- Health Sciences Faculty, Universidad Icesi, Cali, Colombia.,Genetics Department, Fundacion Valle del Lili, Cali, Colombia
| |
Collapse
|
55
|
Timper K, Brüning JC. Hypothalamic circuits regulating appetite and energy homeostasis: pathways to obesity. Dis Model Mech 2018; 10:679-689. [PMID: 28592656 PMCID: PMC5483000 DOI: 10.1242/dmm.026609] [Citation(s) in RCA: 507] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The ‘obesity epidemic’ represents a major global socioeconomic burden that urgently calls for a better understanding of the underlying causes of increased weight gain and its associated metabolic comorbidities, such as type 2 diabetes mellitus and cardiovascular diseases. Improving our understanding of the cellular basis of obesity could set the stage for the development of new therapeutic strategies. The CNS plays a pivotal role in the regulation of energy and glucose homeostasis. Distinct neuronal cell populations, particularly within the arcuate nucleus of the hypothalamus, sense the nutrient status of the organism and integrate signals from peripheral hormones including pancreas-derived insulin and adipocyte-derived leptin to regulate calorie intake, glucose metabolism and energy expenditure. The arcuate neurons are tightly connected to other specialized neuronal subpopulations within the hypothalamus, but also to various extrahypothalamic brain regions, allowing a coordinated behavioral response. This At a Glance article gives an overview of the recent knowledge, mainly derived from rodent models, regarding the CNS-dependent regulation of energy and glucose homeostasis, and illustrates how dysregulation of the neuronal networks involved can lead to overnutrition and obesity. The potential impact of recent research findings in the field on therapeutic treatment strategies for human obesity is also discussed. Summary: This at a glance article gives an overview of the recent knowledge mainly derived from rodent models regarding the CNS-dependent regulation of energy and glucose homeostasis, and depicts how dysregulation of the involved neuronal networks promotes overnutrition and obesity.
Collapse
Affiliation(s)
- Katharina Timper
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, Cologne 50931, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, Cologne 50924, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, Cologne 50931, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, Cologne 50931, Germany .,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, Cologne 50924, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, Cologne 50931, Germany.,National Center for Diabetes Research (DZD), Ingolstädter Land Str. 1, Neuherberg 85764, Germany
| |
Collapse
|
56
|
D’Angelo CS, Varela MC, de Castro CIE, Otto PA, Perez ABA, Lourenço CM, Kim CA, Bertola DR, Kok F, Garcia-Alonso L, Koiffmann CP. Chromosomal microarray analysis in the genetic evaluation of 279 patients with syndromic obesity. Mol Cytogenet 2018; 11:14. [PMID: 29441128 PMCID: PMC5800070 DOI: 10.1186/s13039-018-0363-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/22/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Syndromic obesity is an umbrella term used to describe cases where obesity occurs with additional phenotypes. It often arises as part of a distinct genetic syndrome with Prader-Willi syndrome being a classical example. These rare forms of obesity provide a unique source for identifying obesity-related genetic changes. Chromosomal microarray analysis (CMA) has allowed the characterization of new genetic forms of syndromic obesity, which are due to copy number variants (CNVs); however, CMA in large cohorts requires more study. The aim of this study was to characterize the CNVs detected by CMA in 279 patients with a syndromic obesity phenotype. RESULTS Pathogenic CNVs were detected in 61 patients (22%) and, among them, 35 had overlapping/recurrent CNVs. Genomic imbalance disorders known to cause syndromic obesity were found in 8.2% of cases, most commonly deletions of 1p36, 2q37 and 17p11.2 (5.4%), and we also detected deletions at 1p21.3, 2p25.3, 6q16, 9q34, 16p11.2 distal and proximal, as well as an unbalanced translocation resulting in duplication of the GNB3 gene responsible for a syndromic for of childhood obesity. Deletions of 9p terminal and 22q11.2 proximal/distal were found in 1% and 3% of cases, respectively. They thus emerge as being new putative obesity-susceptibility loci. We found additional CNVs in our study that overlapped with CNVs previously reported in cases of syndromic obesity, including a new case of 13q34 deletion (CHAMP1), bringing to 7 the number of patients in whom such defects have been described in association with obesity. Our findings implicate many genes previously associated with obesity (e.g. PTBP2, TMEM18, MYT1L, POU3F2, SIM1, SH2B1), and also identified other potentially relevant candidates including TAS1R3, ALOX5AP, and GAS6. CONCLUSION Understanding the genetics of obesity has proven difficult, and considerable insight has been obtained from the study of genomic disorders with obesity associated as part of the phenotype. In our study, CNVs known to be causal for syndromic obesity were detected in 8.2% of patients, but we provide evidence for a genetic basis of obesity in as many as 14% of cases. Overall, our results underscore the genetic heterogeneity in syndromic forms of obesity, which imposes a substantial challenge for diagnosis.
Collapse
Affiliation(s)
- Carla Sustek D’Angelo
- Human Genome and Stem Cell Research Center (HUG-CELL), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Rua do Matao no 277, Cidade Universitaria-Butanta, Sao Paulo, SP 05508-090 Brazil
| | - Monica Castro Varela
- Human Genome and Stem Cell Research Center (HUG-CELL), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Rua do Matao no 277, Cidade Universitaria-Butanta, Sao Paulo, SP 05508-090 Brazil
| | - Claudia Irene Emílio de Castro
- Human Genome and Stem Cell Research Center (HUG-CELL), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Rua do Matao no 277, Cidade Universitaria-Butanta, Sao Paulo, SP 05508-090 Brazil
| | - Paulo Alberto Otto
- Human Genome and Stem Cell Research Center (HUG-CELL), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Rua do Matao no 277, Cidade Universitaria-Butanta, Sao Paulo, SP 05508-090 Brazil
| | - Ana Beatriz Alvarez Perez
- Department of Morphology and Genetics, Paulista School of Medicine, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP Brazil
| | - Charles Marques Lourenço
- Neurogenetics Unit, Clinics Hospital of Ribeirao Preto, Faculty of Medicine, University of Sao Paulo, FMRP-USP, Ribeirao Preto, SP Brazil
| | - Chong Ae Kim
- Genetic Unit, Children’s Institute, Faculty of Medicine, University of Sao Paulo, FMUSP, Sao Paulo, SP Brazil
| | - Debora Romeo Bertola
- Genetic Unit, Children’s Institute, Faculty of Medicine, University of Sao Paulo, FMUSP, Sao Paulo, SP Brazil
| | - Fernando Kok
- Department of Neurology, Faculty of Medicine, University of Sao Paulo, FMUSP, Sao Paulo, SP Brazil
| | - Luis Garcia-Alonso
- Department of Morphology and Genetics, Paulista School of Medicine, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP Brazil
| | - Celia Priszkulnik Koiffmann
- Human Genome and Stem Cell Research Center (HUG-CELL), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Rua do Matao no 277, Cidade Universitaria-Butanta, Sao Paulo, SP 05508-090 Brazil
| |
Collapse
|
57
|
Foucan L, Larifla L, Durand E, Rambhojan C, Armand C, Michel CT, Billy R, Dhennin V, De Graeve F, Rabearivelo I, Sand O, Lacorte JM, Froguel P, Bonnefond A. High Prevalence of Rare Monogenic Forms of Obesity in Obese Guadeloupean Afro-Caribbean Children. J Clin Endocrinol Metab 2018; 103:539-545. [PMID: 29216354 DOI: 10.1210/jc.2017-01956] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 11/30/2017] [Indexed: 11/19/2022]
Abstract
CONTEXT The population of Guadeloupe Island exhibits a high prevalence of obesity. OBJECTIVE We aimed to investigate whether rare genetic mutations in genes involved in monogenic obesity (or diabetes) might be causal in this population of Afro-Caribbean ancestry. DESIGN AND SETTING This was a secondary analysis of a study on obesity conducted in schoolchildren from Guadeloupe in 2013 that aimed to assess changes in children's profiles after a lifestyle intervention program. Through next-generation sequencing, we sequenced coding regions of 59 genes involved in monogenic obesity or diabetes in participants from this study. PARTICIPANTS AND INTERVENTIONS A total of 25 obese schoolchildren from Guadeloupe were screened for rare mutations (nonsynonymous, splice-site, or insertion/deletion) in 59 genes. MAIN OUTCOME MEASURES Correlation between phenotypes and mutations of interest. RESULTS We detected five rare heterozygous mutations in five different children with obesity: MC4R p.Ile301Thr and SIM1 p.Val326Thrfs*43 mutations that were pathogenic; SIM1 p.Ser343Pro and SH2B1 p.Pro90His mutations that were likely pathogenic; and NTRK2 p.Leu140Phe that was of uncertain significance. In parallel, we identified seven carriers of mutations in ABCC8 (p.Lys1521Asn and p.Ala625Val) or KCNJ11 (p.Val13Met and p.Val151Met) that were of uncertain significance. CONCLUSIONS We were able to detect pathogenic or likely pathogenic mutations linked to severe obesity in >15% of this population, which is much higher than what we observed in Europeans (∼5%).
