1
|
van Vugt M, Finan C, Chopade S, Providencia R, Bezzina CR, Asselbergs FW, van Setten J, Schmidt AF. Integrating metabolomics and proteomics to identify novel drug targets for heart failure and atrial fibrillation. Genome Med 2024; 16:120. [PMID: 39434187 PMCID: PMC11492627 DOI: 10.1186/s13073-024-01395-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Altered metabolism plays a role in the pathophysiology of cardiac diseases, such as atrial fibrillation (AF) and heart failure (HF). We aimed to identify novel plasma metabolites and proteins associating with cardiac disease. METHODS Mendelian randomisation (MR) was used to assess the association of 174 metabolites measured in up to 86,507 participants with AF, HF, dilated cardiomyopathy (DCM), and non-ischemic cardiomyopathy (NICM). Subsequently, we sourced data on 1567 plasma proteins and performed cis MR to identify proteins affecting the identified metabolites as well as the cardiac diseases. Proteins were prioritised on cardiac expression and druggability, and mapped to biological pathways. RESULTS We identified 35 metabolites associating with cardiac disease. AF was affected by seventeen metabolites, HF by nineteen, DCM by four, and NCIM by taurine. HF was particularly enriched for phosphatidylcholines (p = 0.029) and DCM for acylcarnitines (p = 0.001). Metabolite involvement with AF was more uniform, spanning for example phosphatidylcholines, amino acids, and acylcarnitines. We identified 38 druggable proteins expressed in cardiac tissue, with a directionally concordant effect on metabolites and cardiac disease. We recapitulated known associations, for example between the drug target of digoxin (AT1B2), taurine and NICM risk. Additionally, we identified numerous novel findings, such as higher RET values associating with phosphatidylcholines and decreasing AF and HF. RET is targeted by drugs such as regorafenib which has known cardiotoxic side-effects. Pathway analysis implicated involvement of GDF15 signalling through RET, and ghrelin regulation of energy homeostasis in cardiac pathogenesis. CONCLUSIONS This study identified 35 plasma metabolites involved with cardiac diseases and linked these to 38 druggable proteins, providing actionable leads for drug development.
Collapse
Affiliation(s)
- Marion van Vugt
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands.
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK.
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands.
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands.
| | - Chris Finan
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL British Heart Foundation Research Accelerator, London, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL British Heart Foundation Research Accelerator, London, UK
| | - Rui Providencia
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Connie R Bezzina
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- Department of Experimental Cardiology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- European Reference Network for rare, low prevalence and complex diseases of the heart: ERN GUARD-Heart , Amsterdam, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
| | - A Floriaan Schmidt
- Department of Cardiology, University Medical Center Utrecht, Utrecht University, Division Heart & Lungs, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
- UCL British Heart Foundation Research Accelerator, London, UK
| |
Collapse
|
2
|
Song S, Qiao J, Zhao R, Lu YJ, Wang C, Chang MJ, Zhang HY, Li XF, Wang CH. Identification of novel drug targets for osteoarthritis by integrating genetics and proteomes from blood. J Orthop Surg Res 2024; 19:559. [PMID: 39261869 PMCID: PMC11389225 DOI: 10.1186/s13018-024-05034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative osteoarticular disease, involving genetic predisposition. How the risk variants confer the risk of OA through their effects on proteins remains largely unknown. Therefore, we aimed to discover new and effective drug targets for OA and its subtypes. METHODS A proteome-wide association study (PWAS) was performed based on OA and its subtypes genome-wide association studies (GWAS) summary datasets and the protein quantitative trait loci (pQTL) data. Subsequently, Mendelian randomization (MR) and colocalization analysis was conducted to estimate the associations between protein and OA risk. The replication analysis was performed in an independent dataset of human plasma pQTL data. RESULTS The abundance of seven proteins was causally related to OA, two proteins to knee OA and six proteins to hip OA, respectively. We replicated 2 of these proteins using an independent pQTL dataset. With the further support of colocalization, and higher ECM1 level was causally associated with a higher risk of OA and hip OA. Higher PCSK1 level was causally associated with a lower risk of OA. And higher levels of ITIH1, EFEMP1, and ERLEC1 were associated with decreased risk of hip OA. CONCLUSION Our study provides new insights into the genetic component of protein abundance in OA and a promising therapeutic target for future drug development.
Collapse
Affiliation(s)
- Shan Song
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Jun Qiao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Yu-Jie Lu
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Can Wang
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Min-Jing Chang
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - He-Yi Zhang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Xiao-Feng Li
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Ministry of Education Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, China
| | - Cai-Hong Wang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
3
|
Manco L, Albuquerque D, Aranda B, Rodrigues D, Machado-Rodrigues AM, Padez C. Differential sex-association between PCSK1 polymorphisms and obesity risk in Portuguese children. Am J Hum Biol 2024; 36:e24023. [PMID: 38009939 DOI: 10.1002/ajhb.24023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVES The proprotein convertase subtilisin/Kexin type 1 gene (PCSK1) is implicated in hypothalamic appetite control. Several studies have addressed the relationship between PCSK1 polymorphisms and obesity, although conflicting results were observed. We tested the potential association of four PCSK1 variants with the risk of overweight/obesity and related variables in Portuguese children. METHODS This is a case-control study, where four PCSK1 variants, rs6230 (c.-101T>C), rs6232 (p.N221D), rs6235 (p.S690T), and rs3811942 (c.*265T>C), were analyzed in Portuguese children (aged 5-13 years-old). Anthropometric measures were objectively collected and used to provide weight-for-age, height-for-age, and body mass index (BMI) for age. The indices generated were compared to standard reference values of WHO to obtain the corresponding Z-scores. RESULTS Logistic regression, in the dominant model, revealed no significant associations between the four individual PCSK1 variants and the risk of overweight/obesity in the total population. However, stratifying the sample by sex, a marginally significant association was found between the rs6235 minor C-allele and increased overweight/obesity in boys (n = 345) (OR 1.55 [1.01-2.38] p = .044), but not in girls (n = 340) (OR 0.73 [0.46-1.14] p = .169). Consistently, boys with genotype GG presented lower BMI Z-score (0.62) when compared to those with the genotypes GC + CC (1.04). Testing for different effects in males versus females, a significant interaction was found between the rs6235 polymorphism and sex for BMI Z-score (p = .025). CONCLUSIONS Results of this study suggest for a sex-differentiated association between PCSK1 rs6235 and overweight/ obesity in Portuguese children.
Collapse
Affiliation(s)
- Licínio Manco
- Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - David Albuquerque
- Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
| | - Beatriz Aranda
- Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
| | - Daniela Rodrigues
- Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
| | - Aristides M Machado-Rodrigues
- Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
- Faculty of Sport Sciences and Physical Education, University of Coimbra, Coimbra, Portugal
| | - Cristina Padez
- Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
4
|
Elliott A, Walters RK, Pirinen M, Kurki M, Junna N, Goldstein JI, Reeve MP, Siirtola H, Lemmelä SM, Turley P, Lahtela E, Mehtonen J, Reis K, Elnahas AG, Reigo A, Palta P, Esko T, Mägi R, Palotie A, Daly MJ, Widén E. Distinct and shared genetic architectures of gestational diabetes mellitus and type 2 diabetes. Nat Genet 2024; 56:377-382. [PMID: 38182742 PMCID: PMC10937370 DOI: 10.1038/s41588-023-01607-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 11/07/2023] [Indexed: 01/07/2024]
Abstract
Gestational diabetes mellitus (GDM) is a common metabolic disorder affecting more than 16 million pregnancies annually worldwide1,2. GDM is related to an increased lifetime risk of type 2 diabetes (T2D)1-3, with over a third of women developing T2D within 15 years of their GDM diagnosis. The diseases are hypothesized to share a genetic predisposition1-7, but few studies have sought to uncover the genetic underpinnings of GDM. Most studies have evaluated the impact of T2D loci only8-10, and the three prior genome-wide association studies of GDM11-13 have identified only five loci, limiting the power to assess to what extent variants or biological pathways are specific to GDM. We conducted the largest genome-wide association study of GDM to date in 12,332 cases and 131,109 parous female controls in the FinnGen study and identified 13 GDM-associated loci, including nine new loci. Genetic features distinct from T2D were identified both at the locus and genomic scale. Our results suggest that the genetics of GDM risk falls into the following two distinct categories: one part conventional T2D polygenic risk and one part predominantly influencing mechanisms disrupted in pregnancy. Loci with GDM-predominant effects map to genes related to islet cells, central glucose homeostasis, steroidogenesis and placental expression.
Collapse
Grants
- R00 AG062787 NIA NIH HHS
- R01 MH101244 NIMH NIH HHS
- A.E. was a research Scholar supported by Sarnoff Cardiovascular Research Foundation
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- Academy of Finland (Suomen Akatemia)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- The FinnGen project is funded by two grants from Business Finland (HUS 4685/31/2016 and UH 4386/31/2016) and by eleven industry partners (AbbVie Inc, AstraZeneca UK Ltd, Biogen MA Inc, Celgene Corporation, Celgene International II Sàrl, Genentech Inc, Merck Sharp & Dohme Corp, Pfizer Inc., GlaxoSmithKline, Sanofi, Maze Therapeutics Inc., Janssen Biotech Inc).
- EstBB GWAS analysis is supported by research funding from the Estonian Research Council: Team grant PRG1291 and PRG1911.
Collapse
Affiliation(s)
- Amanda Elliott
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Mitja Kurki
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Nella Junna
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Jacqueline I Goldstein
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mary Pat Reeve
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Harri Siirtola
- TAUCHI Research Center, Faculty of Information Technology and Communication Sciences (ITC), Tampere University, Tampere, Finland
| | - Susanna M Lemmelä
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Patrick Turley
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
- Department of Economics, University of Southern California, Los Angeles, CA, USA
| | - Elisa Lahtela
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Juha Mehtonen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Kadri Reis
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Anu Reigo
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Priit Palta
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Aarno Palotie
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
5
|
Guijo B, Argente J, Martos-Moreno GÁ. The N221D variant in PCSK1 is highly prevalent in childhood obesity and can influence the metabolic profile. J Pediatr Endocrinol Metab 2023; 36:1140-1145. [PMID: 37877373 DOI: 10.1515/jpem-2023-0395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
OBJECTIVES To study the prevalence and influence on metabolic profile of the prohormone-convertase-1 (PCSK1) N221D variant in childhood obesity, proven its role in the leptin-melanocortin signaling pathway as in proinsulin and other prohormone cleavage. METHODS Transversal study of 1066 children with obesity (mean age and BMI Z-score 10.38 ± 3.44 years and +4.38 ± 1.77, respectively), 51.4 % males, 54.4 % prepubertal, 71.5 % Caucasians and 20.8 % Latinos. Anthropometric and metabolic features were compared between patients carrying the N221D variant in PCSK1 and patients with no variants found after next generation sequencing analysis of 17 genes (CREBBP, CPE, HTR2C, KSR2, LEP, LEPR, MAGEL2, MC3R, MC4R, MRAP2, NCOA1, PCSK1, POMC, SH2B1, SIM1, TBX3 and TUB) involved in the leptin-melanocortin pathway. RESULTS No variants were found in 531 patients (49.8 %), while 68 patients carried the PCSK1 N221D variant (42 isolately, and 26 with at least one additional gene variant). Its prevalence was higher in Caucasians vs. Latinos (χ2 7.81; p<0.01). Patients carrying exclusively the PCSK1 N221D variant (n=42) showed lower insulinemia (p<0.05), HOMA index (p<0.05) and area under the curve for insulin in the oral glucose tolerance test (p<0.001) and higher WBISI (p<0.05) than patients with no variants, despite similar obesity severity, age, sex and ethnic distribution. CONCLUSIONS The N221D variant in PCSK1 is highly prevalent in childhood obesity, influenced by ethnicity. Indirect estimation of insulin resistance, based on insulinemia could be byassed in these patients and underestimate their type 2 diabetes mellitus risk.
Collapse
Affiliation(s)
- Blanca Guijo
- Departments of Pediatrics and Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Jesús Argente
- Departments of Pediatrics and Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de alimentación IMDEA, CEIUAM+CSIC, Madrid, Spain
| | - Gabriel Ángel Martos-Moreno
- Departments of Pediatrics and Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
6
|
Keller M, Svensson SIA, Rohde-Zimmermann K, Kovacs P, Böttcher Y. Genetics and Epigenetics in Obesity: What Do We Know so Far? Curr Obes Rep 2023; 12:482-501. [PMID: 37819541 DOI: 10.1007/s13679-023-00526-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE OF REVIEW Enormous progress has been made in understanding the genetic architecture of obesity and the correlation of epigenetic marks with obesity and related traits. This review highlights current research and its challenges in genetics and epigenetics of obesity. RECENT FINDINGS Recent progress in genetics of polygenic traits, particularly represented by genome-wide association studies, led to the discovery of hundreds of genetic variants associated with obesity, which allows constructing polygenic risk scores (PGS). In addition, epigenome-wide association studies helped identifying novel targets and methylation sites being important in the pathophysiology of obesity and which are essential for the generation of methylation risk scores (MRS). Despite their great potential for predicting the individual risk for obesity, the use of PGS and MRS remains challenging. Future research will likely discover more loci being involved in obesity, which will contribute to better understanding of the complex etiology of human obesity. The ultimate goal from a clinical perspective will be generating highly robust and accurate prediction scores allowing clinicians to predict obesity as well as individual responses to body weight loss-specific life-style interventions.