Collapse
Affiliation(s)
- Lydia Foucan
- Research Team on Cardiometabolic Risk, University of Antilles, Pointe-à-Pitre, Guadeloupe, France
- Department of Public Health, University Hospital, Pointe-à-Pitre, Guadeloupe, France
| | - Laurent Larifla
- Research Team on Cardiometabolic Risk, University of Antilles, Pointe-à-Pitre, Guadeloupe, France
- Cardiology Unit, University Hospital, Pointe-à-Pitre, Guadeloupe, France
| | - Emmanuelle Durand
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Christine Rambhojan
- Research Team on Cardiometabolic Risk, University of Antilles, Pointe-à-Pitre, Guadeloupe, France
| | - Christophe Armand
- Research Team on Cardiometabolic Risk, University of Antilles, Pointe-à-Pitre, Guadeloupe, France
- Department of Public Health, University Hospital, Pointe-à-Pitre, Guadeloupe, France
| | - Carl-Thony Michel
- Cardiology Unit, University Hospital, Pointe-à-Pitre, Guadeloupe, France
| | - Rachel Billy
- Cardiology Unit, University Hospital, Pointe-à-Pitre, Guadeloupe, France
| | - Véronique Dhennin
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Franck De Graeve
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Iandry Rabearivelo
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Olivier Sand
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Jean-Marc Lacorte
- Department of Endocrine and Oncological Biochemistry, University Hospitals of Pitié-Salpétrière‒Charles Foix, Paris, France
- Inserm, Research Institute of Cardiovascular Disease, Metabolism and Nutrition, Paris, France
| | - Philippe Froguel
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
| | - Amélie Bonnefond
- CNRS, European Genomic Institute for Diabetes, Institut Pasteur de Lille, University of Lille, Lille, France
- Department of Genomics of Common Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
58
|
|
59
|
van der Klaauw AA. Neuropeptides in Obesity and Metabolic Disease. Clin Chem 2017; 64:173-182. [PMID: 29097517 DOI: 10.1373/clinchem.2017.281568] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/18/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND The global rise in the prevalence of obesity and associated comorbidities such as type 2 diabetes, cardiovascular disease, and cancer represents a major public health concern. CONTENT Studies in rodents with the use of global and targeted gene disruption, and mapping of neurocircuitry by using optogenetics and designer receptors exclusively activated by designer drugs (DREADDs) have greatly advanced our understanding of the neural control of body weight. In conjunction with analytical chemistry techniques involving classical immunoassays and mass spectrometry, many neuropeptides that are key to energy homeostasis have been identified. The actions of neuropeptides are diverse, from paracrine modulation of local neurotransmission to hormonal control of distant target organs. SUMMARY Multiple hormones, such as the adipocyte-derived leptin, insulin, and gut hormones, and nutrients signal peripheral energy state to the central nervous system. Neurons in distinct areas of the hypothalamus and brainstem integrate and translate this information by both direct inhibitory/excitatory projections and anorexigenic or orexigenic neuropeptides into actions on food intake and energy expenditure. The importance of these neuropeptides in human energy balance is most powerfully illustrated by genetic forms of obesity that involve neuropeptides such as melanocortin-4-receptor (MC4R) deficiency. Drugs that mimic the actions of neuropeptides are being tested for the treatment of obesity. Successful therapeutic strategies in obesity will require in-depth knowledge of the neuronal circuits they are working in, the downstream targets, and potential compensatory mechanisms.
Collapse
Affiliation(s)
- Agatha A van der Klaauw
- Department of Clinical Biochemistry, Metabolic Research Laboratories - Institute of Metabolic Science, University of Cambridge, Cambridge, England.
| |
Collapse
|
60
|
Moir L, Bochukova EG, Dumbell R, Banks G, Bains RS, Nolan PM, Scudamore C, Simon M, Watson KA, Keogh J, Henning E, Hendricks A, O'Rahilly S, Barroso I, Sullivan AE, Bersten DC, Whitelaw ML, Kirsch S, Bentley E, Farooqi IS, Cox RD. Disruption of the homeodomain transcription factor orthopedia homeobox (Otp) is associated with obesity and anxiety. Mol Metab 2017; 6:1419-1428. [PMID: 29107289 PMCID: PMC5681237 DOI: 10.1016/j.molmet.2017.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022] Open
Abstract
Objective Genetic studies in obese rodents and humans can provide novel insights into the mechanisms involved in energy homeostasis. Methods In this study, we genetically mapped the chromosomal region underlying the development of severe obesity in a mouse line identified as part of a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis screen. We characterized the metabolic and behavioral phenotype of obese mutant mice and examined changes in hypothalamic gene expression. In humans, we examined genetic data from people with severe early onset obesity. Results We identified an obese mouse heterozygous for a missense mutation (pR108W) in orthopedia homeobox (Otp), a homeodomain containing transcription factor required for the development of neuroendocrine cell lineages in the hypothalamus, a region of the brain important in the regulation of energy homeostasis. OtpR108W/+ mice exhibit increased food intake, weight gain, and anxiety when in novel environments or singly housed, phenotypes that may be partially explained by reduced hypothalamic expression of oxytocin and arginine vasopressin. R108W affects the highly conserved homeodomain, impairs DNA binding, and alters transcriptional activity in cells. We sequenced OTP in 2548 people with severe early-onset obesity and found a rare heterozygous loss of function variant in the homeodomain (Q153R) in a patient who also had features of attention deficit disorder. Conclusions OTP is involved in mammalian energy homeostasis and behavior and appears to be necessary for the development of hypothalamic neural circuits. Further studies will be needed to investigate the contribution of rare variants in OTP to human energy homeostasis. A mouse Otp mutation alters hypothalamic neuropeptide expression leading to increased food intake, obesity and anxiety. In severe early onset obesity, we found a heterozygous LOF variant in a patient with attention deficit disorder features. These studies show for the first time that mutations in the Otp/OTP gene cause obesity.
Collapse
Affiliation(s)
- Lee Moir
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Elena G Bochukova
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK; The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Rebecca Dumbell
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Gareth Banks
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Rasneer S Bains
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Patrick M Nolan
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Cheryl Scudamore
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Michelle Simon
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Kimberly A Watson
- School of Biological Sciences, University of Reading, Reading, Berkshire, UK
| | - Julia Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Audrey Hendricks
- Wellcome Trust Sanger Institute, Cambridge, UK; Department of Mathematical and Statistical Sciences, University of Colorado-Denver, Denver, CO 80204, USA
| | - Stephen O'Rahilly
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | | | | | - Adrienne E Sullivan
- Department Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - David C Bersten
- Department Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - Murray L Whitelaw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK; Department Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - Susan Kirsch
- Department of Endocrinology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada
| | - Elizabeth Bentley
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| | - Roger D Cox
- MRC Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
61
|
Pagel KA, Pejaver V, Lin GN, Nam HJ, Mort M, Cooper DN, Sebat J, Iakoucheva LM, Mooney SD, Radivojac P. When loss-of-function is loss of function: assessing mutational signatures and impact of loss-of-function genetic variants. Bioinformatics 2017; 33:i389-i398. [PMID: 28882004 PMCID: PMC5870554 DOI: 10.1093/bioinformatics/btx272] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MOTIVATION Loss-of-function genetic variants are frequently associated with severe clinical phenotypes, yet many are present in the genomes of healthy individuals. The available methods to assess the impact of these variants rely primarily upon evolutionary conservation with little to no consideration of the structural and functional implications for the protein. They further do not provide information to the user regarding specific molecular alterations potentially causative of disease. RESULTS To address this, we investigate protein features underlying loss-of-function genetic variation and develop a machine learning method, MutPred-LOF, for the discrimination of pathogenic and tolerated variants that can also generate hypotheses on specific molecular events disrupted by the variant. We investigate a large set of human variants derived from the Human Gene Mutation Database, ClinVar and the Exome Aggregation Consortium. Our prediction method shows an area under the Receiver Operating Characteristic curve of 0.85 for all loss-of-function variants and 0.75 for proteins in which both pathogenic and neutral variants have been observed. We applied MutPred-LOF to a set of 1142 de novo vari3ants from neurodevelopmental disorders and find enrichment of pathogenic variants in affected individuals. Overall, our results highlight the potential of computational tools to elucidate causal mechanisms underlying loss of protein function in loss-of-function variants. AVAILABILITY AND IMPLEMENTATION http://mutpred.mutdb.org. CONTACT predrag@indiana.edu.
Collapse
Affiliation(s)
- Kymberleigh A Pagel
- Department of Computer Science and Informatics, Indiana University, Bloomington, IN, USA
| | - Vikas Pejaver
- Department of Computer Science and Informatics, Indiana University, Bloomington, IN, USA
| | - Guan Ning Lin
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Hyun-Jun Nam
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Matthew Mort
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - Jonathan Sebat
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Beyster Center for Psychiatric Genomics, Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Lilia M Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Sean D Mooney
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Predrag Radivojac
- Department of Computer Science and Informatics, Indiana University, Bloomington, IN, USA
| |
Collapse
|
62
|
Kaur Y, de Souza RJ, Gibson WT, Meyre D. A systematic review of genetic syndromes with obesity. Obes Rev 2017; 18:603-634. [PMID: 28346723 DOI: 10.1111/obr.12531] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 11/29/2022]
Abstract
Syndromic monogenic obesity typically follows Mendelian patterns of inheritance and involves the co-presentation of other characteristics, such as mental retardation, dysmorphic features and organ-specific abnormalities. Previous reviews on obesity have reported 20 to 30 syndromes but no systematic review has yet been conducted on syndromic obesity. We searched seven databases using terms such as 'obesity', 'syndrome' and 'gene' to conduct a systematic review of literature on syndromic obesity. Our literature search identified 13,719 references. After abstract and full-text review, 119 relevant papers were eligible, and 42 papers were identified through additional searches. Our analysis of these 161 papers found that 79 obesity syndromes have been reported in literature. Of the 79 syndromes, 19 have been fully genetically elucidated, 11 have been partially elucidated, 27 have been mapped to a chromosomal region and for the remaining 22, neither the gene(s) nor the chromosomal location(s) have yet been identified. Interestingly, 54.4% of the syndromes have not been assigned a name, whereas 13.9% have more than one name. We report on organizational inconsistencies (e.g. naming discrepancies and syndrome classification) and provide suggestions for improvements. Overall, this review illustrates the need for increased clinical and genetic research on syndromes with obesity.
Collapse
Affiliation(s)
- Y Kaur
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - R J de Souza
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - W T Gibson
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,British Columbia Children's Hospital Research Institute, Vancouver, Canada
| | - D Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
63
|
Stanikova D, Buzga M, Krumpolec P, Skopkova M, Surova M, Ukropcova B, Ticha L, Petrasova M, Gabcova D, Huckova M, Piskorova L, Bozensky J, Mokan M, Ukropec J, Zavacka I, Klimes I, Stanik J, Gasperikova D. Genetic analysis of single-minded 1 gene in early-onset severely obese children and adolescents. PLoS One 2017; 12:e0177222. [PMID: 28472148 PMCID: PMC5417716 DOI: 10.1371/journal.pone.0177222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/24/2017] [Indexed: 11/23/2022] Open
Abstract
Background Inactivating mutations of the hypothalamic transcription factor singleminded1 (SIM1) have been shown as a cause of early-onset severe obesity. However, to date, the contribution of SIM1 mutations to the obesity phenotype has only been studied in a few populations. In this study, we screened the functional regions of SIM1 in severely obese children of Slovak and Moravian descent to determine if genetic variants within SIM1 may influence the development of obesity in these populations. Methods The SIM1 promoter region, exons and exon-intron boundaries were sequenced in 126 unrelated obese children and adolescents (2–18 years of age) and 41 adult lean controls of Slovak and Moravian origin. Inclusion criteria for the children and adolescents were a body mass index standard deviation score higher than 2 SD for an appropriate age and sex, and obesity onset at less than 5 years of age. The clinical phenotypes of the SIM1 variant carriers were compared with clinical phenotypes of 4 MC4R variant carriers and with 27 unrelated SIM1 and MC4R mutation negative obese controls that were matched for age and gender. Results Seven previously described SIM1 variants and one novel heterozygous variant p.D134N were identified. The novel variant was predicted to be pathogenic by 7 in silico software analyses and is located at a highly conserved position of the SIM1 protein. The p.D134N variant was found in an 18 year old female proband (BMI 44.2kg/m2; +7.5 SD), and in 3 obese family members. Regardless of early onset severe obesity, the proband and her brother (age 16 years) did not fulfill the criteria of metabolic syndrome. Moreover, the variant carriers had significantly lower preferences for high sugar (p = 0.02) and low fat, low carbohydrate, high protein (p = 0.02) foods compared to the obese controls. Conclusions We have identified a novel SIM1 variant, p.D134N, in 4 obese individuals from a single pedigree which is also associated with lower preference for certain foods.
Collapse
Affiliation(s)
- Daniela Stanikova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Pediatrics, Medical Faculty of Comenius University, Bratislava, Slovakia
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - Marek Buzga
- Department of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Patrik Krumpolec
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Martina Skopkova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Martina Surova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbara Ukropcova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Pathophysiolgy, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Lubica Ticha
- Department of Pediatrics, Medical Faculty of Comenius University, Bratislava, Slovakia
| | - Miroslava Petrasova
- Department of Pediatrics, Medical Faculty of Safarik University, Kosice, Slovakia
| | - Dominika Gabcova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Miroslava Huckova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucie Piskorova
- Department of Pediatrics, Vitkovice Hospital, Ostrava, Czech Republic
| | - Jan Bozensky
- Department of Pediatrics, Vitkovice Hospital, Ostrava, Czech Republic
| | - Marian Mokan
- Department of Internal Medicine, Jessenius Medical Faculty of Comenius University, Martin, Slovakia
| | - Jozef Ukropec
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ivona Zavacka
- Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Iwar Klimes
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Juraj Stanik
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Pediatrics, Medical Faculty of Comenius University, Bratislava, Slovakia
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | - Daniela Gasperikova
- Laboratory of Diabetes and Metabolic Disorders, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- * E-mail:
| |
Collapse
|
64
|
Windholz J, Kovacs P, Schlicke M, Franke C, Mahajan A, Morris AP, Lemke JR, Klammt J, Kiess W, Schöneberg T, Pfäffle R, Körner A. Copy number variations in "classical" obesity candidate genes are not frequently associated with severe early-onset obesity in children. J Pediatr Endocrinol Metab 2017; 30:507-515. [PMID: 28593922 DOI: 10.1515/jpem-2016-0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/06/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Obesity is genetically heterogeneous and highly heritable, although polymorphisms explain the phenotype in only a small proportion of obese children. We investigated the presence of copy number variations (CNVs) in "classical" genes known to be associated with (monogenic) early-onset obesity in children. METHODS In 194 obese Caucasian children selected for early-onset and severe obesity from our obesity cohort we screened for deletions and/or duplications by multiplex ligation-dependent probe amplification reaction (MLPA). As we found one MLPA probe to interfere with a polymorphism in SIM1 we investigated its association with obesity and other phenotypic traits in our extended cohort of 2305 children. RESULTS In the selected subset of most severely obese children, we did not find CNV with MLPA in POMC, LEP, LEPR, MC4R, MC3R or MC2R genes. However, one SIM1 probe located at exon 9 gave signals suggestive for SIM1 insufficiency in 52 patients. Polymerase chain reaction (PCR) analysis identified this as a false positive result due to interference with single nucleotide polymorphism (SNP) rs3734354/rs3734355. We, therefore, investigated for associations of this polymorphism with obesity and metabolic traits in our extended cohort. We found rs3734354/rs3734355 to be associated with body mass index-standard deviation score (BMI-SDS) (p = 0.003), but not with parameters of insulin metabolism, blood pressure or food intake. CONCLUSIONS In our modest sample of severely obese children, we were unable to find CNVs in well-established monogenic obesity genes. Nevertheless, we found an association of rs3734354 in SIM1 with obesity of early-onset type in children, although not with obesity-related traits.
Collapse
Affiliation(s)
- Jan Windholz
- Pediatric Research Center, University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig
| | - Peter Kovacs
- Leipzig University Medical Center, IFB AdiposityDiseases, University of Leipzig, Leipzig
| | - Marina Schlicke
- Pediatric Research Center, University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig
| | - Christin Franke
- Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Johannes R Lemke
- Institute of Human Genetics, Medical Faculty, University of Leipzig, Leipzig
| | - Jürgen Klammt
- Pediatric Research Center, University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig
| | - Wieland Kiess
- Pediatric Research Center, University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig
| | - Torsten Schöneberg
- Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig
| | - Roland Pfäffle
- Pediatric Research Center, University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig
| | - Antje Körner
- Pediatric Research Center, University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig
| |
Collapse
|
65
|
Recent progress in genetics, epigenetics and metagenomics unveils the pathophysiology of human obesity. Clin Sci (Lond) 2017; 130:943-86. [PMID: 27154742 DOI: 10.1042/cs20160136] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/24/2016] [Indexed: 12/19/2022]
Abstract
In high-, middle- and low-income countries, the rising prevalence of obesity is the underlying cause of numerous health complications and increased mortality. Being a complex and heritable disorder, obesity results from the interplay between genetic susceptibility, epigenetics, metagenomics and the environment. Attempts at understanding the genetic basis of obesity have identified numerous genes associated with syndromic monogenic, non-syndromic monogenic, oligogenic and polygenic obesity. The genetics of leanness are also considered relevant as it mirrors some of obesity's aetiologies. In this report, we summarize ten genetically elucidated obesity syndromes, some of which are involved in ciliary functioning. We comprehensively review 11 monogenic obesity genes identified to date and their role in energy maintenance as part of the leptin-melanocortin pathway. With the emergence of genome-wide association studies over the last decade, 227 genetic variants involved in different biological pathways (central nervous system, food sensing and digestion, adipocyte differentiation, insulin signalling, lipid metabolism, muscle and liver biology, gut microbiota) have been associated with polygenic obesity. Advances in obligatory and facilitated epigenetic variation, and gene-environment interaction studies have partly accounted for the missing heritability of obesity and provided additional insight into its aetiology. The role of gut microbiota in obesity pathophysiology, as well as the 12 genes associated with lipodystrophies is discussed. Furthermore, in an attempt to improve future studies and merge the gap between research and clinical practice, we provide suggestions on how high-throughput '-omic' data can be integrated in order to get closer to the new age of personalized medicine.
Collapse
|
66
|
Liu R, Zou Y, Hong J, Cao M, Cui B, Zhang H, Chen M, Shi J, Ning T, Zhao S, Liu W, Xiong H, Wei C, Qiu Z, Gu W, Zhang Y, Li W, Miao L, Sun Y, Yang M, Wang R, Ma Q, Xu M, Xu Y, Wang T, Chan KHK, Zuo X, Chen H, Qi L, Lai S, Duan S, Song B, Bi Y, Liu S, Wang W, Ning G, Wang J. Rare Loss-of-Function Variants in NPC1 Predispose to Human Obesity. Diabetes 2017; 66:935-947. [PMID: 28130309 DOI: 10.2337/db16-0877] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/16/2017] [Indexed: 11/13/2022]
Abstract
Some Shanghai Clinical Center f a role of Niemann-Pick type C1 (NPC1) for obesity traits. However, whether the loss-of-function mutations in NPC1 cause adiposity in humans remains unknown. We recruited 25 probands with rare autosomal-recessive Niemann-Pick type C (NP-C) disease and their parents in assessment of the effect of heterozygous NPC1 mutations on adiposity. We found that male NPC1+/- carriers had a significantly higher BMI than matched control subjects or the whole population-based control subjects. Consistently, male NPC1+/- mice had increased fat storage while eating a high-fat diet. We further conducted an in-depth assessment of rare variants in the NPC1 gene in young, severely obese subjects and lean control subjects and identified 17 rare nonsynonymous/frameshift variants in NPC1 (minor allele frequency <1%) that were significantly associated with an increased risk of obesity (3.40% vs. 0.73%, respectively, in obese patients and control subjects, P = 0.0008, odds ratio = 4.8, 95% CI 1.7-13.2), indicating that rare NPC1 variants were enriched in young, morbidly obese Chinese subjects. Importantly, participants carrying rare variants with severely damaged cholesterol-transporting ability had more fat accumulation than those with mild/no damage rare variants. In summary, rare loss-of-function NPC1 mutations were identified as being associated with human adiposity with a high penetrance, providing potential therapeutic interventions for obesity in addition to the role of NPC1 in the familial NP-C disease.