Collapse
Affiliation(s)
- Maria Keller
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Medical Center, University of Leipzig, 04103, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Stina Ingrid Alice Svensson
- EpiGen, Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Kerstin Rohde-Zimmermann
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Medical Center, University of Leipzig, 04103, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Medical Center, University of Leipzig, 04103, Leipzig, Germany
| | - Yvonne Böttcher
- EpiGen, Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway.
- EpiGen, Medical Division, Akershus University Hospital, 1478, Lørenskog, Norway.
| |
Collapse
|
7
|
Velazquez-Roman J, Angulo-Zamudio UA, Leon-Sicairos N, Flores-Villaseñor H, Benitez-Baez M, Espinoza-Salomón A, Karam-León A, Villamil-Ramírez H, Canizales-Quinteros S, Macías-Kauffer L, Monroy-Higuera J, Acosta-Smith E, Canizalez-Roman A. Association of PCSK1 and PPARG1 Allelic Variants with Obesity and Metabolic Syndrome in Mexican Adults. Genes (Basel) 2023; 14:1775. [PMID: 37761915 PMCID: PMC10531047 DOI: 10.3390/genes14091775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Metabolic diseases, including obesity, diabetes, and metabolic syndrome, are among the most important public health challenges worldwide. Metabolic diseases are classified as multifactorial diseases in which genetic variants such as single-nucleotide polymorphisms (SNPs) may play an important role. The present study aimed to identify associations linking allelic variants of the PCSK1, TMEM18, GPX5, ZPR1, ZBTB16, and PPARG1 genes with anthropometric and biochemical traits and metabolic diseases (obesity or metabolic syndrome) in an adult population from northwestern Mexico. METHODS Blood samples were collected from 523 subjects, including 247 with normal weight, 276 with obesity, and 147 with metabolic syndrome. Anthropometric and biochemical characteristics were recorded, and single-nucleotide polymorphisms (SNPs) were genotyped by real-time PCR. RESULTS PCSK1 was significantly (p < 0.05) associated with BMI, weight, and waist-to-hip ratio; TMEM18 was significantly associated with systolic blood pressure and triglyceride levels; GPX5 was significantly associated with HDL cholesterol levels. In addition, PCSK1 was associated with obesity (p = 1.0 × 10-4) and metabolic syndrome (p = 3.0 × 10-3), whereas PPARG1 was associated with obesity (p = 0.044). CONCLUSIONS The associations found in this study, mainly between allelic variants of PCSK1 and metabolic traits, obesity, and metabolic syndrome, may represent a risk for developing metabolic diseases in adult subjects from northwestern Mexico.
Collapse
Affiliation(s)
- Jorge Velazquez-Roman
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
| | - Uriel A. Angulo-Zamudio
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
| | - Nidia Leon-Sicairos
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
- Pediatric Hospital of Sinaloa, Constitución 530, Jorge Almada, Culiacan Sinaloa 80200, Mexico
| | - Hector Flores-Villaseñor
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
- The Sinaloa State Public Health Laboratory, Secretariat of Health, Culiacan Sinaloa 80020, Mexico
| | - Miriam Benitez-Baez
- Programa de Doctorado, Posgrado Integral en Biotecnología, FCQB, UAS, Culiacan Sinaloa 80013, Mexico; (M.B.-B.); (A.K.-L.); (J.M.-H.)
| | - Ana Espinoza-Salomón
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
| | - Alejandra Karam-León
- Programa de Doctorado, Posgrado Integral en Biotecnología, FCQB, UAS, Culiacan Sinaloa 80013, Mexico; (M.B.-B.); (A.K.-L.); (J.M.-H.)
| | - Hugo Villamil-Ramírez
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/INMEGEN, Mexico City 04510, Mexico; (H.V.-R.); (S.C.-Q.); (L.M.-K.)
| | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/INMEGEN, Mexico City 04510, Mexico; (H.V.-R.); (S.C.-Q.); (L.M.-K.)
| | - Luis Macías-Kauffer
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/INMEGEN, Mexico City 04510, Mexico; (H.V.-R.); (S.C.-Q.); (L.M.-K.)
| | - Jose Monroy-Higuera
- Programa de Doctorado, Posgrado Integral en Biotecnología, FCQB, UAS, Culiacan Sinaloa 80013, Mexico; (M.B.-B.); (A.K.-L.); (J.M.-H.)
| | - Erika Acosta-Smith
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
| | - Adrian Canizalez-Roman
- School of Medicine, Autonomous University of Sinaloa, Culiacan Sinaloa 80019, Mexico; (J.V.-R.); (U.A.A.-Z.); (N.L.-S.); (H.F.-V.); (A.E.-S.); (E.A.-S.)
- The Women’s Hospital, Secretariat of Health, Culiacan Sinaloa 80020, Mexico
| |
Collapse
|
8
|
Zhang X, Hu LG, Lei Y, Stolina M, Homann O, Wang S, Véniant MM, Hsu YH. A transcriptomic and proteomic atlas of obesity and type 2 diabetes in cynomolgus monkeys. Cell Rep 2023; 42:112952. [PMID: 37556324 DOI: 10.1016/j.celrep.2023.112952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2023] [Accepted: 07/23/2023] [Indexed: 08/11/2023] Open
Abstract
Obesity and type 2 diabetes (T2D) remain major global healthcare challenges, and developing therapeutics necessitates using nonhuman primate models. Here, we present a transcriptomic and proteomic atlas of all the major organs of cynomolgus monkeys with spontaneous obesity or T2D in comparison to healthy controls. Molecular changes occur predominantly in the adipose tissues of individuals with obesity, while extensive expression perturbations among T2D individuals are observed in many tissues such as the liver and kidney. Immune-response-related pathways are upregulated in obesity and T2D, whereas metabolism and mitochondrial pathways are downregulated. Moreover, we highlight some potential therapeutic targets, including SLC2A1 and PCSK1 in obesity as well as SLC30A8 and SLC2A2 in T2D. Our study provides a resource for exploring the complex molecular mechanism of obesity and T2D and developing therapies for these diseases, with limitations including lack of hypothalamus, isolated islets of Langerhans, longitudinal data, and body fat percentage.
Collapse
Affiliation(s)
- Xianglong Zhang
- Center for Research Acceleration by Digital Innovation (CRADI), Amgen Research, South San Francisco, CA 94080, USA
| | | | - Ying Lei
- Research China, Amgen Research, Shanghai 200020, China
| | - Marina Stolina
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA 91320, USA
| | - Oliver Homann
- Center for Research Acceleration by Digital Innovation (CRADI), Amgen Research, South San Francisco, CA 94080, USA
| | - Songli Wang
- Research Biomics, Amgen Research, South San Francisco, CA 94080, USA
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA 91320, USA.
| | - Yi-Hsiang Hsu
- Marcus Institute for Aging Research and Harvard Medical School, Boston, MA 02131, USA.
| |
Collapse
|
9
|
Folon L, Baron M, Toussaint B, Vaillant E, Boissel M, Scherrer V, Loiselle H, Leloire A, Badreddine A, Balkau B, Charpentier G, Franc S, Marre M, Aboulouard S, Salzet M, Canouil M, Derhourhi M, Froguel P, Bonnefond A. Contribution of heterozygous PCSK1 variants to obesity and implications for precision medicine: a case-control study. Lancet Diabetes Endocrinol 2023; 11:182-190. [PMID: 36822744 DOI: 10.1016/s2213-8587(22)00392-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 02/24/2023]
Abstract
BACKGROUND Rare biallelic pathogenic mutations in PCSK1 (encoding proprotein convertase subtilisin/kexin type 1 [PC1/3]) cause early-onset obesity associated with various endocrinopathies. Setmelanotide has been approved for carriers of these biallelic mutations in the past 3 years. We aimed to perform a large-scale functional genomic study focusing on rare heterozygous variants of PCSK1 to decipher their putative impact on obesity risk. METHODS This case-control study included all participants with overweight and obesity (ie, cases) or healthy weight (ie, controls) from the RaDiO study of three community-based and one hospital-based cohort in France recruited between Jan 1, 1995, and Dec 31, 2000. In adults older than 18 years, healthy weight was defined as BMI of less than 25·0 kg/m2, overweight as 25·0-29·9 kg/m2, and obesity as 30·0 kg/m2 or higher. Participants with type 2 diabetes had fasting glucose of 7·0 mmol/L or higher or used treatment for hyperglycaemia (or both) and were negative for islet or insulin autoantibodies. Functional assessment of rare missense variants of PCSK1 was performed. Pathogenicity clusters of variants were determined with machine learning. The effect of each cluster of PCSK1 variants on obesity was assessed using the adjusted mixed-effects score test. FINDINGS All 13 coding exons of PCSK1 were sequenced in 9320 participants (including 7260 adults and 2060 children and adolescents) recruited from the RaDiO study. We detected 65 rare heterozygous PCSK1 variants, including four null variants and 61 missense variants that were analysed in vitro and clustered into five groups (A-E), according to enzymatic activity. Compared with the wild-type, 15 missense variants led to complete PC1/3 loss of function (group A; reference) and rare exome variant ensemble learner (REVEL) led to 15 (25%) false positives and four (7%) false negatives. Carrying complete loss-of-function or null PCSK1 variants was significantly associated with obesity (six [86%] of seven carriers vs 1518 [35%] of 4395 non-carriers; OR 9·3 [95% CI 1·5-177·4]; p=0·014) and higher BMI (32·0 kg/m2 [SD 9·3] in carriers vs 27·3 kg/m2 [6·5] in non-carriers; mean effect π 6·94 [SE 1·95]; p=0·00029). Clusters of PCSK1 variants with partial or neutral effect on PC1/3 activity did not have an effect on obesity or overweight and on BMI. INTERPRETATION Only carriers of heterozygous, null, or complete loss-of-function PCSK1 variants cause monogenic obesity and, therefore, might be eligible for setmelanotide. In silico tests were unable to accurately detect these variants, which suggests that in vitro assays are necessary to determine the variant pathogenicity for genetic diagnosis and precision medicine purposes. FUNDING Agence Nationale de la Recherche, European Research Council, National Center for Precision Diabetic Medicine, European Regional Development Fund, Hauts-de-France Regional Council, and the European Metropolis of Lille.
Collapse
Affiliation(s)
- Lise Folon
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Morgane Baron
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Bénédicte Toussaint
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Emmanuel Vaillant
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Mathilde Boissel
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Victoria Scherrer
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Hélène Loiselle
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Audrey Leloire
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Alaa Badreddine
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Beverley Balkau
- Paris-Saclay University, Paris-Sud University, Université de Versailles Saint-Quentin-en-Yvelines, Center for Research in Epidemiology and Population Health, Inserm U1018 Clinical Epidemiology, Villejuif, France
| | - Guillaume Charpentier
- Centre d'Étude et de Recherche pour l'Intensification du Traitement du Diabète, Evry, France
| | - Sylvia Franc
- Centre d'Étude et de Recherche pour l'Intensification du Traitement du Diabète, Evry, France; Department of Diabetes, Sud-Francilien Hospital, Paris-Sud University, Corbeil-Essonnes, France
| | - Michel Marre
- Institut Necker-Enfants Malades, INSERM, Université de Paris, Paris, France; Clinique Ambroise Paré, Neuilly-sur-Seine, France
| | - Soulaimane Aboulouard
- Université de Lille, Lille, France; Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse, Lille, France
| | - Michel Salzet
- Université de Lille, Lille, France; Inserm U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse, Lille, France
| | - Mickaël Canouil
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Mehdi Derhourhi
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France
| | - Philippe Froguel
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France; Department of Metabolism, Imperial College London, London, UK
| | - Amélie Bonnefond
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille University Hospital, Lille, France; Université de Lille, Lille, France; Department of Metabolism, Imperial College London, London, UK.
| |
Collapse
|
10
|
Elliott A, Walters RK, Pirinen M, Kurki M, Junna N, Goldstein J, Reeve M, Siirtola H, Lemmelä S, Turley P, Palotie A, Daly M, Widén E. Distinct and shared genetic architectures of Gestational diabetes mellitus and Type 2 Diabetes Mellitus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.16.23286014. [PMID: 36865330 PMCID: PMC9980250 DOI: 10.1101/2023.02.16.23286014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Gestational diabetes mellitus (GDM) affects more than 16 million pregnancies annually worldwide and is related to an increased lifetime risk of Type 2 diabetes (T2D). The diseases are hypothesized to share a genetic predisposition, but there are few GWAS studies of GDM and none of them is sufficiently powered to assess whether any variants or biological pathways are specific to GDM. We conducted the largest genome-wide association study of GDM to date in 12,332 cases and 131,109 parous female controls in the FinnGen Study and identified 13 GDM-associated loci including 8 novel loci. Genetic features distinct from T2D were identified both at the locus and genomic scale. Our results suggest that the genetics of GDM risk falls into two distinct categories - one part conventional T2D polygenic risk and one part predominantly influencing mechanisms disrupted in pregnancy. Loci with GDM-predominant effects map to genes related to islet cells, central glucose homeostasis, steroidogenesis, and placental expression. These results pave the way for an improved biological understanding of GDM pathophysiology and its role in the development and course of T2D.
Collapse
Affiliation(s)
- A. Elliott
- Analytic and Translational Genetics Unit, Massachusetts Gen. Hosp., Boston, MA
- Stanley Ctr. for Psychiatric Res., Broad Inst. of Harvard and MIT, Cambridge, MA
- Harvard Med. Sch., Boston, MA
| | - R. K. Walters
- Analytic and Translational Genetics Unit, Massachusetts Gen. Hosp., Boston, MA
- Stanley Ctr. for Psychiatric Res., Broad Inst. of Harvard and MIT, Cambridge, MA
- Harvard Med. Sch., Boston, MA
| | - M. Pirinen
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - M. Kurki
- Analytic and Translational Genetics Unit, Massachusetts Gen. Hosp., Boston, MA
- Stanley Ctr. for Psychiatric Res., Broad Inst. of Harvard and MIT, Cambridge, MA
| | - N. Junna
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
| | - J. Goldstein
- Stanley Ctr. for Psychiatric Res., Broad Inst. of Harvard and MIT, Cambridge, MA
| | - M.P. Reeve
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
| | - H. Siirtola
- TAUCHI Research Center, Faculty of Information Technology and Communication Sciences (ITC), Tampere University, Tampere, Finland
| | - S. Lemmelä
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - P. Turley
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
- Department of Economics, University of Southern California, Los Angeles, CA, USA
| | | | - A. Palotie
- Analytic and Translational Genetics Unit, Massachusetts Gen. Hosp., Boston, MA
- Stanley Ctr. for Psychiatric Res., Broad Inst. of Harvard and MIT, Cambridge, MA
- Harvard Med. Sch., Boston, MA
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
| | - M. Daly
- Analytic and Translational Genetics Unit, Massachusetts Gen. Hosp., Boston, MA
- Stanley Ctr. for Psychiatric Res., Broad Inst. of Harvard and MIT, Cambridge, MA
- Harvard Med. Sch., Boston, MA
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
| | - E. Widén
- Institute for Molecular Med. Finland, Helsinki Institute of Life Sciences., University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Bernard A, Ojeda Naharros I, Yue X, Mifsud F, Blake A, Bourgain-Guglielmetti F, Ciprin J, Zhang S, McDaid E, Kim K, Nachury MV, Reiter JF, Vaisse C. MRAP2 regulates energy homeostasis by promoting primary cilia localization of MC4R. JCI Insight 2023; 8:e155900. [PMID: 36692018 PMCID: PMC9977312 DOI: 10.1172/jci.insight.155900] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 11/30/2022] [Indexed: 01/24/2023] Open
Abstract
The G protein-coupled receptor melanocortin-4 receptor (MC4R) and its associated protein melanocortin receptor-associated protein 2 (MRAP2) are essential for the regulation of food intake and body weight in humans. MC4R localizes and functions at the neuronal primary cilium, a microtubule-based organelle that senses and relays extracellular signals. Here, we demonstrate that MRAP2 is critical for the weight-regulating function of MC4R neurons and the ciliary localization of MC4R. More generally, our study also reveals that GPCR localization to primary cilia can require specific accessory proteins that may not be present in heterologous cell culture systems. Our findings further demonstrate that targeting of MC4R to neuronal primary cilia is essential for the control of long-term energy homeostasis and suggest that genetic disruption of MC4R ciliary localization may frequently underlie inherited forms of obesity.
Collapse
Affiliation(s)
| | | | - Xinyu Yue
- Department of Medicine and The Diabetes Center
| | | | - Abbey Blake
- Department of Medicine and The Diabetes Center
| | | | | | - Sumei Zhang
- Department of Medicine and The Diabetes Center
| | - Erin McDaid
- Department of Medicine and The Diabetes Center
| | - Kellan Kim
- Department of Medicine and The Diabetes Center
| | | | - Jeremy F. Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | | |
Collapse
|
12
|
Gentry AE, Alexander JC, Ahangari M, Peterson RE, Miles MF, Bettinger JC, Davies AG, Groteweil M, Bacanu SA, Kendler KS, Riley BP, Webb BT. Case-only exome variation analysis of severe alcohol dependence using a multivariate hierarchical gene clustering approach. PLoS One 2023; 18:e0283985. [PMID: 37098020 PMCID: PMC10128939 DOI: 10.1371/journal.pone.0283985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Variation in genes involved in ethanol metabolism has been shown to influence risk for alcohol dependence (AD) including protective loss of function alleles in ethanol metabolizing genes. We therefore hypothesized that people with severe AD would exhibit different patterns of rare functional variation in genes with strong prior evidence for influencing ethanol metabolism and response when compared to genes not meeting these criteria. OBJECTIVE Leverage a novel case only design and Whole Exome Sequencing (WES) of severe AD cases from the island of Ireland to quantify differences in functional variation between genes associated with ethanol metabolism and/or response and their matched control genes. METHODS First, three sets of ethanol related genes were identified including those a) involved in alcohol metabolism in humans b) showing altered expression in mouse brain after alcohol exposure, and altering ethanol behavioral responses in invertebrate models. These genes of interest (GOI) sets were matched to control gene sets using multivariate hierarchical clustering of gene-level summary features from gnomAD. Using WES data from 190 individuals with severe AD, GOI were compared to matched control genes using logistic regression to detect aggregate differences in abundance of loss of function, missense, and synonymous variants, respectively. RESULTS Three non-independent sets of 10, 117, and 359 genes were queried against control gene sets of 139, 1522, and 3360 matched genes, respectively. Significant differences were not detected in the number of functional variants in the primary set of ethanol-metabolizing genes. In both the mouse expression and invertebrate sets, we observed an increased number of synonymous variants in GOI over matched control genes. Post-hoc simulations showed the estimated effects sizes observed are unlikely to be under-estimated. CONCLUSION The proposed method demonstrates a computationally viable and statistically appropriate approach for genetic analysis of case-only data for hypothesized gene sets supported by empirical evidence.
Collapse
Affiliation(s)
- Amanda Elswick Gentry
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jeffry C Alexander
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Mohammad Ahangari
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Integrative Life Sciences Ph.D. Program, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Roseann E Peterson
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry and Behavioral Sciences, Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, New York, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michael F Miles
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jill C Bettinger
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Andrew G Davies
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Mike Groteweil
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Silviu A Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Brien P Riley
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Bradley T Webb
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, North Caroline, United States of America
| |
Collapse
|
13
|
Littleton SH, Grant SFA. Strategies to identify causal common genetic variants and corresponding effector genes for paediatric obesity. Pediatr Obes 2022; 17:e12968. [PMID: 35971868 DOI: 10.1111/ijpo.12968] [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: 01/24/2022] [Revised: 07/24/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Childhood obesity rates are on the rise, but there are currently no effective therapies available to slow or halt their progression. Although environmental and lifestyle factors have been implicated in its pathogenesis, childhood obesity is considered a complex disorder with a clear genetic component. Intense genome-wide association study (GWAS) efforts through large-scale collaborations have enabled the discovery of genetic loci robustly associated with childhood obesity beyond the classic FTO locus. That said, GWAS itself does not pinpoint the actual underlying causal effector genes, but rather just yields association signals in the genome. OBJECTIVE This review aims to outline what has been elucidated thus far on the genetic aetiology of commong childhood obesity and to describe strategies to identify and validate both causal common genetic variants and their corresponding effector genes. RESULTS Relevant cell types for molecular studies can be identified by gene set enrichment analysis and considering known biology of obesity-related physiological processes. Putatively causal single nucleotide polymorphisms (SNPs) can be identified by several methods including statistical fine mapping and 'assay for transposase accessible chromatin sequencing' (ATAC-seq). Variant to gene mapping can then nominate effector genes likely regulated by cis-regulatory elements harbouring putatively causal SNPs. A SNP's cis-regulatory activity can be functionally validated by several in vitro methods including luciferase assay and CRISPR approaches. These CRISPR approaches can also be used to investigate how dysregulatn of effector genes may confer obesity risk. CONCLUSION Uncovering the causative genes related to GWAS signals and elucidating their functional contributions to paediatric obesity with these strategies will deepen our understanding of this disease and serve better treatment outcomes.
Collapse
Affiliation(s)
- Sheridan H Littleton
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, USA.,Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA.,Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, USA.,Divisons of Genetics and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, USA.,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| |
Collapse
|
14
|
Guevara-Ramírez P, Cadena-Ullauri S, Ruiz-Pozo VA, Tamayo-Trujillo R, Paz-Cruz E, Simancas-Racines D, Zambrano AK. Genetics, genomics, and diet interactions in obesity in the Latin American environment. Front Nutr 2022; 9:1063286. [PMID: 36532520 PMCID: PMC9751379 DOI: 10.3389/fnut.2022.1063286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 10/25/2022] [Indexed: 08/25/2023] Open
Abstract
Obesity is a chronic disease characterized by abnormal or excessive fat accumulation that could impact an individual's health; moreover, the World Health Organization (WHO) has declared obesity a global epidemic since 1997. In Latin America, in 2016, reports indicated that 24.2% of the adult population was obese. The environmental factor or specific behaviors like dietary intake or physical activity have a vital role in the development of a condition like obesity, but the interaction of genes could contribute to that predisposition. Hence, it is vital to understand the relationship between genes and disease. Indeed, genetics in nutrition studies the genetic variations and their effect on dietary response; while genomics in nutrition studies the role of nutrients in gene expression. The present review represents a compendium of the dietary behaviors in the Latin American environment and the interactions of genes with their single nucleotide polymorphisms (SNPs) associated with obesity, including the risk allele frequencies in the Latin American population. Additionally, a bibliographical selection of several studies has been included; these studies examined the impact that dietary patterns in Latin American environments have on the expression of numerous genes involved in obesity-associated metabolic pathways.
Collapse
Affiliation(s)
- Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Daniel Simancas-Racines
- Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
15
|
Loss of hypothalamic Furin affects POMC to proACTH cleavage and feeding behavior in high-fat diet-fed mice. Mol Metab 2022; 66:101627. [PMID: 36374777 PMCID: PMC9664468 DOI: 10.1016/j.molmet.2022.101627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE The hypothalamus regulates feeding and glucose homeostasis through the balanced action of different neuropeptides, which are cleaved and activated by the proprotein convertases PC1/3 and PC2. However, the recent association of polymorphisms in the proprotein convertase FURIN with type 2 diabetes, metabolic syndrome, and obesity, prompted us to investigate the role of FURIN in hypothalamic neurons controlling glucose and feeding. METHODS POMC-Cre+/- mice were bred with Furinfl/fl mice to generate conditional knockout mice with Furin-deletion in neurons expressing proopiomelanocortin (POMCFurKO), and Furinfl/fl mice were used as controls. POMCFurKO and controls were periodically monitored on both normal chow diet and high fat diet (HFD) for body weight and glucose tolerance by established in-vivo procedures. Food intake was measured in HFD-fed FurKO and controls. Hypothalamic Pomc mRNA was measured by RT-qPCR. ELISAs quantified POMC protein and resulting peptides in the hypothalamic extracts of POMCFurKO mice and controls. The in-vitro processing of POMC was studied by biochemical techniques in HEK293T and CHO cell lines lacking FURIN. RESULTS In control mice, Furin mRNA levels were significantly upregulated on HFD feeding, suggesting an increased demand for FURIN activity in obesogenic conditions. Under these conditions, the POMCFurKO mice were hyperphagic and had increased body weight compared to Furinfl/fl mice. Moreover, protein levels of POMC were elevated and ACTH concentrations markedly reduced. Also, the ratio of α-MSH/POMC was decreased in POMCFurKO mice compared to controls. This indicates that POMC processing was significantly reduced in the hypothalami of POMCFurKO mice, highlighting for the first time the involvement of FURIN in the cleavage of POMC. Importantly, we found that in vitro, the first stage in processing where POMC is cleaved into proACTH was achieved by FURIN but not by PC1/3 or the other proprotein convertases in cell lines lacking a regulated secretory pathway. CONCLUSIONS These results suggest that FURIN processes POMC into proACTH before sorting into the regulated secretory pathway, challenging the dogma that PC1/3 and PC2 are the only convertases responsible for POMC cleavage. Furthermore, its deletion affects feeding behaviors under obesogenic conditions.