Collapse
Affiliation(s)
- Ruixin Liu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yaoyu Zou
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Laboratory of Endocrinology and Metabolism, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jie Hong
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Min Cao
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Bin Cui
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Laboratory of Endocrinology and Metabolism, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Huiwen Zhang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Maopei Chen
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Juan Shi
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Tinglu Ning
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Laboratory of Endocrinology and Metabolism, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Shaoqian Zhao
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Wen Liu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Hui Xiong
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Cuijie Wei
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Zhengqing Qiu
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiqiong Gu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yifei Zhang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Wanyu Li
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Lin Miao
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yingkai Sun
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Minglan Yang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Rui Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Qinyun Ma
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Min Xu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yu Xu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Tiange Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Kei-Hang Katie Chan
- Department of Epidemiology and Center for Global Cardiometabolic Health, School of Public Health, and Department of Medicine (Endocrinology), The Warren Alpert Medical School, Brown University, Providence, RI
| | - Xianbo Zuo
- Institute of Dermatology and Department of Dermatology at No. 1 Hospital, Anhui Medical University, Hefei, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Shenghan Lai
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD
| | - Shumin Duan
- The Institute of Neuroscience, Zhejiang University, Hangzhou, China
| | - Baoliang Song
- College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Yufang Bi
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Simin Liu
- Department of Epidemiology and Center for Global Cardiometabolic Health, School of Public Health, and Department of Medicine (Endocrinology), The Warren Alpert Medical School, Brown University, Providence, RI
| | - Weiqing Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Guang Ning
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Laboratory of Endocrinology and Metabolism, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jiqiu Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, National Key Laboratory for Medical Genomes, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| |
Collapse
|
67
|
Styne DM, Arslanian SA, Connor EL, Farooqi IS, Murad MH, Silverstein JH, Yanovski JA. Pediatric Obesity-Assessment, Treatment, and Prevention: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2017; 102:709-757. [PMID: 28359099 PMCID: PMC6283429 DOI: 10.1210/jc.2016-2573] [Citation(s) in RCA: 688] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023]
Abstract
COSPONSORING ASSOCIATIONS The European Society of Endocrinology and the Pediatric Endocrine Society. This guideline was funded by the Endocrine Society. OBJECTIVE To formulate clinical practice guidelines for the assessment, treatment, and prevention of pediatric obesity. PARTICIPANTS The participants include an Endocrine Society-appointed Task Force of 6 experts, a methodologist, and a medical writer. EVIDENCE This evidence-based guideline was developed using the Grading of Recommendations, Assessment, Development, and Evaluation approach to describe the strength of recommendations and the quality of evidence. The Task Force commissioned 2 systematic reviews and used the best available evidence from other published systematic reviews and individual studies. CONSENSUS PROCESS One group meeting, several conference calls, and e-mail communications enabled consensus. Endocrine Society committees and members and co-sponsoring organizations reviewed and commented on preliminary drafts of this guideline. CONCLUSION Pediatric obesity remains an ongoing serious international health concern affecting ∼17% of US children and adolescents, threatening their adult health and longevity. Pediatric obesity has its basis in genetic susceptibilities influenced by a permissive environment starting in utero and extending through childhood and adolescence. Endocrine etiologies for obesity are rare and usually are accompanied by attenuated growth patterns. Pediatric comorbidities are common and long-term health complications often result; screening for comorbidities of obesity should be applied in a hierarchal, logical manner for early identification before more serious complications result. Genetic screening for rare syndromes is indicated only in the presence of specific historical or physical features. The psychological toll of pediatric obesity on the individual and family necessitates screening for mental health issues and counseling as indicated. The prevention of pediatric obesity by promoting healthful diet, activity, and environment should be a primary goal, as achieving effective, long-lasting results with lifestyle modification once obesity occurs is difficult. Although some behavioral and pharmacotherapy studies report modest success, additional research into accessible and effective methods for preventing and treating pediatric obesity is needed. The use of weight loss medications during childhood and adolescence should be restricted to clinical trials. Increasing evidence demonstrates the effectiveness of bariatric surgery in the most seriously affected mature teenagers who have failed lifestyle modification, but the use of surgery requires experienced teams with resources for long-term follow-up. Adolescents undergoing lifestyle therapy, medication regimens, or bariatric surgery for obesity will need cohesive planning to help them effectively transition to adult care, with continued necessary monitoring, support, and intervention. Transition programs for obesity are an uncharted area requiring further research for efficacy. Despite a significant increase in research on pediatric obesity since the initial publication of these guidelines 8 years ago, further study is needed of the genetic and biological factors that increase the risk of weight gain and influence the response to therapeutic interventions. Also needed are more studies to better understand the genetic and biological factors that cause an obese individual to manifest one comorbidity vs another or to be free of comorbidities. Furthermore, continued investigation into the most effective methods of preventing and treating obesity and into methods for changing environmental and economic factors that will lead to worldwide cultural changes in diet and activity should be priorities. Particular attention to determining ways to effect systemic changes in food environments and total daily mobility, as well as methods for sustaining healthy body mass index changes, is of importance.
Collapse
Affiliation(s)
- Dennis M Styne
- University of California Davis, Sacramento, California 95817
| | | | | | | | | | | | | |
Collapse
|
68
|
Burnett LC, LeDuc CA, Sulsona CR, Paull D, Rausch R, Eddiry S, Carli JFM, Morabito MV, Skowronski AA, Hubner G, Zimmer M, Wang L, Day R, Levy B, Fennoy I, Dubern B, Poitou C, Clement K, Butler MG, Rosenbaum M, Salles JP, Tauber M, Driscoll DJ, Egli D, Leibel RL. Deficiency in prohormone convertase PC1 impairs prohormone processing in Prader-Willi syndrome. J Clin Invest 2017; 127:293-305. [PMID: 27941249 PMCID: PMC5199710 DOI: 10.1172/jci88648] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/20/2016] [Indexed: 12/17/2022] Open
Abstract
Prader-Willi syndrome (PWS) is caused by a loss of paternally expressed genes in an imprinted region of chromosome 15q. Among the canonical PWS phenotypes are hyperphagic obesity, central hypogonadism, and low growth hormone (GH). Rare microdeletions in PWS patients define a 91-kb minimum critical deletion region encompassing 3 genes, including the noncoding RNA gene SNORD116. Here, we found that protein and transcript levels of nescient helix loop helix 2 (NHLH2) and the prohormone convertase PC1 (encoded by PCSK1) were reduced in PWS patient induced pluripotent stem cell-derived (iPSC-derived) neurons. Moreover, Nhlh2 and Pcsk1 expression were reduced in hypothalami of fasted Snord116 paternal knockout (Snord116p-/m+) mice. Hypothalamic Agrp and Npy remained elevated following refeeding in association with relative hyperphagia in Snord116p-/m+ mice. Nhlh2-deficient mice display growth deficiencies as adolescents and hypogonadism, hyperphagia, and obesity as adults. Nhlh2 has also been shown to promote Pcsk1 expression. Humans and mice deficient in PC1 display hyperphagic obesity, hypogonadism, decreased GH, and hypoinsulinemic diabetes due to impaired prohormone processing. Here, we found that Snord116p-/m+ mice displayed in vivo functional defects in prohormone processing of proinsulin, pro-GH-releasing hormone, and proghrelin in association with reductions in islet, hypothalamic, and stomach PC1 content. Our findings suggest that the major neuroendocrine features of PWS are due to PC1 deficiency.
Collapse
Affiliation(s)
- Lisa C. Burnett
- Institute of Human Nutrition
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Charles A. LeDuc
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- New York Obesity Research Center, New York, New York, USA
| | - Carlos R. Sulsona
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida College of Medicine Gainesville, Florida, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Richard Rausch
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Sanaa Eddiry
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, CNRS UMR 5282, INSERM UMR 1043, Université Paul Sabatier, Toulouse, France
| | - Jayne F. Martin Carli
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | - Michael V. Morabito
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Alicja A. Skowronski
- Institute of Human Nutrition
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | | | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Liheng Wang
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Robert Day
- Institut de pharmacologie de Sherbrooke, Department of Surgery, Division of Urology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Brynn Levy
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Ilene Fennoy
- Department of Pediatrics, Division of Pediatric Diabetes, Endocrinology and Metabolism, Columbia University, New York, New York, USA
| | - Beatrice Dubern
- Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Sorbonne University, University Pierre et Marie-Curie, INSERM UMRS 1166, Paris, France
| | - Christine Poitou
- Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Sorbonne University, University Pierre et Marie-Curie, INSERM UMRS 1166, Paris, France
| | - Karine Clement
- Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Sorbonne University, University Pierre et Marie-Curie, INSERM UMRS 1166, Paris, France
| | - Merlin G. Butler
- Department of Psychiatry and Behavioral Sciences, Division of Research and Genetics, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Michael Rosenbaum
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Jean Pierre Salles
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, CNRS UMR 5282, INSERM UMR 1043, Université Paul Sabatier, Toulouse, France
- Unité d’Endocrinologie, Hôpital des Enfants, and
| | - Maithe Tauber
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, CNRS UMR 5282, INSERM UMR 1043, Université Paul Sabatier, Toulouse, France
- Unité d’Endocrinologie, Hôpital des Enfants, and
- Centre de Référence du Syndrome de Prader-Willi, CHU Toulouse, Toulouse, France
| | - Daniel J. Driscoll
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida College of Medicine Gainesville, Florida, USA
- Center for Epigenetics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Dieter Egli
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Rudolph L. Leibel
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- New York Obesity Research Center, New York, New York, USA
| |
Collapse
|
69
|
Ayuso M, Fernández A, Núñez Y, Benítez R, Isabel B, Fernández AI, Rey AI, González-Bulnes A, Medrano JF, Cánovas Á, López-Bote CJ, Óvilo C. Developmental Stage, Muscle and Genetic Type Modify Muscle Transcriptome in Pigs: Effects on Gene Expression and Regulatory Factors Involved in Growth and Metabolism. PLoS One 2016; 11:e0167858. [PMID: 27936208 PMCID: PMC5148031 DOI: 10.1371/journal.pone.0167858] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/21/2016] [Indexed: 01/08/2023] Open
Abstract
Iberian pig production includes purebred (IB) and Duroc-crossbred (IBxDU) pigs, which show important differences in growth, fattening and tissue composition. This experiment was conducted to investigate the effects of genetic type and muscle (Longissimus dorsi (LD) vs Biceps femoris (BF)) on gene expression and transcriptional regulation at two developmental stages. Nine IB and 10 IBxDU piglets were slaughtered at birth, and seven IB and 10 IBxDU at four months of age (growing period). Carcass traits and LD intramuscular fat (IMF) content were measured. Muscle transcriptome was analyzed on LD samples with RNA-Seq technology. Carcasses were smaller in IB than in IBxDU neonates (p < 0.001), while growing IB pigs showed greater IMF content (p < 0.05). Gene expression was affected (p < 0.01 and Fold change > 1.5) by the developmental stage (5,812 genes), muscle type (135 genes), and genetic type (261 genes at birth and 113 at growth). Newborns transcriptome reflected a highly proliferative developmental stage, while older pigs showed upregulation of catabolic and muscle functioning processes. Regarding the genetic type effect, IBxDU newborns showed enrichment of gene pathways involved in muscle growth, in agreement with the higher prenatal growth observed in these pigs. However, IB growing pigs showed enrichment of pathways involved in protein deposition and cellular growth, supporting the compensatory gain experienced by IB pigs during this period. Moreover, newborn and growing IB pigs showed more active glucose and lipid metabolism than IBxDU pigs. Moreover, LD muscle seems to have more active muscular and cell growth, while BF points towards lipid metabolism and fat deposition. Several regulators controlling transcriptome changes in both genotypes were identified across muscles and ages (SIM1, PVALB, MEFs, TCF7L2 or FOXO1), being strong candidate genes to drive expression and thus, phenotypic differences between IB and IBxDU pigs. Many of the identified regulators were known to be involved in muscle and adipose tissues development, but others not previously associated with pig muscle growth were also identified, as PVALB, KLF1 or IRF2. The present study discloses potential molecular mechanisms underlying phenotypic differences observed between IB and IBxDU pigs and highlights candidate genes implicated in these molecular mechanisms.
Collapse
Affiliation(s)
- Miriam Ayuso
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Yolanda Núñez
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| | - Rita Benítez
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| | - Beatriz Isabel
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Ana I. Rey
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Juan F. Medrano
- Department of Animal Science, University of California Davis, Davis, California, United States of America
| | - Ángela Cánovas
- Department of Animal Science, University of California Davis, Davis, California, United States of America
| | - Clemente J. López-Bote
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| |
Collapse
|
70
|
Wu D, Su X, Potluri N, Kim Y, Rastinejad F. NPAS1-ARNT and NPAS3-ARNT crystal structures implicate the bHLH-PAS family as multi-ligand binding transcription factors. eLife 2016; 5. [PMID: 27782878 PMCID: PMC5111884 DOI: 10.7554/elife.18790] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023] Open
Abstract
The neuronal PAS domain proteins NPAS1 and NPAS3 are members of the basic helix-loop-helix-PER-ARNT-SIM (bHLH-PAS) family, and their genetic deficiencies are linked to a variety of human psychiatric disorders including schizophrenia, autism spectrum disorders and bipolar disease. NPAS1 and NPAS3 must each heterodimerize with the aryl hydrocarbon receptor nuclear translocator (ARNT), to form functional transcription complexes capable of DNA binding and gene regulation. Here we examined the crystal structures of multi-domain NPAS1-ARNT and NPAS3-ARNT-DNA complexes, discovering each to contain four putative ligand-binding pockets. Through expanded architectural comparisons between these complexes and HIF-1α-ARNT, HIF-2α-ARNT and CLOCK-BMAL1, we show the wider mammalian bHLH-PAS family is capable of multi-ligand-binding and presents as an ideal class of transcription factors for direct targeting by small-molecule drugs. DOI:http://dx.doi.org/10.7554/eLife.18790.001 Transcription factors are proteins that can bind to DNA to regulate the activity of genes. One family of transcription factors in mammals is known as the bHLH-PAS family, which consists of sixteen members including NPAS1 and NPAS3. These two proteins are both found in nerve cells, and genetic mutations that affect NPAS1 or NPAS3 have been linked to psychiatric conditions in humans. Therefore, researchers would like to discover new drugs that can bind to these proteins and control their activities in nerve cells. Understanding the three-dimensional structure of a protein can aid the discovery of small molecules that can bind to these proteins and act as drugs. Proteins in the bHLH-PAS family have to form pairs in order to bind to DNA: NPAS1 and NPAS3 both interact with another bHLH-PAS protein called ARNT, but it is not clear exactly how this works. In 2015, a team of researchers described the shapes that ARNT adopts when it forms pairs with two other bHLH-PAS proteins that are important for sensing when oxygen levels drop in cells. Here, Wu et al. – including many of the researchers involved in the earlier work – have used a technique called X-ray crystallography to determine the three-dimensional shapes of NPAS1 when it is bound to ARNT, and NPAS3 when it is bound to both ARNT and DNA. The experiments show that each of these structures contains four distinct pockets that certain small molecules might be able to bind to. The NPAS1 and NPAS3 structures are similar to each other and to the previously discovered bHLH-PAS structures involved in oxygen sensing. Further analysis of other bHLH-PAS proteins suggests that all the members of this protein family are likely to be able to bind to small molecules and should therefore be considered as potential drug targets. The next step following on from this work is to identify small molecules that bind to bHLH-PAS proteins, which will help to reveal the genes that are regulated by this family. In the future, these small molecules may have the potential to be developed into new drugs to treat psychiatric conditions and other diseases in humans. DOI:http://dx.doi.org/10.7554/eLife.18790.002
Collapse
Affiliation(s)
- Dalei Wu
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, United States
| | - Xiaoyu Su
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, United States
| | - Nalini Potluri
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, United States
| | - Youngchang Kim
- Structural Biology Center, Biosciences Division, Argonne National Laboratory, Argonne, United States
| | - Fraydoon Rastinejad
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, United States
| |
Collapse
|
71
|
Wu D, Rastinejad F. Structural characterization of mammalian bHLH-PAS transcription factors. Curr Opin Struct Biol 2016; 43:1-9. [PMID: 27721191 DOI: 10.1016/j.sbi.2016.09.011] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/06/2016] [Accepted: 09/21/2016] [Indexed: 11/28/2022]
Abstract
The mammalian basic helix-loop-helix-PER-ARNT-SIM (bHLH-PAS) transcription factors share common architectural features that include a bHLH DNA-binding domain and tandemly positioned PAS domains. The sixteen members of this family include the hypoxia-inducible factors (HIF-1α and HIF-2α), ARNT (also known as HIF-1β), CLOCK and BMAL1. Most bHLH-PAS proteins have been genetically linked to variety of diseases in humans, including cancers, metabolic syndromes and psychiatric conditions. To function as transcription factors, the bHLH-PAS proteins must form heterodimeric complexes. Recent crystallographic studies of HIF-α-ARNT and CLOCK-BMAL1 complexes have unveiled the organization of their multi-domain bHLH-PAS-A-PAS-B segments, revealing how these architectures can give rise to unique patterns of heterodimerization. As our structural understanding becomes better integrated with ligand-discovery and target gene identification, a more comprehensive picture of their architectural and functional properties will emerge.
Collapse
Affiliation(s)
- Dalei Wu
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA; State Key Laboratory of Microbial Technology, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Fraydoon Rastinejad
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA.
| |
Collapse
|
72
|
Identification of mutations through dominant screening for obesity using C57BL/6 substrains. Sci Rep 2016; 6:32453. [PMID: 27585985 PMCID: PMC5009433 DOI: 10.1038/srep32453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/10/2016] [Indexed: 01/05/2023] Open
Abstract
The discovery of leptin substantiated the usefulness of a forward genetic approach in elucidating the molecular network regulating energy metabolism. However, no successful dominant screening for obesity has been reported, which may be due to the influence of quantitative trait loci between the screening and counter strains and the low fertility of obese mice. Here, we performed a dominant screening for obesity using C57BL/6 substrains, C57BL/6J and C57BL/6N, with the routine use of in vitro fertilization. The screening of more than 5000 mutagenized mice established two obese pedigrees in which single nucleotide substitutions in Mc4r and Sim1 genes were identified through whole-exome sequencing. The mutation in the Mc4r gene produces a premature stop codon, and the mutant SIM1 protein lacks transcriptional activity, showing that the haploinsufficiency of SIM1 and MC4R results in obesity. We further examined the hypothalamic neuropeptide expressions in the mutant pedigrees and mice with diet-induced obesity, which showed that each obesity mouse model has distinct neuropeptide expression profiles. This forward genetic screening scheme is useful and applicable to any research field in which mouse models work.
Collapse
|
73
|
Cheon CK. Genetics of Prader-Willi syndrome and Prader-Will-Like syndrome. Ann Pediatr Endocrinol Metab 2016; 21:126-135. [PMID: 27777904 PMCID: PMC5073158 DOI: 10.6065/apem.2016.21.3.126] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 11/29/2022] Open
Abstract
The Prader-Willi syndrome (PWS) is a human imprinting disorder resulting from genomic alterations that inactivate imprinted, paternally expressed genes in human chromosome region 15q11-q13. This genetic condition appears to be a contiguous gene syndrome caused by the loss of at least 2 of a number of genes expressed exclusively from the paternal allele, including SNRPN, MKRN3, MAGEL2, NDN and several snoRNAs, but it is not yet well known which specific genes in this region are associated with this syndrome. Prader-Will-Like syndrome (PWLS) share features of the PWS phenotype and the gene functions disrupted in PWLS are likely to lie in genetic pathways that are important for the development of PWS phenotype. However, the genetic basis of these rare disorders differs and the absence of a correct diagnosis may worsen the prognosis of these individuals due to the endocrine-metabolic malfunctioning associated with the PWS. Therefore, clinicians face a challenge in determining when to request the specific molecular test used to identify patients with classical PWS because the signs and symptoms of PWS are common to other syndromes such as PWLS. This review aims to provide an overview of current knowledge relating to the genetics of PWS and PWLS, with an emphasis on identification of patients that may benefit from further investigation and genetic screening.
Collapse
Affiliation(s)
- Chong Kun Cheon
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
74
|
Gao Y, Sun T. Molecular regulation of hypothalamic development and physiological functions. Mol Neurobiol 2016; 53:4275-85. [PMID: 26223804 PMCID: PMC4733441 DOI: 10.1007/s12035-015-9367-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/17/2015] [Indexed: 01/08/2023]
Abstract
The hypothalamus is composed of many heterogeneous nuclei that control distinct physiological functions. Investigating molecular mechanisms that regulate the specification of these nuclei and specific neuronal subtypes, and their contribution to diverse hypothalamic functions, is an exciting research focus. Here, we begin by summarizing the hypothalamic functions of feeding regulation, sleep-wake cycles, stress responses, and circadian rhythm, and describing their anatomical bases. Next, we review the molecular regulation of formation of hypothalamic territories, specification of nuclei and subnuclei, and generation of specific neurons. Finally, we highlight physiological and behavioral consequences of altered hypothalamic development. Identifying molecules that regulate hypothalamic development and function will increase our understanding of hypothalamus-related disorders, such as obesity and diabetes, and aid in the development of therapies aimed specifically at their etiologies.
Collapse
Affiliation(s)
- Yanxia Gao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Tao Sun
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, Box 60, New York, NY, 10065, USA.
| |
Collapse
|
75
|
Spikol ED, Laverriere CE, Robnett M, Carter G, Wolfe E, Glasgow E. Zebrafish Models of Prader-Willi Syndrome: Fast Track to Pharmacotherapeutics. Diseases 2016; 4. [PMID: 27857842 PMCID: PMC5110251 DOI: 10.3390/diseases4010013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a rare genetic neurodevelopmental disorder characterized by an insatiable appetite, leading to chronic overeating and obesity. Additional features include short stature, intellectual disability, behavioral problems and incomplete sexual development. Although significant progress has been made in understanding the genetic basis of PWS, the mechanisms underlying the pathogenesis of the disorder remain poorly understood. Treatment for PWS consists mainly of palliative therapies; curative therapies are sorely needed. Zebrafish, Danio rerio, represent a promising way forward for elucidating physiological problems such as obesity and identifying new pharmacotherapeutic options for PWS. Over the last decade, an increased appreciation for the highly conserved biology among vertebrates and the ability to perform high-throughput drug screening has seen an explosion in the use of zebrafish for disease modeling and drug discovery. Here, we review recent advances in developing zebrafish models of human disease. Aspects of zebrafish genetics and physiology that are relevant to PWS will be discussed, and the advantages and disadvantages of zebrafish models will be contrasted with current animal models for this syndrome. Finally, we will present a paradigm for drug screening in zebrafish that is potentially the fastest route for identifying and delivering curative pharmacotherapies to PWS patients.