Collapse
|
16
|
Hinney A, Körner A, Fischer-Posovszky P. The promise of new anti-obesity therapies arising from knowledge of genetic obesity traits. Nat Rev Endocrinol 2022; 18:623-637. [PMID: 35902734 PMCID: PMC9330928 DOI: 10.1038/s41574-022-00716-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 02/07/2023]
Abstract
Obesity is a multifactorial and complex disease that often manifests in early childhood with a lifelong burden. Polygenic and monogenic obesity are driven by the interaction between genetic predisposition and environmental factors. Polygenic variants are frequent and confer small effect sizes. Rare monogenic obesity syndromes are caused by defined pathogenic variants in single genes with large effect sizes. Most of these genes are involved in the central nervous regulation of body weight; for example, genes of the leptin-melanocortin pathway. Clinically, patients with monogenic obesity present with impaired satiety, hyperphagia and pronounced food-seeking behaviour in early childhood, which leads to severe early-onset obesity. With the advent of novel pharmacological treatment options emerging for monogenic obesity syndromes that target the central melanocortin pathway, genetic testing is recommended for patients with rapid weight gain in infancy and additional clinical suggestive features. Likewise, patients with obesity associated with hypothalamic damage or other forms of syndromic obesity involving energy regulatory circuits could benefit from these novel pharmacological treatment options. Early identification of patients affected by syndromic obesity will lead to appropriate treatment, thereby preventing the development of obesity sequelae, avoiding failure of conservative treatment approaches and alleviating stigmatization of patients and their families.
Collapse
Affiliation(s)
- Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy and University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Antje Körner
- Leipzig University, Medical Faculty, Hospital for Children and Adolescents, Centre of Paediatric Research (CPL), Leipzig, Germany
- LIFE Child, Leipzig Research Centre for Civilization Diseases, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | | |
Collapse
|
17
|
Abstract
Gestational diabetes mellitus (GDM) traditionally refers to abnormal glucose tolerance with onset or first recognition during pregnancy. GDM has long been associated with obstetric and neonatal complications primarily relating to higher infant birthweight and is increasingly recognized as a risk factor for future maternal and offspring cardiometabolic disease. The prevalence of GDM continues to rise internationally due to epidemiological factors including the increase in background rates of obesity in women of reproductive age and rising maternal age and the implementation of the revised International Association of the Diabetes and Pregnancy Study Groups' criteria and diagnostic procedures for GDM. The current lack of international consensus for the diagnosis of GDM reflects its complex historical evolution and pragmatic antenatal resource considerations given GDM is now 1 of the most common complications of pregnancy. Regardless, the contemporary clinical approach to GDM should be informed not only by its short-term complications but also by its longer term prognosis. Recent data demonstrate the effect of early in utero exposure to maternal hyperglycemia, with evidence for fetal overgrowth present prior to the traditional diagnosis of GDM from 24 weeks' gestation, as well as the durable adverse impact of maternal hyperglycemia on child and adolescent metabolism. The major contribution of GDM to the global epidemic of intergenerational cardiometabolic disease highlights the importance of identifying GDM as an early risk factor for type 2 diabetes and cardiovascular disease, broadening the prevailing clinical approach to address longer term maternal and offspring complications following a diagnosis of GDM.
Collapse
Affiliation(s)
- Arianne Sweeting
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Jencia Wong
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Helen R Murphy
- Diabetes in Pregnancy Team, Cambridge University Hospitals, Cambridge, UK
- Norwich Medical School, Bob Champion Research and Education Building, University of East Anglia, Norwich, UK
- Division of Women’s Health, Kings College London, London, UK
| | - Glynis P Ross
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Močnik M, Zagradišnik B, Marčun Varda N. Assessing 48 SNPs in Hypertensive Paediatric Patients and Young Adults with Review of Genetic Background of Essential Hypertension. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1262. [PMID: 36010152 PMCID: PMC9406300 DOI: 10.3390/children9081262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
Essential hypertension in paediatric patients and young adults is rising, mostly on account of obesity-related hypertension. Clinically, the difference between obese hypertensive and non-obese hypertensive individuals is evident; yet, the pathophysiology of essential and obesity-related hypertension is multifactorial, complex and not fully understood. The aim of our study was to obtain a comprehensive view of the clinical differences between obesity-related hypertension and hypertension in non-obese paediatric patients and young adults and to do genetic tests to possibly highlight some of the pathophysiological differences with a review of their genetic backgrounds. Four hundred and thirty-six hypertensive paediatric patients and young adults were included in the study, and a study of 48 single-nucleotide polymorphisms, using Kompetitive allele specific PCR, was conducted. The subjects were divided into 243 non-obese participants with hypertension and 193 obese participants with hypertension. The data for the clinical comparison of both groups were collected as well. The differences in some clinical and biochemical parameters were confirmed. Genetic tests showed a significant difference in one allele frequency between both groups in five SNPs: rs6232, rs6235, rs12145833, rs59744560 and rs9568856. In rs6235 and rs59744560, a direct effect of different allele states could be implied. Obesity-related hypertension at a young age differs from essential hypertension in those non-obese. The reported genetic differences could be important in understanding the complex pathophysiology of early-onset obesity-related hypertension and should be further evaluated.
Collapse
Affiliation(s)
- Mirjam Močnik
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
| | - Boris Zagradišnik
- Laboratory of Medical Genetics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
- Medical Faculty, University of Maribor, Taborska 8, 2000 Maribor, Slovenia
| |
Collapse
|
19
|
Meier DT, Rachid L, Wiedemann SJ, Traub S, Trimigliozzi K, Stawiski M, Sauteur L, Winter DV, Le Foll C, Brégère C, Guzman R, Odermatt A, Böni-Schnetzler M, Donath MY. Prohormone convertase 1/3 deficiency causes obesity due to impaired proinsulin processing. Nat Commun 2022; 13:4761. [PMID: 35963866 PMCID: PMC9376086 DOI: 10.1038/s41467-022-32509-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
Defective insulin processing is associated with obesity and diabetes. Prohormone convertase 1/3 (PC1/3) is an endopeptidase required for the processing of neurotransmitters and hormones. PC1/3 deficiency and genome-wide association studies relate PC1/3 with early onset obesity. Here, we find that deletion of PC1/3 in obesity-related neuronal cells expressing proopiomelanocortin mildly and transiently change body weight and fail to produce a phenotype when targeted to Agouti-related peptide- or nestin-expressing tissues. In contrast, pancreatic β cell-specific PC1/3 ablation induces hyperphagia with consecutive obesity despite uncontrolled diabetes with glucosuria. Obesity develops not due to impaired pro-islet amyloid polypeptide processing but due to impaired insulin maturation. Proinsulin crosses the blood-brain-barrier but does not induce central satiety. Accordingly, insulin therapy prevents hyperphagia. Further, islet PC1/3 expression levels negatively correlate with body mass index in humans. In this work, we show that impaired PC1/3-mediated proinsulin processing, as observed in human prediabetes, promotes hyperphagic obesity. Defective insulin secretion is observed early in the development of diabetes. Here the authors report that β cell-specific deficiency of the insulin prohormone convertase 1/3 (PC1/3) leads not only to hyperglycemia, but also to hyperphagic obesity in mice.
Collapse
Affiliation(s)
- Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland. .,Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Leila Rachid
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Sophia J Wiedemann
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Shuyang Traub
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Kelly Trimigliozzi
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Stawiski
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Loïc Sauteur
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Denise V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, 8057, Zurich, Switzerland
| | - Catherine Brégère
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University of Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University of Basel, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Marianne Böni-Schnetzler
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
Nawaz S, Chinnadurai R, Al Chalabi S, Evans P, Kalra PA, Syed AA, Sinha S. Obesity and Chronic Kidney Disease A Current Review. Obes Sci Pract 2022; 9:61-74. [PMID: 37034567 PMCID: PMC10073820 DOI: 10.1002/osp4.629] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/02/2022] [Accepted: 07/04/2022] [Indexed: 11/08/2022] Open
Abstract
Background Obesity poses significant challenges to healthcare globally, particularly through its bi-directional relationship with co-morbid metabolic conditions such as type 2 diabetes and hypertension. There is also emerging evidence of an association between obesity and chronic kidney disease (CKD) which is less well characterized. Methods A literature search of electronic libraries was conducted to identify and present a narrative review of the interplay between obesity and CKD. Findings Obesity may predispose to CKD directly as it is linked to the histopathological finding of obesity-related glomerulopathy and indirectly through its widely recognized complications such as atherosclerosis, hypertension, and type 2 diabetes. The biochemical and endocrine products of adipose tissue contribute to pathophysiological processes such as inflammation, oxidative stress, endothelial dysfunction, and proteinuria. The prevention and management of obesity may prove critical in counteracting both the development and advancement of CKD. Moreover, measures of abdominal adiposity such as waist circumference, are generally associated with worse morbidity and mortality in individuals receiving maintenance hemodialysis. Conclusion Obesity is a risk factor for the onset and progression of CKD and should be recognized as a potential target for a preventative public health approach to reduce CKD rates within the general population. Future research should focus on the use of glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors in patients with CKD and obesity due to their multi-faceted actions on major outcomes.
Collapse
Affiliation(s)
- Saira Nawaz
- Faculty of Biology Medicine and Health University of Manchester Manchester UK
| | - Rajkumar Chinnadurai
- Faculty of Biology Medicine and Health University of Manchester Manchester UK
- Department of Renal Medicine Salford Royal Hospital Northern Care Alliance NHS Foundation Trust Salford UK
| | - Saif Al Chalabi
- Faculty of Biology Medicine and Health University of Manchester Manchester UK
- Department of Renal Medicine Salford Royal Hospital Northern Care Alliance NHS Foundation Trust Salford UK
| | - Philip Evans
- Department of Renal Medicine Liverpool University Hospitals NHS Foundation Trust Liverpool UK
| | - Philip A Kalra
- Faculty of Biology Medicine and Health University of Manchester Manchester UK
- Department of Renal Medicine Salford Royal Hospital Northern Care Alliance NHS Foundation Trust Salford UK
| | - Akheel A. Syed
- Faculty of Biology Medicine and Health University of Manchester Manchester UK
- Department of Diabetes Endocrinology and Obesity Medicine Salford Royal Hospital Northern Care Alliance NHS Foundation Trust Salford UK
| | - Smeeta Sinha
- Faculty of Biology Medicine and Health University of Manchester Manchester UK
- Department of Renal Medicine Salford Royal Hospital Northern Care Alliance NHS Foundation Trust Salford UK
| |
Collapse
|
21
|
Roberts KJ, Ariza AJ, Selvaraj K, Quadri M, Mangarelli C, Neault S, Davis EE, Binns HJ. Testing for rare genetic causes of obesity: findings and experiences from a pediatric weight management program. Int J Obes (Lond) 2022; 46:1493-1501. [PMID: 35562395 PMCID: PMC9105591 DOI: 10.1038/s41366-022-01139-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Genetic screening for youth with obesity in the absence of syndromic findings has not been part of obesity management. For children with early onset obesity, genetic screening is recommended for those having clinical features of genetic obesity syndromes (including hyperphagia). OBJECTIVES The overarching goal of this work is to report the findings and experiences from one pediatric weight management program that implemented targeted sequencing analysis for genes known to cause rare genetic disorders of obesity. SUBJECTS/METHODS This exploratory study evaluated youth tested over an 18-month period using a panel of 40-genes in the melanocortin 4 receptor pathway. Medical records were reviewed for demographic and visit information, including body mass index (BMI) percent of 95th percentile (%BMIp95) and two eating behaviors. RESULTS Of 117 subjects: 51.3% were male; 53.8% Hispanic; mean age 10.2 years (SD 3.8); mean %BMIp95 157% (SD 29%). Most subjects were self- or caregiver-reported to have overeating to excess or binge eating (80.3%) and sneaking food or eating in secret (59.0%). Among analyzed genes, 72 subjects (61.5%) had at least one variant reported; 50 (42.7%) had a single variant reported; 22 (18.8%) had 2-4 variants reported; most variants were rare (<0.05% minor allele frequency [MAF]), and of uncertain significance; all variants were heterozygous. Nine subjects (7.7%) had a variant reported as PSCK1 "risk" or MC4R "likely pathogenic"; 39 (33.3%) had a Bardet-Biedl Syndrome (BBS) gene variant (4 with "pathogenic" or "likely pathogenic" variants). Therefore, 9 youth (7.7%) had gene variants previously identified as increasing risk for obesity and 4 youth (3.4%) had BBS carrier status. CONCLUSIONS Panel testing identified rare variants of uncertain significance in most youth tested, and infrequently identified variants previously reported to increase the risk for obesity. Further research in larger cohorts is needed to understand how genetic variants influence the expression of non-syndromic obesity.