Collapse
|
76
|
Geets E, Zegers D, Beckers S, Verrijken A, Massa G, Van Hoorenbeeck K, Verhulst S, Van Gaal L, Van Hul W. Copy number variation (CNV) analysis and mutation analysis of the 6q14.1-6q16.3 genes SIM1 and MRAP2 in Prader Willi like patients. Mol Genet Metab 2016; 117:383-8. [PMID: 26795956 DOI: 10.1016/j.ymgme.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/07/2016] [Accepted: 01/07/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Prader-Willi syndrome (PWS), caused by a paternal defect on 15q11.2-q13, is the most common form of syndromic obesity. However, patients clinically diagnosed with PWS do not always show this defect on chromosome 15q and are therefore molecularly categorized as Prader Willi like (PWL). Deletions at 6q14.1-q16.3 encompassing MRAP2 and SIM1 were reported in some individuals with a PWL phenotype. In addition, a few mutations in SIM1 and MRAP2 were also previously identified in cohorts of obese individuals. Therefore, we decided to perform copy number variation analysis of the 6q14.1-6q16.3 region followed by mutation analysis of SIM1 and MRAP2 in a PWL cohort. METHODS A genome-wide microarray analysis was performed in a group of 109 PWL patients. Next, we screened 94 PWL patients for mutations in SIM1 and MRAP2 using high-resolution melting curve analysis and Sanger sequencing. Additionally, 363 obese children and adolescents were screened for mutations in MRAP2. RESULTS No gene harboring deletions were identified at the 6q14.1-q16.3 region in the 109 PWL patients. SIM1 mutation analysis resulted in the identification of one very rare nonsynonymous variant p.P352S (rs3734354). Another rare nonsynonymous variant, p.A40S, was detected in the MRAP2 gene. No variants were identified in the 363 obese individuals. CONCLUSIONS In contrast to literature reports, no gene harboring deletions were identified in the SIM1 and MRAP2 regions in our PWL cohort. Secondly, taking into account their very low minor allele frequencies in public sequencing databases and the results of in silico prediction programs, further functional analysis of p.P352S found in SIM1 and p.A40S found in MRAP2 is useful. This would provide further support for a possible role of SIM1 and MRAP2 in the pathogenesis of the PWL phenotype albeit in a limited number of patients.
Collapse
Affiliation(s)
- Ellen Geets
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Doreen Zegers
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Sigri Beckers
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, Belgium
| | - Guy Massa
- Department of Pediatrics, Jessa Hospital, Hasselt, Belgium
| | | | - Stijn Verhulst
- Department of Pediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Luc Van Gaal
- Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Antwerp, Belgium
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
77
|
Small 6q16.1 Deletions Encompassing POU3F2 Cause Susceptibility to Obesity and Variable Developmental Delay with Intellectual Disability. Am J Hum Genet 2016; 98:363-72. [PMID: 26833329 DOI: 10.1016/j.ajhg.2015.12.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/15/2015] [Indexed: 12/22/2022] Open
Abstract
Genetic studies of intellectual disability and identification of monogenic causes of obesity in humans have made immense contribution toward the understanding of the brain and control of body mass. The leptin > melanocortin > SIM1 pathway is dysregulated in multiple monogenic human obesity syndromes but its downstream targets are still unknown. In ten individuals from six families, with overlapping 6q16.1 deletions, we describe a disorder of variable developmental delay, intellectual disability, and susceptibility to obesity and hyperphagia. The 6q16.1 deletions segregated with the phenotype in multiplex families and were shown to be de novo in four families, and there was dramatic phenotypic overlap among affected individuals who were independently ascertained without bias from clinical features. Analysis of the deletions revealed a ∼350 kb critical region on chromosome 6q16.1 that encompasses a gene for proneuronal transcription factor POU3F2, which is important for hypothalamic development and function. Using morpholino and mutant zebrafish models, we show that POU3F2 lies downstream of SIM1 and controls oxytocin expression in the hypothalamic neuroendocrine preoptic area. We show that this finding is consistent with the expression patterns of POU3F2 and related genes in the human brain. Our work helps to further delineate the neuro-endocrine control of energy balance/body mass and demonstrates that this molecular pathway is conserved across multiple species.
Collapse
|
78
|
Saeed S, Bonnefond A, Manzoor J, Shabbir F, Ayesha H, Philippe J, Durand E, Crouch H, Sand O, Ali M, Butt T, Rathore AW, Falchi M, Arslan M, Froguel P. Genetic variants in LEP, LEPR, and MC4R explain 30% of severe obesity in children from a consanguineous population. Obesity (Silver Spring) 2015; 23:1687-95. [PMID: 26179253 DOI: 10.1002/oby.21142] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 03/24/2015] [Accepted: 04/10/2015] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Single gene mutations leading to severe obesity have so far been identified in 3-5% cases in European populations. However, prevalence of these pathogenic mutations has not systematically been examined in specific consanguineous populations. Here we describe the incidence of obesity-associated mutations through a step-wise sequence analysis, in a cohort of 73 Pakistani children with severe obesity from consanguineous families. METHODS Initially, all subjects were screened for mutations in coding regions of leptin (LEP) and melanocortin 4 receptor (MC4R) genes by direct sequencing. Subjects negative for mutation in these genes were screened using microdroplet PCR enrichment and NGS. Genomic structural variation was assessed by genotyping. Serum leptin, insulin, and cortisol were determined by ELISA. RESULTS Among 73 children with severe obesity (BMI SDS > 3.0), we identified 22 probands and 5 relatives, carrying 10 different loss-of-function homozygous mutations in LEP, leptin receptor (LEPR), and MC4R genes, including 4 novel variants. Hypercortisolemia was significantly emphasized in LEP mutation carriers. CONCLUSIONS The prevalence of pathogenic mutations in genes known to directly influence leptin-melanocortin signaling is 30% in our cohort. The results of this study emphasize the desirability of undertaking systematic and in-depth genetic analysis of cases with severe obesity in specific consanguineous populations.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Amélie Bonnefond
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Jaida Manzoor
- Department of Paediatric Endocrinology, Children's Hospital, Lahore, Pakistan
| | - Faiza Shabbir
- Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
| | - Hina Ayesha
- Department of Paediatrics, Punjab Medical College, Faisalabad, Pakistan
| | - Julien Philippe
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Emmanuelle Durand
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Hutokshi Crouch
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Olivier Sand
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Muhammad Ali
- Department of Paediatrics, Mayo Hospital, King Edward Medical University, Lahore, Pakistan
| | - Taeed Butt
- Department of Paediatrics, Fatima Memorial Hospital, Lahore, Pakistan
| | - Ahsan W Rathore
- Department of Paediatric Endocrinology, Children's Hospital, Lahore, Pakistan
| | - Mario Falchi
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Muhammad Arslan
- Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
- Centre for Research in Molecular Medicine, the University of Lahore, Lahore, Pakistan
| | - Philippe Froguel
- Department of Genomics of Common Disease, Imperial College London, London, UK
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| |
Collapse
|
79
|
Abstract
The global rise in the prevalence of obesity and associated co-morbidities such as type 2 diabetes, cardiovascular disease, and cancer represents a major public health concern. The biological response to increased consumption of palatable foods or a reduction in energy expenditure is highly variable between individuals. A more detailed mechanistic understanding of the molecular, physiological, and behavioral pathways involved in the development of obesity in susceptible individuals is critical for identifying effective mechanism-based preventative and therapeutic interventions.
Collapse
Affiliation(s)
- Agatha A van der Klaauw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| |
Collapse
|
80
|
Yazdi FT, Clee SM, Meyre D. Obesity genetics in mouse and human: back and forth, and back again. PeerJ 2015; 3:e856. [PMID: 25825681 PMCID: PMC4375971 DOI: 10.7717/peerj.856] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 03/05/2015] [Indexed: 12/19/2022] Open
Abstract
Obesity is a major public health concern. This condition results from a constant and complex interplay between predisposing genes and environmental stimuli. Current attempts to manage obesity have been moderately effective and a better understanding of the etiology of obesity is required for the development of more successful and personalized prevention and treatment options. To that effect, mouse models have been an essential tool in expanding our understanding of obesity, due to the availability of their complete genome sequence, genetically identified and defined strains, various tools for genetic manipulation and the accessibility of target tissues for obesity that are not easily attainable from humans. Our knowledge of monogenic obesity in humans greatly benefited from the mouse obesity genetics field. Genes underlying highly penetrant forms of monogenic obesity are part of the leptin-melanocortin pathway in the hypothalamus. Recently, hypothesis-generating genome-wide association studies for polygenic obesity traits in humans have led to the identification of 119 common gene variants with modest effect, most of them having an unknown function. These discoveries have led to novel animal models and have illuminated new biologic pathways. Integrated mouse-human genetic approaches have firmly established new obesity candidate genes. Innovative strategies recently developed by scientists are described in this review to accelerate the identification of causal genes and deepen our understanding of obesity etiology. An exhaustive dissection of the molecular roots of obesity may ultimately help to tackle the growing obesity epidemic worldwide.
Collapse
Affiliation(s)
- Fereshteh T. Yazdi
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Susanne M. Clee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
81
|
Abstract
Type 2 diabetes (T2D) had long been referred to as the "geneticist's nightmare." Genome-wide association studies have fully confirmed the polygenic nature of T2D, demonstrating the role of many genes in T2D risk. The increasingly busier picture of T2D genetics is quite difficult to understand for the diabetes research community, which can create misunderstandings with geneticists, and can eventually limit both basic research and translational outcomes of these genetic discoveries. The present review wishes to lift the fog around genetics of T2D with the hope that it will foster integrated diabetes modeling approaches from genetic defects to personalized medicine.
Collapse
Affiliation(s)
- Amélie Bonnefond
- CNRS-UMR8199, Lille Pasteur Institute, Lille 59000, France; Lille University, Lille 59000, France; European Genomic Institute for Diabetes (EGID), Lille 59000, France
| | - Philippe Froguel
- CNRS-UMR8199, Lille Pasteur Institute, Lille 59000, France; Lille University, Lille 59000, France; European Genomic Institute for Diabetes (EGID), Lille 59000, France; Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London W12 0NN, UK.
| |
Collapse
|
82
|
|
83
|
Altirriba J, Poher AL, Rohner-Jeanrenaud F. Chronic Oxytocin Administration as a Treatment Against Impaired Leptin Signaling or Leptin Resistance in Obesity. Front Endocrinol (Lausanne) 2015; 6:119. [PMID: 26300847 PMCID: PMC4525065 DOI: 10.3389/fendo.2015.00119] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/22/2015] [Indexed: 12/31/2022] Open
Abstract
This review summarizes the existing literature on the effects of oxytocin administration in the treatment of obesity in different animal models and in humans, focusing on the central control of food intake, the oxytocin effects on adipose tissue, and the relationships between oxytocin and leptin. Oxytocin is a hypothalamic nonapeptide synthesized mainly in the paraventricular and supraoptic nuclei projecting to the pituitary, where it reaches the peripheral circulation, as well as to other brain regions. Moreover, leptin modulates oxytocin levels and activates oxytocin neurons in the hypothalamic paraventricular nucleus, which innervates the nucleus of the solitary tract, partly responsible for the brain-elicited oxytocin effects. Taking into account that oxytocin is located downstream leptin, it was hypothesized that oxytocin treatment would be effective in decreasing body weight in leptin-resistant DIO animals, as well as in those with leptin or with leptin receptor deficiency. Several groups have demonstrated that in such animal models (rats, mice, and rhesus monkeys), central or peripheral oxytocin administration decreases body weight, mainly due to a decrease in fat mass, demonstrating that an oxytocin treatment is able to partly overcome leptin deficiency or resistance. Moreover, a pilot clinical study demonstrated the efficiency of oxytocin in the treatment of obesity in human subjects, confirming the results obtained in the different animal models. Larger multicenter studies are now needed to determine whether the beneficial effects of oxytocin treatment can apply not only to obese but also to type 2 diabetic patients. These studies should also shed some light on the molecular mechanisms of oxytocin action in humans.
Collapse
Affiliation(s)
- Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- *Correspondence: Jordi Altirriba, Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, 1, rue Michel-Servet, Geneva CH-1211, Switzerland,
| | - Anne-Laure Poher
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Françoise Rohner-Jeanrenaud
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
84
|
Montagne L, Raimondo A, Delobel B, Duban-Bedu B, Noblet FS, Dechaume A, Bersten DC, Meyre D, Whitelaw ML, Froguel P, Bonnefond A. Identification of two novel loss-of-function SIM1 mutations in two overweight children with developmental delay. Obesity (Silver Spring) 2014; 22:2621-4. [PMID: 25234154 DOI: 10.1002/oby.20886] [Citation(s) in RCA: 21] [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: 07/26/2014] [Accepted: 08/14/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Several deletions of chromosome 6q, including SIM1, were reported in obese patients with developmental delay. Furthermore, rare loss-of-function SIM1 mutations were shown to contribute to severe obesity, yet the role of these mutations in developmental delay remained unclear. Here, SIM1 in children with neurodevelopmental abnormalities was screened and the functional effect of the identified mutations was investigated. METHODS SIM1 was sequenced in 283 children presenting with developmental delay and at least overweight. The effect of the identified mutations on SIM1 transcriptional activity in stable human cell lines was assessed using luciferase gene reporter assays. RESULTS Two novel mutations (c.886A>G/p.R296G and c.925A>G/p.S309G) in two boys with variable degrees of cognitive deficits and weight issues were identified. The child mutated for p.R296G presented with a generally more severe phenotype than the p.S309G carrier (obesity, compulsive eating, neonatal hypotonia versus overweight only), while both mutations had strong loss-of-function effects on SIM1 transcriptional activity. CONCLUSIONS Severe loss-of-function SIM1 mutations can be associated with a spectrum of developmental delay phenotypes and obesity. Our data suggest that SIM1 sequencing should be performed more systematically in patients with developmental delay, even in the absence of severe obesity. These results deserve further SIM1 screening studies.
Collapse
Affiliation(s)
- Louise Montagne
- European Genomic Institute for Diabetes, Lille, France. Correspondence: Philippe Froguel ; CNRS-UMR8199, Lille Pasteur Institute, Lille, France; Lille 2 University, Lille, France; Department of Pediatrics, Saint Antoine Pediatric Hospital, Saint Vincent de Paul Hospital, Catholic University of Lille, Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Incomplete penetrance and phenotypic variability of 6q16 deletions including SIM1. Eur J Hum Genet 2014; 23:1010-8. [PMID: 25351778 DOI: 10.1038/ejhg.2014.230] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/12/2014] [Accepted: 09/16/2014] [Indexed: 11/08/2022] Open
Abstract
6q16 deletions have been described in patients with a Prader-Willi-like (PWS-like) phenotype. Recent studies have shown that certain rare single-minded 1 (SIM1) loss-of-function variants were associated with a high intra-familial risk for obesity with or without features of PWS-like syndrome. Although SIM1 seems to have a key role in the phenotype of patients carrying 6q16 deletions, some data support a contribution of other genes, such as GRIK2, to explain associated behavioural problems. We describe 15 new patients in whom de novo 6q16 deletions were characterised by comparative genomic hybridisation or single-nucleotide polymorphism (SNP) array analysis, including the first patient with fetopathological data. This fetus showed dysmorphic facial features, cerebellar and cerebral migration defects with neuronal heterotopias, and fusion of brain nuclei. The size of the deletion in the 14 living patients ranged from 1.73 to 7.84 Mb, and the fetus had the largest deletion (14 Mb). Genotype-phenotype correlations confirmed the major role for SIM1 haploinsufficiency in obesity and the PWS-like phenotype. Nevertheless, only 8 of 13 patients with SIM1 deletion exhibited obesity, in agreement with incomplete penetrance of SIM1 haploinsufficiency. This study in the largest series reported to date confirms that the PWS-like phenotype is strongly linked to 6q16.2q16.3 deletions and varies considerably in its clinical expression. The possible involvement of other genes in the 6q16.2q16.3-deletion phenotype is discussed.
Collapse
|
86
|
Abstract
The heritability of obesity has long been appreciated and the genetics of obesity has been the focus of intensive study for decades. Early studies elucidating genetic factors involved in rare monogenic and syndromic forms of extreme obesity focused attention on dysfunction of hypothalamic leptin-related pathways in the control of food intake as a major contributor. Subsequent genome-wide association studies of common genetic variants identified novel loci that are involved in more common forms of obesity across populations of diverse ethnicities and ages. The subsequent search for factors contributing to the heritability of obesity not explained by these 2 approaches ("missing heritability") has revealed additional rare variants, copy number variants, and epigenetic changes that contribute. Although clinical applications of these findings have been limited to date, the increasing understanding of the interplay of these genetic factors with environmental conditions, such as the increased availability of high calorie foods and decreased energy expenditure of sedentary lifestyles, promises to accelerate the translation of genetic findings into more successful preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Jill Waalen
- The Scripps Research Institute and the Scripps Translational Science Institute, La Jolla, California.
| |
Collapse
|
87
|
Characterization of human variants in obesity-related SIM1 protein identifies a hot-spot for dimerization with the partner protein ARNT2. Biochem J 2014; 461:403-12. [DOI: 10.1042/bj20131618] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Several non-synonymous variants found in obese patients disrupt function of the transcription factor SIM1 (single-minded 1) by impairing binding to an essential partner protein. The clustering of these variants reveals a mutational hot-spot critical for function of SIM1 and the related protein SIM2.
Collapse
|
88
|
Tolson KP, Gemelli T, Meyer D, Yazdani U, Kozlitina J, Zinn AR. Inducible neuronal inactivation of Sim1 in adult mice causes hyperphagic obesity. Endocrinology 2014; 155:2436-44. [PMID: 24773343 PMCID: PMC4060186 DOI: 10.1210/en.2013-2125] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Germline haploinsufficiency of human or mouse Sim1 is associated with hyperphagic obesity. Sim1 encodes a transcription factor required for proper formation of the paraventricular (PVN), supraoptic, and anterior periventricular hypothalamic nuclei. Sim1 expression persists in these neurons in adult mice, raising the question of whether it plays a physiologic role in regulation of energy balance. We previously showed that Sim1 heterozygous mice had normal numbers of PVN neurons that were hyporesponsive to melanocortin 4 receptor agonism and showed reduced oxytocin expression. Furthermore, conditional postnatal neuronal inactivation of Sim1 also caused hyperphagic obesity and decreased hypothalamic oxytocin expression. PVN projections to the hindbrain, where oxytocin is thought to act to modulate satiety, were anatomically intact in both Sim1 heterozygous and conditional knockout mice. These experiments provided evidence that Sim1 functions in energy balance apart from its role in hypothalamic development but did not rule out effects of Sim1 deficiency on postnatal hypothalamic maturation. To address this possibility, we used a tamoxifen-inducible, neural-specific Cre transgene to conditionally inactivate Sim1 in adult mice with mature hypothalamic circuitry. Induced Sim1 inactivation caused increased food and water intake and decreased expression of PVN neuropeptides, especially oxytocin and vasopressin, with no change in energy expenditure. Sim1 expression was not required for survival of PVN neurons. The results corroborate previous evidence that Sim1 acts physiologically as well as developmentally to regulate body weight. Inducible knockout mice provide a system for studying Sim1's physiologic function in energy balance and identifying its relevant transcriptional targets in the hypothalamus.