Collapse
Affiliation(s)
- Karyn J Roberts
- College of Nursing, University of Wisconsin-Milwaukee, PO Box 413, Milwaukee, WI, 53201-0413, USA. .,Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Adolfo J Ariza
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Kavitha Selvaraj
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Maheen Quadri
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Caren Mangarelli
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Sarah Neault
- Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Erica E Davis
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Helen J Binns
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
22
|
Ahamed SK, Barek MA, Roy UK, Kouser M, Reza MS, Mannan AB, Alam MA, Uddin SMN. A review on association and correlation of genetic variants with eating disorders and obesity. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Now, eating disorders and obesity and their correlations are danger signal in worldwide which is caused by multifactor and associated with significant mortality and morbidity.
Main body
Every aspect of a patient’s life is influenced by eating disorders and obesity and their correlations. Due to frequent seeing of obese patients, eating disorders have been included in the review as they can sometimes be associated with obesity. However, it should be noted that most patients having eating disorder are at risk to be obese or overweight. This research explores the risk factors for the two disorders, as well as the assessment of medical complications and treatment recommendations for the disorders. In these two disorders, there is also a correlation. The essential consideration is that eating disorders are impulse-control disorders which are similar to addictive behaviors in some aspects. So it is a crying need to treat a patient with obesity and eating disorders simultaneously to ensure success. Genome-wide association studies (GWASs) have increased our knowledge of the pathophysiology of eating disorders (EDs) and obesity and their correlation.
Conclusion
This review enlightens on the summary of eating disorder, obesity, genotypic traits, molecular relations, interaction, correlation, and effect of eating disorder and obesity which outline potential future directions and clinical implications for patients with EDs and obesity.
Collapse
|
23
|
Duclaux-Loras R, Bourgeois P, Lavrut PM, Charbit-Henrion F, Bonniaud-Blot P, Maudinas R, Bournez M, Faure M, Cerf-Bensussan N, Lachaux A, Peretti N, Fabre A. A novel mutation of PCSK1 responsible for PC1/3 deficiency in two siblings. Clin Res Hepatol Gastroenterol 2021; 45:101640. [PMID: 33662777 DOI: 10.1016/j.clinre.2021.101640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 10/09/2020] [Accepted: 01/15/2021] [Indexed: 02/04/2023]
Abstract
Proprotein convertase 1 (PCSK1, PC1/3) deficiency is an uncommon cause of neonatal malabsorptive diarrhoea associated with endocrinopathies that are due to the disrupted processing of a large number of prohormones, including proinsulin. To date, only 26 cases have been reported. Herein, we describe two siblings with typical features including severe congenital diarrhoea, central diabetes insipidus, growth hormone deficiency, and hypoadrenalism. Next generation sequencing found a homozygous missense mutation in exon 5 of PCSK1 gene, c.500A>C (p.Asp167Ala), located within the catalytic domain. Both patients presented a high level of proinsulin. In the first years of life they required parenteral nutrition and hormone replacement therapy. The patients, aged 3 and 1.5 years, experienced several infectious episodes associated with septic shocks. While the mechanism underlying intestinal failure remains poorly investigated, parenteral nutrition is essential in order to ensure normal growth in early childhood.
Collapse
Affiliation(s)
- Rémi Duclaux-Loras
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service de Gastroentérologie, Hépathologie et Nutrition Pédiatrique, Bron, France; INSERM U1111, Centre International de Recherche en Infectiologie, Lyon, France.
| | - Patrice Bourgeois
- Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Pierre-Marie Lavrut
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Anatomopathologie, Bron, France
| | - Fabienne Charbit-Henrion
- Service de Génétique Moléculaire, Necker-Enfants Malades Hospital, Assistance Publique des Hôpitaux de Paris, Université de Paris et Institut Imagine, Inserm UMR1163 Intestinal Immunity, Paris, France
| | | | | | | | - Mathias Faure
- INSERM U1111, Centre International de Recherche en Infectiologie, Lyon, France
| | - Nadine Cerf-Bensussan
- Université de Paris et Institut Imagine, Inserm UMR1163 Intestinal Immunity, Paris, France
| | - Alain Lachaux
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service de Gastroentérologie, Hépathologie et Nutrition Pédiatrique, Bron, France
| | - Noel Peretti
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service de Gastroentérologie, Hépathologie et Nutrition Pédiatrique, Bron, France
| | - Alexandre Fabre
- Service de Pédiatrie Multidisciplinaire, Hôpital de la Timone Enfants, APHM, 13385 Marseille, France
| |
Collapse
|
24
|
Courbage S, Poitou C, Le Beyec-Le Bihan J, Karsenty A, Lemale J, Pelloux V, Lacorte JM, Carel JC, Lecomte N, Storey C, De Filippo G, Coupaye M, Oppert JM, Tounian P, Clément K, Dubern B. Implication of Heterozygous Variants in Genes of the Leptin-Melanocortin Pathway in Severe Obesity. J Clin Endocrinol Metab 2021; 106:2991-3006. [PMID: 34097736 DOI: 10.1210/clinem/dgab404] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Unlike homozygous variants, the implication of heterozygous variants on the leptin-melanocortin pathway in severe obesity has not been established. OBJECTIVE To describe the frequency, the phenotype, and the genotype-phenotype relationship for heterozygous variants in LEP, LEPR, POMC, and PCSK1 in severe obesity. METHODS In this retrospective study, genotyping was performed on at least 1 of the LEP, LEPR, POMC, and PCSK1 genes in 1486 probands with severe obesity (600 children, 886 adults). The phenotype was collected in 60 subjects with heterozygous variants and 16 with homozygous variants. We analyzed variant frequency, body mass index (BMI), age of obesity onset, food impulsivity, and endocrine abnormalities. RESULTS The frequency of subjects with homozygous variants was 1.7% (n = 26), and 6.7% (n = 100) with heterozygous variants. Adults with homozygous variants had a higher BMI (66 vs 53 kg/m2, P = .015), an earlier onset of obesity (0.4 vs 5.4 years, P < .001), more often food impulsivity (83% vs 42%, P = .04), and endocrine abnormalities (75% vs 26%, P < .01). The BMI was higher for subjects with high-impact heterozygous variants (61 vs 50 kg/m², P = .045) and those with a second heterozygous variant on the pathway (65 vs 49 kg/m², P < .01). In children, no significant differences were found for the age of obesity onset and BMI. CONCLUSION Heterozygous variants in LEP, LEPR, POMC, and PCSK1 are frequent in severe obesity and sometimes associated with a phenotype close to that of homozygotes. These data suggest a systematic search for variants in severe early-onset obesity, to discuss therapy that targets this key pathway.
Collapse
Affiliation(s)
- Sophie Courbage
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Pediatric Nutrition and Gastroenterology Department, Armand-Trousseau Hospital, Sorbonne University, Paris, France
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics) Research Unit, Paris, France
| | - Christine Poitou
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics) Research Unit, Paris, France
| | - Johanne Le Beyec-Le Bihan
- Assistance Publique-Hôpitaux de Paris (AP-HP), Endocrine and Oncological Biochemistry Department, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Alexandra Karsenty
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Pediatric Nutrition and Gastroenterology Department, Armand-Trousseau Hospital, Sorbonne University, Paris, France
| | - Julie Lemale
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Pediatric Nutrition and Gastroenterology Department, Armand-Trousseau Hospital, Sorbonne University, Paris, France
| | - Véronique Pelloux
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics) Research Unit, Paris, France
| | - Jean-Marc Lacorte
- Assistance Publique-Hôpitaux de Paris (AP-HP), Endocrine and Oncological Biochemistry Department, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Jean-Claude Carel
- Université de Paris, F-75019, Paris, France; AP-HP.Nord Université de Paris. Hôpital Universitaire Robert-Debré, Service d'Endocrinologie Diabétologie Pédiatrique F-75019, Paris, France
| | - Nathalie Lecomte
- Université de Paris, F-75019, Paris, France; AP-HP.Nord Université de Paris. Hôpital Universitaire Robert-Debré, Service d'Endocrinologie Diabétologie Pédiatrique F-75019, Paris, France
| | - Caroline Storey
- Université de Paris, F-75019, Paris, France; AP-HP.Nord Université de Paris. Hôpital Universitaire Robert-Debré, Service d'Endocrinologie Diabétologie Pédiatrique F-75019, Paris, France
| | - Gianpaolo De Filippo
- Université de Paris, F-75019, Paris, France; AP-HP.Nord Université de Paris. Hôpital Universitaire Robert-Debré, Service d'Endocrinologie Diabétologie Pédiatrique F-75019, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Bicêtre Hospital, Medicine for Adolescents Department, Le Kremlin-Bicêtre, France
| | - Muriel Coupaye
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Explorations Fonctionnelles Department, Louis-Mourier Hospital, Centre Intégré Nord Francilien de l'Obésité (CINFO) and Université de Paris, Centre de Recherche sur l'Inflammation, Inserm UMRS 1149, Paris, France
| | - Jean-Michel Oppert
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
| | - Patrick Tounian
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Pediatric Nutrition and Gastroenterology Department, Armand-Trousseau Hospital, Sorbonne University, Paris, France
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics) Research Unit, Paris, France
| | - Karine Clément
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics) Research Unit, Paris, France
| | - Béatrice Dubern
- Assistance Publique-Hôpitaux de Paris (AP-HP), Reference Centre for Rare Diseases (PRADORT, Prader-Willi Syndrome and other Rare Forms of Obesity with Eating Behavior Disorders), Pediatric Nutrition and Gastroenterology Department, Armand-Trousseau Hospital, Sorbonne University, Paris, France
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics) Research Unit, Paris, France
| |
Collapse
|
25
|
Тимашева ЯР, Балхиярова ЖР, Кочетова ОВ. [Current state of the obesity research: genetic aspects, the role of microbiome, and susceptibility to COVID-19]. PROBLEMY ENDOKRINOLOGII 2021; 67:20-35. [PMID: 34533011 PMCID: PMC9753850 DOI: 10.14341/probl12775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 11/06/2022]
Abstract
Obesity affects over 700 million people worldwide and its prevalence keeps growing steadily. The problem is particularly relevant due to the increased risk of COVID-19 complications and mortality in obese patients. Obesity prevalence increase is often associated with the influence of environmental and behavioural factors, leading to stigmatization of people with obesity due to beliefs that their problems are caused by poor lifestyle choices. However, hereditary predisposition to obesity has been established, likely polygenic in nature. Morbid obesity can result from rare mutations having a significant effect on energy metabolism and fat deposition, but the majority of patients does not present with monogenic forms. Microbiome low diversity significantly correlates with metabolic disorders (inflammation, insulin resistance), and the success of weight loss (bariatric) surgery. However, data on the long-term consequences of bariatric surgery and changes in the microbiome composition and genetic diversity before and after surgery are currently lacking. In this review, we summarize the results of studies of the genetic characteristics of obesity patients, molecular mechanisms of obesity, contributing to the unfavourable course of coronavirus infection, and the evolution of their microbiome during bariatric surgery, elucidating the mechanisms of disease development and creating opportunities to identify potential new treatment targets and design effective personalized approaches for the diagnosis, management, and prevention of obesity.
Collapse
Affiliation(s)
- Я. Р. Тимашева
- Институт биохимии и генетики Уфимского федерального исследовательского центра Российской академии наук;
Башкирский государственный медицинский университет
| | - Ж. Р. Балхиярова
- Институт биохимии и генетики Уфимского федерального исследовательского центра Российской академии наук;
Башкирский государственный медицинский университет;
Университет Суррея
| | - О. В. Кочетова
- Институт биохимии и генетики Уфимского федерального исследовательского центра Российской академии наук
| |
Collapse
|
26
|
Ramzy A, Kieffer TJ. Altered islet prohormone processing: A cause or consequence of diabetes? Physiol Rev 2021; 102:155-208. [PMID: 34280055 DOI: 10.1152/physrev.00008.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peptide hormones are first produced as larger precursor prohormones that require endoproteolytic cleavage to liberate the mature hormones. A structurally conserved but functionally distinct family of nine prohormone convertase enzymes (PCs) are responsible for cleavage of protein precursors of which PC1/3 and PC2 are known to be exclusive to neuroendocrine cells and responsible for prohormone cleavage. Differential expression of PCs within tissues define prohormone processing; whereas glucagon is the major product liberated from proglucagon via PC2 in pancreatic α-cells, proglucagon is preferentially processed by PC1/3 in intestinal L cells to produce glucagon-like peptides 1 and 2 (GLP-1, GLP-2). Beyond our understanding of processing of islet prohormones in healthy islets, there is convincing evidence that proinsulin, proIAPP, and proglucagon processing is altered during prediabetes and diabetes. There is predictive value of elevated circulating proinsulin or proinsulin : C-peptide ratio for progression to type 2 diabetes and elevated proinsulin or proinsulin : C-peptide is predictive for development of type 1 diabetes in at risk groups. After onset of diabetes, patients have elevated circulating proinsulin and proIAPP and proinsulin may be an autoantigen in type 1 diabetes. Further, preclinical studies reveal that α-cells have altered proglucagon processing during diabetes leading to increased GLP-1 production. We conclude that despite strong associative data, current evidence is inconclusive on the potential causal role of impaired prohormone processing in diabetes, and suggest that future work should focus on resolving the question of whether altered prohormone processing is a causal driver or merely a consequence of diabetes pathology.
Collapse
Affiliation(s)
- Adam Ramzy
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Chauhdary Z, Rehman K, Akash MSH. The composite alliance of FTO locus with obesity-related genetic variants. Clin Exp Pharmacol Physiol 2021; 48:954-965. [PMID: 33735452 DOI: 10.1111/1440-1681.13498] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/05/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
Obesity has become a genuine global pandemic due to lifestyle and environmental modifications, and is associated with chronic lethal comorbidities. Various environmental factors such as lack of physical activity due to modernization and higher intake of energy-rich diets are primary obesogenic factors in pathogenesis of obesity. Genome-wide association study has identified the crucial role of FTO (fat mass and obesity) in human obesity. A bunch of SNPs in the first intron of FTO has been identified and subsequently correlated to body mass index and body composition. Findings of in silico, in vitro, and in vivo studies have manifested the robust role of FTO in regulation of energy expenditure and food consumption. Numerous studies have highlighted the mechanistic pathways behind the concomitant functions of FTO in adipogenesis and body size. Current investigation has also revealed the link of FTO neighbouring genes i.e., RPGRIP1L, IRX3 and IRX5 and epigenetic factors with obesity phenotypes. The motive behind this review is to cite the consequences of FTO on obesity vulnerability.
Collapse
Affiliation(s)
- Zunera Chauhdary
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
28
|
Abstract
The kexin-like proprotein convertases perform the initial proteolytic cleavages that ultimately generate a variety of different mature peptide and proteins, ranging from brain neuropeptides to endocrine peptide hormones, to structural proteins, among others. In this review, we present a general introduction to proprotein convertase structure and biochemistry, followed by a comprehensive discussion of each member of the kexin-like subfamily of proprotein convertases. We summarize current knowledge of human proprotein convertase insufficiency syndromes, including genome-wide analyses of convertase polymorphisms, and compare these to convertase null and mutant mouse models. These mouse models have illuminated our understanding of the roles specific convertases play in human disease and have led to the identification of convertase-specific substrates; for example, the identification of procorin as a specific PACE4 substrate in the heart. We also discuss the limitations of mouse null models in interpreting human disease, such as differential precursor cleavage due to species-specific sequence differences, and the challenges presented by functional redundancy among convertases in attempting to assign specific cleavages and/or physiological roles. However, in most cases, knockout mouse models have added substantively both to our knowledge of diseases caused by human proprotein convertase insufficiency and to our appreciation of their normal physiological roles, as clearly seen in the case of the furin, proprotein convertase 1/3, and proprotein convertase 5/6 mouse models. The creation of more sophisticated mouse models with tissue- or temporally-restricted expression of specific convertases will improve our understanding of human proprotein convertase insufficiency and potentially provide support for the emerging concept of therapeutic inhibition of convertases.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Ahmed ABM, Alsaleem BMR. Enteroendocrine Dysfunction in Two Saudi Sisters. Case Rep Gastroenterol 2021; 15:290-295. [PMID: 33790717 PMCID: PMC7989775 DOI: 10.1159/000511761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/18/2020] [Indexed: 12/29/2022] Open
Abstract
Proprotein convertase (PC) deficiency is a rare autosomal recessive disorder caused by mutations in proprotein convertase subtilisin/kexin type 1 (PCSK1). It is characterized by severe malabsorptive early-onset diarrhea, obesity, and systemic endocrinopathies. Only few cases have been reported in the literature; we have add two female sisters with some difference in clinical progress. Herein, we describe two sisters with congenital osmotic diarrhea diagnosed with PC1/3 deficiency, causing malabsorptive diarrhea and enteroendocrine dysfunction, who presented with chronic enteropathy with hypernatremia but with different expressivity. PC1/3 deficiency presents with symptoms and signs that mimic glucose-galactose malabsorption. Because of the clinical paucity and heterogeneity of congenital enteropathies, whole-exome sequencing may be of great help towards early diagnosis and effective treatment.
Collapse
Affiliation(s)
- Amna Basheer M Ahmed
- Department of Pediatric Gastroenterology/Hepatology, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Badr M Rasheed Alsaleem
- Department of Pediatric Gastroenterology/Hepatology, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Chalmers J, Tung YCL, Liu CH, O'Kane CJ, O'Rahilly S, Yeo GSH. A multicomponent screen for feeding behaviour and nutritional status in Drosophila to interrogate mammalian appetite-related genes. Mol Metab 2021; 43:101127. [PMID: 33242659 PMCID: PMC7753202 DOI: 10.1016/j.molmet.2020.101127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE More than 300 genetic variants have been robustly associated with measures of human adiposity. Highly penetrant mutations causing human obesity do so largely by disrupting satiety pathways in the brain and increasing food intake. Most of the common obesity-predisposing variants are in, or near, genes expressed highly in the brain, but little is known of their function. Exploring the biology of these genes at scale in mammalian systems is challenging. We sought to establish and validate the use of a multicomponent screen for feeding behaviour phenotypes, taking advantage of the tractable model organism Drosophila melanogaster. METHODS We validated a screen for feeding behaviour in Drosophila by comparing results after disrupting the expression of centrally expressed genes that influence energy balance in flies to those of 10 control genes. We then used this screen to explore the effects of disrupted expression of genes either a) implicated in energy homeostasis through human genome-wide association studies (GWAS) or b) expressed and nutritionally responsive in specific populations of hypothalamic neurons with a known role in feeding/fasting. RESULTS Using data from the validation study to classify responses, we studied 53 Drosophila orthologues of genes implicated by human GWAS in body mass index and found that 15 significantly influenced feeding behaviour or energy homeostasis in the Drosophila screen. We then studied 50 Drosophila homologues of 47 murine genes reciprocally nutritionally regulated in POMC and agouti-related peptide neurons. Seven of these 50 genes were found by our screen to influence feeding behaviour in flies. CONCLUSION We demonstrated the utility of Drosophila as a tractable model organism in a high-throughput genetic screen for food intake phenotypes. This simple, cost-efficient strategy is ideal for high-throughput interrogation of genes implicated in feeding behaviour and obesity in mammals and will facilitate the process of reaching a functional understanding of obesity pathogenesis.
Collapse
Affiliation(s)
- J Chalmers
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Y C L Tung
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - C H Liu
- Department of Physiology, Development and Neuroscience, Cambridge University, Downing St, Cambridge, CB2 3EG, UK.
| | - C J O'Kane
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK.
| | - S O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - G S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
31
|
Diels S, Vanden Berghe W, Van Hul W. Insights into the multifactorial causation of obesity by integrated genetic and epigenetic analysis. Obes Rev 2020; 21:e13019. [PMID: 32170999 DOI: 10.1111/obr.13019] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/24/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Obesity is a highly heritable multifactorial disease that places an enormous burden on human health. Its increasing prevalence and the concomitant-reduced life expectancy has intensified the search for new analytical methods that can reduce the knowledge gap between genetic susceptibility and functional consequences of the disease pathology. Although the influence of genetics and epigenetics has been studied independently in the past, there is increasing evidence that genetic variants interact with environmental factors through epigenetic regulation. This suggests that a combined analysis of genetic and epigenetic variation may be more effective in characterizing the obesity phenotype. To date, limited genome-wide integrative analyses have been performed. In this review, we provide an overview of the latest findings, advantages, and challenges and discuss future perspectives.
Collapse
Affiliation(s)
- Sara Diels
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Wim Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
32
|
El Hajj Chehadeh S, Osman W, Nazar S, Jerman L, Alghafri A, Sajwani A, Alawlaqi M, AlObeidli M, Jelinek HF, AlAnouti F, Khalaf K, Alsafar H. Implication of genetic variants in overweight and obesity susceptibility among the young Arab population of the United Arab Emirates. Gene 2020; 739:144509. [PMID: 32109558 DOI: 10.1016/j.gene.2020.144509] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Overweight and obesity are major risk factors for Type 2 Diabetes Mellitus (T2DM), cardiovascular disease (CVD) and cancer. Genetic predisposition has been shown to play a key role in obesity, and genome-wide association studies (GWAS) have identified multiple loci linked with obesity in various ethnic groups. The aim of this study was to validate the reported genetic variants associated with obesity and overweight in a young UAE Arab population. METHODS Twenty-two associated single nucleotide polymorphisms (SNPs) at 11 loci (FTO, MC4R, TMEM18, KCTD15, MTCH2, SH2B1, TFAP2B, GNPDA2, NEGR1, PCSK1 and BDNF) were studied in 392 controls and 318 overweight/obese young Emiratis (aged 18-35 years). RESULTS After adjusting for age and smoking, rs3751812 of the FTO gene was associated with overweight/obesity in male participants (p-value < 0.016), while SNPs rs17782313, rs571312 of the MC4R gene and rs12463617 of the TMEM18 gene were significantly associated with overweight/obesity in female participants (p-value = 0.001, 0.028, 0.044, respectively). Follow-up association tests and logistic regression revealed the contribution of the FTO rs3751812 and MC4R rs571213 SNPs to the risk of overweight/obesity after adjusting for age, sex and smoking (p-value = 0.044, 0.049, respectively). In addition, the FTO rs3751812 was associated with the risk of overweight/obesity after adjusting for the effect of other markers (rs17782313, rs571312, rs2867125, rs6548238 and rs12463617) (p-value = 0.035). A significant gene-gene interaction was seen between FTO, MCR4 and TMEM18 (p-value = 0.013). CONCLUSIONS Our data demonstrates that rs3751812 of the FTO gene is the key SNP associated with risk of overweight/obesity among the young UAE Arab population, in alignment with previous findings. Our results also indicate that the identified genes stratify with sex and risk of overweight/obesity. In addition to their direct association with overweight/obesity, rs17782313 and rs571312, as well as rs2867125 and rs6548238, may have a modifying effect on the risk of overweight/obesity caused by the rs3751812. Population-specific, sex-specific genetic profiling is important in understanding the heritability of obesity.
Collapse
Affiliation(s)
| | - Wael Osman
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; College of Arts and Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Suna Nazar
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Laila Jerman
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ameera Alghafri
- College of Medicine, Mohammad Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Ali Sajwani
- College of Medicine, Mohammad Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mohamed Alawlaqi
- School of Medicine, The Royal College of Surgeons, Dublin, Ireland
| | - Mohamed AlObeidli
- College of Medicine and Health Sciences, United Arab Emirates University, AlAin, United Arab Emirates
| | - Herbert F Jelinek
- School of Community Health, Charles Sturt University, Albury, Australia; Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Fatme AlAnouti
- College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Kinda Khalaf
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Habiba Alsafar
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
33
|
Muhsin NIA, Bentley L, Bai Y, Goldsworthy M, Cox RD. A novel mutation in the mouse Pcsk1 gene showing obesity and diabetes. Mamm Genome 2020; 31:17-29. [PMID: 31974728 PMCID: PMC7060156 DOI: 10.1007/s00335-020-09826-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022]
Abstract
The proprotein convertase subtilisin/Kexin type 1 (PCSK1/PC1) protein processes inactive pro-hormone precursors into biologically active hormones in a number of neuroendocrine and endocrine cell types. Patients with recessive mutations in PCSK1 exhibit a complex spectrum of traits including obesity, diarrhoea and endocrine disorders. We describe here a new mouse model with a point mutation in the Pcsk1 gene that exhibits obesity, hyperphagia, transient diarrhoea and hyperproinsulinaemia, phenotypes consistent with human patient traits. The mutation results in a pV96L amino acid substitution and changes the first nucleotide of mouse exon 3 leading to skipping of that exon and in homozygotes very little full-length transcript. Overexpression of the exon 3 deleted protein or the 96L protein results in ER retention in Neuro2a cells. This is the second Pcsk1 mouse model to display obesity phenotypes, contrasting knockout mouse alleles. This model will be useful in investigating the basis of endocrine disease resulting from prohormone processing defects.