Collapse
Affiliation(s)
- Kristen P Tolson
- McDermott Center for Human Growth and Development (K.P.T., T.G., D.M., U.Y., J.K., A.R.Z.) and Department of Internal Medicine (A.R.Z.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8591
| | | | | | | | | | | |
Collapse
|
89
|
Philippe J, Stijnen P, Meyre D, De Graeve F, Thuillier D, Delplanque J, Gyapay G, Sand O, Creemers JW, Froguel P, Bonnefond A. A nonsense loss-of-function mutation in PCSK1 contributes to dominantly inherited human obesity. Int J Obes (Lond) 2014; 39:295-302. [PMID: 24890885 DOI: 10.1038/ijo.2014.96] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 04/29/2014] [Accepted: 05/15/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND A significant proportion of severe familial forms of obesity remain genetically elusive. Taking advantage of our unique cohort of multigenerational obese families, we aimed to assess the contribution of rare mutations in 29 common obesity-associated genes to familial obesity, and to evaluate in these families the putative presence of nine known monogenic forms of obesity. METHODS Through next-generation sequencing, we sequenced the coding regions of 34 genes involved in polygenic and/or monogenic forms of obesity in 201 participants (75 normal weight individuals, 54 overweight individuals and 72 individuals with obesity class I, II or III) from 13 French families. In vitro functional analyses were performed to investigate the mutation PCSK1-p.Arg80* which was identified in a family. RESULTS A novel heterozygous nonsense variant in PCSK1 (p.Arg80*), encoding a propeptide truncated to less than two exons (out of 14), was found to co-segregate with obesity in a three-generation family. We demonstrated that this mutation inhibits PCSK1 enzyme activity and that this inhibition most likely does not involve a strong physical interaction. Furthermore, both mutations PCSK1-p.Asn180Ser and POMC-p.Phe144Leu, which had previously been reported to be associated with severe obesity, were also identified in this study, but did not co-segregate with obesity. Finally, we did not identify any rare mutations co-segregating with obesity in common obesity susceptibility genes, except for CADM2 and QPCTL, where we found two novel variants (p.Arg81His and p.Leu98Pro, respectively) in three obese individuals. CONCLUSIONS We showed for the first time that a nonsense mutation in PCSK1 was likely to cause dominantly inherited human obesity, due to the inhibiting properties of the propeptide fragment encoded by the null allele. Furthermore, the present family sequencing design challenged the contribution of previously reported mutations to monogenic or at least severe obesity.
Collapse
Affiliation(s)
- J Philippe
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France
| | - P Stijnen
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - D Meyre
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France [4] Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - F De Graeve
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France
| | - D Thuillier
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France
| | - J Delplanque
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France
| | | | - O Sand
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France
| | - J W Creemers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - P Froguel
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France [4] Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - A Bonnefond
- 1] CNRS-UMR8199, Institut de Biologie de Lille, Lille Pasteur Institute, Lille, France [2] Lille 2 University, Lille, France [3] European Genomic Institute for Diabetes (EGID), Lille, France
| |
Collapse
|
90
|
Abstract
Under normal conditions, food intake and energy expenditure are balanced by a homeostatic system that maintains stability of body fat content over time. However, this homeostatic system can be overridden by the activation of 'emergency response circuits' that mediate feeding responses to emergent or stressful stimuli. Inhibition of these circuits is therefore permissive for normal energy homeostasis to occur, and their chronic activation can cause profound, even life-threatening, changes in body fat mass. This Review highlights how the interplay between homeostatic and emergency feeding circuits influences the biologically defended level of body weight under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Gregory J Morton
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington 98109, USA
| | - Thomas H Meek
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington 98109, USA
| | - Michael W Schwartz
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington 98109, USA
| |
Collapse
|
91
|
Kim MJ, Oksenberg N, Hoffmann TJ, Vaisse C, Ahituv N. Functional characterization of SIM1-associated enhancers. Hum Mol Genet 2014; 23:1700-8. [PMID: 24203700 PMCID: PMC3943516 DOI: 10.1093/hmg/ddt559] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/15/2013] [Accepted: 10/31/2013] [Indexed: 12/20/2022] Open
Abstract
Haploinsufficiency of the single-minded homology 1 (SIM1) gene in humans and mice leads to severe obesity, suggesting that altered expression of SIM1, by way of regulatory elements such as enhancers, could predispose individuals to obesity. Here, we identified transcriptional enhancers that could regulate SIM1, using comparative genomics coupled with zebrafish and mouse transgenic enhancer assays. Owing to the dual role of Sim1 in hypothalamic development and in adult energy homeostasis, the enhancer activity of these sequences was annotated from embryonic to adult age. Of the seventeen tested sequences, two SIM1 candidate enhancers (SCE2 and SCE8) were found to have brain-enhancer activity in zebrafish. Both SCE2 and SCE8 also exhibited embryonic brain-enhancer expression in mice, and time course analysis of SCE2 activity showed overlapping expression with Sim1 from embryonic to adult age, notably in the hypothalamus in adult mice. Using a deletion series, we identified the critical region in SCE2 that is needed for enhancer activity in the developing brain. Sequencing this region in obese and lean cohorts revealed a higher prevalence of single nucleotide polymorphisms (SNPs) that were unique to obese individuals, with one variant reducing developmental-enhancer activity in zebrafish. In summary, we have characterized two brain enhancers in the SIM1 locus and identified a set of obesity-specific SNPs within one of them, which may predispose individuals to obesity.
Collapse
Affiliation(s)
- Mee J. Kim
- Department of Bioengineering and Therapeutic Sciences
- Institute for Human Genetics
| | - Nir Oksenberg
- Department of Bioengineering and Therapeutic Sciences
- Institute for Human Genetics
| | - Thomas J. Hoffmann
- Institute for Human Genetics
- Department of Epidemiology and Biostatistics and
| | - Christian Vaisse
- Institute for Human Genetics
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences
- Institute for Human Genetics
| |
Collapse
|
92
|
Bersten DC, Bruning JB, Peet DJ, Whitelaw ML. Human variants in the neuronal basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) transcription factor complex NPAS4/ARNT2 disrupt function. PLoS One 2014; 9:e85768. [PMID: 24465693 PMCID: PMC3894988 DOI: 10.1371/journal.pone.0085768] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/06/2013] [Indexed: 11/25/2022] Open
Abstract
Neuronal Per-Arnt-Sim homology (PAS) Factor 4 (NPAS4) is a neuronal activity-dependent transcription factor which heterodimerises with ARNT2 to regulate genes involved in inhibitory synapse formation. NPAS4 functions to maintain excitatory/inhibitory balance in neurons, while mouse models have shown it to play roles in memory formation, social interaction and neurodegeneration. NPAS4 has therefore been implicated in a number of neuropsychiatric or neurodegenerative diseases which are underpinned by defects in excitatory/inhibitory balance. Here we have explored a broad set of non-synonymous human variants in NPAS4 and ARNT2 for disruption of NPAS4 function. We found two variants in NPAS4 (F147S and E257K) and two variants in ARNT2 (R46W and R107H) which significantly reduced transcriptional activity of the heterodimer on a luciferase reporter gene. Furthermore, we found that NPAS4.F147S was unable to activate expression of the NPAS4 target gene BDNF due to reduced dimerisation with ARNT2. Homology modelling predicts F147 in NPAS4 to lie at the dimer interface, where it appears to directly contribute to protein/protein interaction. We also found that reduced transcriptional activation by ARNT2 R46W was due to disruption of nuclear localisation. These results provide insight into the mechanisms of NPAS4/ARNT dimerisation and transcriptional activation and have potential implications for cognitive phenotypic variation and diseases such as autism, schizophrenia and dementia.
Collapse
Affiliation(s)
- David C. Bersten
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - John B. Bruning
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Daniel J. Peet
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Murray L. Whitelaw
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
93
|
Abstract
Obesity and its related metabolic consequences represent a major public health problem. Huge changes within the environment have undoubtedly contributed to the increased prevalence of obesity but genetic factors are also critical in determining an individual's predisposition to gain weight. The last two decades have seen a huge increase in the understanding of the mechanisms controlling appetitive behavior, body composition, and energy expenditure. Many regions throughout the central nervous system play critical roles in these processes but the hypothalamus, in particular, receives and orchestrates a variety of signals to bring about coordinated changes in energy balance. Reviewing data from human genetic and model organism studies, we consider how disruptions of hypothalamic pathways evolved to maintain energy homeostasis and go on to cause obesity. We highlight ongoing technological developments which continue to lead to novel insights and discuss how this increased knowledge may lead to effective therapeutic interventions in the future.
Collapse
Affiliation(s)
- Rachel Larder
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Chung Thong Lim
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Anthony P Coll
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
94
|
Ramachandrappa S, Raimondo A, Cali AM, Keogh JM, Henning E, Saeed S, Thompson A, Garg S, Bochukova EG, Brage S, Trowse V, Wheeler E, Sullivan AE, Dattani M, Clayton PE, Datta V, Bruning JB, Wareham NJ, O’Rahilly S, Peet DJ, Barroso I, Whitelaw ML, Farooqi IS, Farooqi IS. Rare variants in single-minded 1 (SIM1) are associated with severe obesity. J Clin Invest 2013; 123:3042-50. [PMID: 23778139 PMCID: PMC3696558 DOI: 10.1172/jci68016] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 04/18/2013] [Indexed: 02/02/2023] Open
Abstract
Single-minded 1 (SIM1) is a basic helix-loop-helix transcription factor involved in the development and function of the paraventricular nucleus of the hypothalamus. Obesity has been reported in Sim1 haploinsufficient mice and in a patient with a balanced translocation disrupting SIM1. We sequenced the coding region of SIM1 in 2,100 patients with severe, early onset obesity and in 1,680 controls. Thirteen different heterozygous variants in SIM1 were identified in 28 unrelated severely obese patients. Nine of the 13 variants significantly reduced the ability of SIM1 to activate a SIM1-responsive reporter gene when studied in stably transfected cells coexpressing the heterodimeric partners of SIM1 (ARNT or ARNT2). SIM1 variants with reduced activity cosegregated with obesity in extended family studies with variable penetrance. We studied the phenotype of patients carrying variants that exhibited reduced activity in vitro. Variant carriers exhibited increased ad libitum food intake at a test meal, normal basal metabolic rate, and evidence of autonomic dysfunction. Eleven of the 13 probands had evidence of a neurobehavioral phenotype. The phenotypic similarities between patients with SIM1 deficiency and melanocortin 4 receptor (MC4R) deficiency suggest that some of the effects of SIM1 deficiency on energy homeostasis are mediated by altered melanocortin signaling.
Collapse
Affiliation(s)
- Shwetha Ramachandrappa
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Anne Raimondo
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Anna M.G. Cali
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Julia M. Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Sadia Saeed
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Amanda Thompson
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Sumedha Garg
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Elena G. Bochukova
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Soren Brage
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Victoria Trowse
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Eleanor Wheeler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Adrienne E. Sullivan
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Mehul Dattani
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Peter E. Clayton
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Vippan Datta
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - John B. Bruning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Nick J. Wareham
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Stephen O’Rahilly
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Daniel J. Peet
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Ines Barroso
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - Murray L. Whitelaw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | - I. Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Discipline of Biochemistry, School of Molecular and Biomedical Science and Australian Research Council Special Research Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, Australia.
Wellcome Trust Sanger Institute, Cambridge, United Kingdom.
MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom.
Clinical and Molecular Genetics Unit, University College London Institute of Child Health, London, United Kingdom.
Manchester Academic Health Sciences Centre, Royal Manchester Children’s Hospital, Manchester, United Kingdom.
Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, United Kingdom
| | | |
Collapse
|