Collapse
Affiliation(s)
- Nor I A Muhsin
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Liz Bentley
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Ying Bai
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Michelle Goldsworthy
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Roger D Cox
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
34
|
Tam V, Patel N, Turcotte M, Bossé Y, Paré G, Meyre D. Benefits and limitations of genome-wide association studies. Nat Rev Genet 2019; 20:467-484. [PMID: 31068683 DOI: 10.1038/s41576-019-0127-1] [Citation(s) in RCA: 979] [Impact Index Per Article: 195.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies (GWAS) involve testing genetic variants across the genomes of many individuals to identify genotype-phenotype associations. GWAS have revolutionized the field of complex disease genetics over the past decade, providing numerous compelling associations for human complex traits and diseases. Despite clear successes in identifying novel disease susceptibility genes and biological pathways and in translating these findings into clinical care, GWAS have not been without controversy. Prominent criticisms include concerns that GWAS will eventually implicate the entire genome in disease predisposition and that most association signals reflect variants and genes with no direct biological relevance to disease. In this Review, we comprehensively assess the benefits and limitations of GWAS in human populations and discuss the relevance of performing more GWAS.
Collapse
Affiliation(s)
- Vivian Tam
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Nikunj Patel
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Michelle Turcotte
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec City, Québec, Canada.,Department of Molecular Medicine, Laval University, Québec City, Quebec, Canada
| | - Guillaume Paré
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada. .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada. .,Inserm UMRS 954 N-GERE (Nutrition-Genetics-Environmental Risks), University of Lorraine, Faculty of Medicine, Nancy, France.
| |
Collapse
|
35
|
Akıncı A, Türkkahraman D, Tekedereli İ, Özer L, Evren B, Şahin İ, Kalkan T, Çürek Y, Çamtosun E, Döğer E, Bideci A, Güven A, Eren E, Sangün Ö, Çayır A, Bilir P, Törel Ergür A, Ercan O. Novel Mutations in Obesity-related Genes in Turkish Children with Non-syndromic Early Onset Severe Obesity: A Multicentre Study. J Clin Res Pediatr Endocrinol 2019; 11:341-349. [PMID: 30991789 PMCID: PMC6878344 DOI: 10.4274/jcrpe.galenos.2019.2019.0021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Non syndromic monogenic obesity is a rare cause of early onset severe obesity in the childhood period. This form may not be distinguishable from other forms of severe obesity without genetic analysis, particularly if patients do not exibit any physical abnormalities or developmental delay. The aim of this study was to screen 41 different obesity-related genes in children with non-syndromic early onset severe obesity. METHODS Children with severe (body mass index-standard deviation score >3) and early onset (<7 years) obesity were screened by next-generation sequencing based, targeted DNA custom panel for 41 known-obesity-related genes and the results were confirmed by Sanger technique. RESULTS Six novel variants were identified in five candidate genes in seven out of 105 children with severe obesity; two in SIM1 (p.W306C and p.Q36X), one in POMC (p.Y160H), one in PCSK1 (p.W130G fs Ter8), two in MC4R (p.D126E) and one in LEPR (p.Q4H). Additionally, two previously known variations in MC4R were identified in four patients (p.R165W in three, and p.V166I in one). CONCLUSION We identified six novel and four previously described variants in six obesity-related genes in 11 out of 105 childrens with early onset severe obesity. The prevalence of monogenic obesity was 10.4% in our cohort.
Collapse
Affiliation(s)
- Ayşehan Akıncı
- nönü University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Malatya, Turkey,* Address for Correspondence: İnönü University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Malatya, Turkey Phone: +90 532 643 53 50 E-mail:
| | - Doğa Türkkahraman
- Antalya Training and Research Hospital, Clinic of Pediatric Endocrinology and Diabetes, Antalya, Turkey
| | - İbrahim Tekedereli
- İnönü University Faculty of Medicine, Department of Molecular Genetics, Malatya, Turkey
| | - Leyla Özer
- Yüksek İhtisas University Faculty of Medicine, Department of Molecular Genetics, Ankara, Turkey
| | - Bahri Evren
- İnönü University Faculty of Medicine, Department of Endocrinology and Diabetes, Malatya, Turkey
| | - İbrahim Şahin
- İnönü University Faculty of Medicine, Department of Endocrinology and Diabetes, Malatya, Turkey
| | - Tarkan Kalkan
- Antalya Training and Research Hospital, Clinic of Molecular Genetics, Antalya, Turkey
| | - Yusuf Çürek
- Antalya Training and Research Hospital, Clinic of Pediatric Endocrinology and Diabetes, Antalya, Turkey
| | - Emine Çamtosun
- nönü University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Malatya, Turkey
| | - Esra Döğer
- Gazi University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Ankara, Turkey
| | - Aysun Bideci
- Gazi University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Ankara, Turkey
| | - Ayla Güven
- Göztepe Training and Research Hospital, Clinic of Pediatric Endocrinology and Diabetes, İstanbul, Turkey
| | - Erdal Eren
- Uludağ University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Bursa, Turkey
| | - Özlem Sangün
- Başkent University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Adana, Turkey
| | - Atilla Çayır
- Erzurum Training and Reseach Hospital, Clinic of Pediatric Endocrinology and Diabetes, Erzurum, Turkey
| | - Pelin Bilir
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Ankara, Turkey
| | - Ayça Törel Ergür
- Ufuk University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Ankara, Turkey
| | - Oya Ercan
- İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Departments of Pediatric Endocrinology and Diabetes, and Adolescent, İstanbul, Turkey
| |
Collapse
|
36
|
Jarvela TS, Shakya M, Bachor T, White A, Low MJ, Lindberg I. Reduced Stability and pH-Dependent Activity of a Common Obesity-Linked PCSK1 Polymorphism, N221D. Endocrinology 2019; 160:2630-2645. [PMID: 31504391 PMCID: PMC6892424 DOI: 10.1210/en.2019-00418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023]
Abstract
Common mutations in the human prohormone convertase (PC)1/3 gene (PCKSI) are linked to increased risk of obesity. Previous work has shown that the rs6232 single-nucleotide polymorphism (N221D) results in slightly decreased activity, although whether this decrease underlies obesity risk is not clear. We observed significantly decreased activity of the N221D PC1/3 enzyme at the pH of the trans-Golgi network; at this pH, the mutant enzyme was less stable than wild-type enzyme. Recombinant N221D PC1/3 also showed enhanced susceptibility to heat stress. Enhanced susceptibility to tunicamycin-induced endoplasmic reticulum stress was observed in AtT-20/PC2 cell clones in which murine PC1/3 was replaced by human N221D PC1/3, as compared with wild-type human PC1/3. However, N221D PC1/3-expressing AtT-20/PC2 clones processed proopiomelanocortin to α-MSH similarly to wild-type PC1/3. We also generated a CRISPR-edited mouse line expressing the N221D mutation in the PCKSI gene. When homozygous N221D mice were fed either a standard or a high-fat diet, we found no increase in body weight compared with their wild-type sibling controls. Sexual dimorphism was observed in pituitary ACTH for both genotypes, with females exhibiting lower levels of pituitary ACTH. In contrast, hypothalamic α-MSH content for both genotypes was higher in females compared with males. Hypothalamic corticotropin-like intermediate peptide content was higher in wild-type females compared with wild-type, but not N221D, males. Taken together, these data suggest that the increased obesity risk linked to the N221D allele in humans may be due in part to PC1/3-induced loss of resilience to stressors rather than strictly to decreased enzymatic activity on peptide precursors.
Collapse
Affiliation(s)
- Timothy S Jarvela
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Surbhi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Tomas Bachor
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anne White
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
- Correspondence: Iris Lindberg, PhD, Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Room S267, Baltimore, Maryland 21210. E-mail:
| |
Collapse
|
37
|
Rohde K, Keller M, la Cour Poulsen L, Blüher M, Kovacs P, Böttcher Y. Genetics and epigenetics in obesity. Metabolism 2019; 92:37-50. [PMID: 30399374 DOI: 10.1016/j.metabol.2018.10.007] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/15/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022]
Abstract
Obesity is among the most threatening health burdens worldwide and its prevalence has markedly increased over the last decades. Obesity maybe considered a heritable trait. Identifications of rare cases of monogenic obesity unveiled that hypothalamic circuits and the brain-adipose axis play an important role in the regulation of energy homeostasis, appetite, hunger and satiety. For example, mutations in the leptin gene cause obesity through almost unsuppressed overeating. Common (multifactorial) obesity, most likely resulting from a concerted interplay of genetic, epigenetic and environmental factors, is clearly linked to genetic predisposition by multiple risk variants, which, however only account for a minor part of the general BMI variability. Although GWAS opened new avenues in elucidating the complex genetics behind common obesity, understanding the biological mechanisms relative to the specific risk contributing to obesity remains poorly understood. Non-genetic factors such as eating behavior or physical activity strongly modulate the individual risk for developing obesity. These factors may interact with genetic predisposition for obesity through epigenetic mechanisms. Thus, here, we review the current knowledge about monogenic and common (multifactorial) obesity highlighting the important recent advances in our knowledge on how epigenetic regulation is involved in the etiology of obesity.
Collapse
Affiliation(s)
- Kerstin Rohde
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany; University of Oslo, Institute of Clinical Medicine, Oslo 0316, Norway.
| | - Maria Keller
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany.
| | - Lars la Cour Poulsen
- Akershus University Hospital, Department of Clinical Molecular Biology, Medical Division, Lørenskog 1478, Norway.
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig 04103, Germany.
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany.
| | - Yvonne Böttcher
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany; University of Oslo, Institute of Clinical Medicine, Oslo 0316, Norway; Akershus University Hospital, Department of Clinical Molecular Biology, Medical Division, Lørenskog 1478, Norway.
| |
Collapse
|
38
|
Teitelman G. Heterogeneous Expression of Proinsulin Processing Enzymes in Beta Cells of Non-diabetic and Type 2 Diabetic Humans. J Histochem Cytochem 2019; 67:385-400. [PMID: 30759032 DOI: 10.1369/0022155419831641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Although there is evidence indicating transcriptional and functional heterogeneity in human beta cells, it is unclear whether this heterogeneity extends to the expression level of the enzymes that process proinsulin to insulin in beta cells. To address this question, the expression levels of prohormone convertases (PC) 1/3, proprotein convertase 2 (PC2), and carboxypeptidase E (CPE) were determined in immune-stained sections of human pancreas. In non-diabetic donors, the level of proprotein convertase 1/3 (PC1/3) expression varied among beta cells of each islet but the average per islet was similar for all islets of each donor. Although the average PC1/3 expression of all islets examined per sample was unique for each pancreas, donors had similar levels of proinsulin/insulin expression. PC2 expression in beta cells showed less pronounced inter- and intraislet variation while CPE levels were fairly constant. The relationship between PC1/3 and PC2 expression levels was variable among different donors. Type 2 diabetes had an uneven effect on the expression levels of all three enzymes as they decrease only in some islets in a section. These findings suggest the presence of intraislet, but not interislet, variation in the expression of the proinsulin processing enzymes in non-diabetic subjects and a heterogeneous effect of type 2 diabetes on enzyme expression in islets.
Collapse
Affiliation(s)
- Gladys Teitelman
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
39
|
Tam V, Turcotte M, Meyre D. Established and emerging strategies to crack the genetic code of obesity. Obes Rev 2019; 20:212-240. [PMID: 30353704 DOI: 10.1111/obr.12770] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022]
Abstract
Tremendous progress has been made in the genetic elucidation of obesity over the past two decades, driven largely by technological, methodological and organizational innovations. Current strategies for identifying obesity-predisposing loci/genes, including cytogenetics, linkage analysis, homozygosity mapping, admixture mapping, candidate gene studies, genome-wide association studies, custom genotyping arrays, whole-exome sequencing and targeted exome sequencing, have achieved differing levels of success, and the identified loci in aggregate explain only a modest fraction of the estimated heritability of obesity. This review outlines the successes and limitations of these approaches and proposes novel strategies, including the use of exceptionally large sample sizes, the study of diverse ethnic groups and deep phenotypes and the application of innovative methods and study designs, to identify the remaining obesity-predisposing genes. The use of both established and emerging strategies has the potential to crack the genetic code of obesity in the not-too-distant future. The resulting knowledge is likely to yield improvements in obesity prediction, prevention and care.
Collapse
Affiliation(s)
- V Tam
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - M Turcotte
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - D Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
40
|
Harno E, Gali Ramamoorthy T, Coll AP, White A. POMC: The Physiological Power of Hormone Processing. Physiol Rev 2019; 98:2381-2430. [PMID: 30156493 DOI: 10.1152/physrev.00024.2017] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pro-opiomelanocortin (POMC) is the archetypal polypeptide precursor of hormones and neuropeptides. In this review, we examine the variability in the individual peptides produced in different tissues and the impact of the simultaneous presence of their precursors or fragments. We also discuss the problems inherent in accurately measuring which of the precursors and their derived peptides are present in biological samples. We address how not being able to measure all the combinations of precursors and fragments quantitatively has affected our understanding of the pathophysiology associated with POMC processing. To understand how different ratios of peptides arise, we describe the role of the pro-hormone convertases (PCs) and their tissue specificities and consider the cellular processing pathways which enable regulated secretion of different peptides that play crucial roles in integrating a range of vital physiological functions. In the pituitary, correct processing of POMC peptides is essential to maintain the hypothalamic-pituitary-adrenal axis, and this processing can be disrupted in POMC-expressing tumors. In hypothalamic neurons expressing POMC, abnormalities in processing critically impact on the regulation of appetite, energy homeostasis, and body composition. More work is needed to understand whether expression of the POMC gene in a tissue equates to release of bioactive peptides. We suggest that this comprehensive view of POMC processing, with a focus on gaining a better understanding of the combination of peptides produced and their relative bioactivity, is a necessity for all involved in studying this fascinating physiological regulatory phenomenon.
Collapse
Affiliation(s)
- Erika Harno
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| | - Thanuja Gali Ramamoorthy
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| | - Anthony P Coll
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| | - Anne White
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| |
Collapse
|
41
|
Kong X, Xing X, Zhang X, Hong J, Yang W. Sexual Dimorphism of a Genetic Risk Score for Obesity and Related Traits among Chinese Patients with Type 2 Diabetes. Obes Facts 2019; 12:328-343. [PMID: 31167208 PMCID: PMC6696887 DOI: 10.1159/000500490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity is more prevalent in men than in women in China, especially within the middle-aged population. OBJECTIVES The present study aims to determine the contribution of sexual dimorphisms to obesity and related traits in terms of the mechanisms involving the obesity-related genetic variants among patients of Chinese Han ancestry with type 2 diabetes. METHOD In the Chinese National Diabetes and Metabolic Disorders Study, 2,555 out of 4,036 patients with type 2 diabetes were treatment naive, including 1,142 men and 1,413 women. Single-nucleotide polymorphisms (SNP) from 18 genomic loci previously found to be associated with obesity-related traits were successfully genotyped, and a genetic risk score (GRS) was constructed by summing the risk alleles for obesity. RESULTS Single SNP analysis showed that genetic variants in SLC30A10, TMEM18, GNPDA2, PRL, TFAP2B, BDNF, MTCH2, FTO, and MC4R were nominally associated with waist circumference (WC), BMI, and risk for abdominal or general obesity in the untreated patients with type 2 diabetes, as well as in the total group of patients with type 2 diabetes (untreated and treated) (p < 0.05). Interactions between sex and SNP in PRL, MTCH2,and FTO were detected (p < 0.05). In the untreated patients with diabetes, the GRS was nominally associated with WC (β = 0.0032, SE = 0.0011; p = 0.003), BMI (β = 0.0030, SE = 0.0013; p = 0.027), and increased risk for abdominal (OR = 1.08; 95% CI 1.02-1.13; p = 0.004) or general obesity (OR = 1.07; 95% CI 1.02-1.13; p = 0.011) in men but not in women. GRS-sex interactions were detected in the determinant of WC (p = 0.019) and abdominal obesity (p = 0.016). Among patients aged 30-60 years, GRS was found to be significantly associated with WC (β = 0.0050, SE = 0.0016; p = 0.002) and abdominal obesity (OR = 1.10; 95% CI 1.04-1.17; p = 0.001) and nominally associated with BMI (β = 0.0057, SE = 0.0020; p = 0.005) and general obesity (OR = 1.07; 95% CI 1.01-1.14; p = 0.027) in men, whereas in women none of the associations were detected. GRS-sex interactions were present in the determinant of WC (p = 0.015), BMI (p = 0.032), and abdominal obesity (p = 0.012). Among patients aged 60 years or older, neither an association of GRS with obesity-related traits nor GRS-sex interactions were detected. CONCLUSIONS Genetic factors contribute to obesity-related traits in a sex-dependent pattern among middle-aged Chinese, and men tend to be more susceptible to the genetic risk of obesity.
Collapse
Affiliation(s)
- Xiaomu Kong
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoyan Xing
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Xuelian Zhang
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Jing Hong
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China
| | - Wenying Yang
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, China,
| |
Collapse
|
42
|
Andersen MK, Hansen T. Genetics of metabolic traits in Greenlanders: lessons from an isolated population. J Intern Med 2018; 284:464-477. [PMID: 30101502 DOI: 10.1111/joim.12814] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we describe the extraordinary population of Greenland, which differs from large outbred populations of Europe and Asia, both in terms of population history and living conditions. Many years in isolation, small population size and an extreme environment have shaped the genetic composition of the Greenlandic population. The unique genetic background combined with the transition from a traditional Inuit lifestyle and diet, to a more Westernized lifestyle, has led to an increase in the prevalence of metabolic conditions like obesity, where the prevalence from 1993 to 2010 has increased from 16.4% to 19.4% among men, and from 13.0% to 25.4% among women, type 2 diabetes and cardiovascular diseases. The genetic susceptibility to metabolic conditions has been explored in Greenlanders, as well as other isolated populations, taking advantage of population-genetic properties of these populations. During the last 10 years, these studies have provided examples of loci showing evidence of positive selection, due to adaption to Arctic climate and Inuit diet, including TBC1D4 and FADS/CPT1A, and have facilitated the discovery of several loci associated with metabolic phenotypes. Most recently, the c.2433-1G>A loss-of-function variant in ADCY3 associated with obesity and type 2 diabetes was described. This locus has provided novel biological insights, as it has been shown that reduced ADCY3 function causes obesity through disrupted function in primary cilia. Future studies of isolated populations will likely provide further genetic as well as biological insights.
Collapse
Affiliation(s)
- M K Andersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - T Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Chen YC, Taylor AJ, Verchere CB. Islet prohormone processing in health and disease. Diabetes Obes Metab 2018; 20 Suppl 2:64-76. [PMID: 30230179 DOI: 10.1111/dom.13401] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/30/2018] [Accepted: 05/30/2018] [Indexed: 12/15/2022]
Abstract
Biosynthesis of peptide hormones by pancreatic islet endocrine cells is a tightly orchestrated process that is critical for metabolic homeostasis. Like neuroendocrine peptides, insulin and other islet hormones are first synthesized as larger precursor molecules that are processed to their mature secreted products through a series of proteolytic cleavages, mediated by the prohormone convertases Pc1/3 and Pc2, and carboxypeptidase E. Additional posttranslational modifications including C-terminal amidation of the β-cell peptide islet amyloid polypeptide (IAPP) by peptidyl-glycine α-amidating monooxygenase (Pam) may also occur. Genome-wide association studies (GWAS) have showed genetic linkage of these processing enzymes to obesity, β-cell dysfunction, and type 2 diabetes (T2D), pointing to their important roles in metabolism and blood glucose regulation. In both type 1 diabetes (T1D) and T2D, and in the face of metabolic or inflammatory stresses, islet prohormone processing may become impaired; indeed elevated proinsulin:insulin (PI:I) ratios are a hallmark of the β-cell dysfunction in T2D. Recent studies suggest that genetic or acquired defects in proIAPP processing may lead to the production and secretion of incompletely processed forms of proIAPP that could contribute to T2D pathogenesis, and additionally that impaired processing of both PI and proIAPP may be characteristic of β-cell dysfunction in T1D. In islet α-cells, the prohormone proglucagon is normally processed to bioactive glucagon by Pc2 but may express Pc1/3 under certain conditions leading to production of GLP-1(7-36NH2 ). A better understanding of how β-cell processing of PI and proIAPP, as well as α-cell processing of proglucagon, are impacted by genetic susceptibility and in the face of diabetogenic stresses, may lead to new therapeutic approaches for improving islet function in diabetes.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Surgery, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
| | - Austin J Taylor
- Department of Surgery, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
| | - C Bruce Verchere
- Department of Surgery, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
44
|
Ayers KL, Glicksberg BS, Garfield AS, Longerich S, White JA, Yang P, Du L, Chittenden TW, Gulcher JR, Roy S, Fiedorek F, Gottesdiener K, Cohen S, North KE, Schadt EE, Li SD, Chen R, Van der Ploeg LHT. Melanocortin 4 Receptor Pathway Dysfunction in Obesity: Patient Stratification Aimed at MC4R Agonist Treatment. J Clin Endocrinol Metab 2018; 103:2601-2612. [PMID: 29726959 PMCID: PMC7263790 DOI: 10.1210/jc.2018-00258] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/27/2018] [Indexed: 01/03/2023]
Abstract
CONTEXT The hypothalamic melanocortin 4 receptor (MC4R) pathway serves a critical role in regulating body weight. Loss of function (LoF) mutations in the MC4R pathway, including mutations in the pro-opiomelanocortin (POMC), prohormone convertase 1 (PCSK1), leptin receptor (LEPR), or MC4R genes, have been shown to cause early-onset severe obesity. METHODS Through a comprehensive epidemiological analysis of known and predicted LoF variants in the POMC, PCSK1, and LEPR genes, we sought to estimate the number of US individuals with biallelic MC4R pathway LoF variants. RESULTS We predict ~650 α-melanocyte-stimulating hormone (MSH)/POMC, 8500 PCSK1, and 3600 LEPR homozygous and compound heterozygous individuals in the United States, cumulatively enumerating >12,800 MC4R pathway-deficient obese patients. Few of these variants have been genetically diagnosed to date. These estimates increase when we include a small subset of less rare variants: β-MSH/POMC,PCSK1 N221D, and a PCSK1 LoF variant (T640A). To further define the MC4R pathway and its potential impact on obesity, we tested associations between body mass index (BMI) and LoF mutation burden in the POMC, PCSK1, and LEPR genes in various populations. We show that the cumulative allele burden in individuals with two or more LoF alleles in one or more genes in the MC4R pathway are predisposed to a higher BMI than noncarriers or heterozygous LoF carriers with a defect in only one gene. CONCLUSIONS Our analysis represents a genetically rationalized study of the hypothalamic MC4R pathway aimed at genetic patient stratification to determine which obese subpopulations should be studied to elucidate MC4R agonist (e.g., setmelanotide) treatment responsiveness.
Collapse
Affiliation(s)
- Kristin L Ayers
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Sema4, Stamford, Connecticut
| | - Benjamin S Glicksberg
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | | | - Lei Du
- WuXiNextCode, Cambridge, Massachusetts
| | | | | | - Sophie Roy
- Rhythm Pharmaceuticals, Boston, Massachusetts
| | | | | | | | - Kari E North
- University of North Carolina, Chapel Hill, North Carolina
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Sema4, Stamford, Connecticut
| | - Shuyu D Li
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Sema4, Stamford, Connecticut
- Correspondence and Reprint Requests: Shuyu D. Li, PhD, or Rong Chen, PhD, Icahn School of Medicine at Mount Sinai, 1255 5th Avenue, New York, New York 10029. E-mail: or; or Lex H. T. Van der Ploeg, PhD, Rhythm Pharmaceuticals, 500 Boylston Street, Boston, Massachusetts 02116. E-mail:
| | - Rong Chen
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Sema4, Stamford, Connecticut
- Correspondence and Reprint Requests: Shuyu D. Li, PhD, or Rong Chen, PhD, Icahn School of Medicine at Mount Sinai, 1255 5th Avenue, New York, New York 10029. E-mail: or; or Lex H. T. Van der Ploeg, PhD, Rhythm Pharmaceuticals, 500 Boylston Street, Boston, Massachusetts 02116. E-mail:
| | - Lex H T Van der Ploeg
- Rhythm Pharmaceuticals, Boston, Massachusetts
- Correspondence and Reprint Requests: Shuyu D. Li, PhD, or Rong Chen, PhD, Icahn School of Medicine at Mount Sinai, 1255 5th Avenue, New York, New York 10029. E-mail: or; or Lex H. T. Van der Ploeg, PhD, Rhythm Pharmaceuticals, 500 Boylston Street, Boston, Massachusetts 02116. E-mail:
| |
Collapse
|
45
|
Dong SS, Zhang YJ, Chen YX, Yao S, Hao RH, Rong Y, Niu HM, Chen JB, Guo Y, Yang TL. Comprehensive review and annotation of susceptibility SNPs associated with obesity-related traits. Obes Rev 2018. [PMID: 29527783 DOI: 10.1111/obr.12677] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We aimed to summarize the results of genetic association studies for obesity and provide a comprehensive annotation of all susceptibility single nucleotide polymorphisms (SNPs). A total of 72 studies were summarized, resulting in 90,361 susceptibility SNPs (738 index SNPs and 89,623 linkage disequilibrium SNPs). Over 90% of the susceptibility SNPs are located in non-coding regions, and it is challenging to understand their functional significance. Therefore, we annotated these SNPs by using various functional databases. We identified 24,623 functional SNPs, including 4 nonsense SNPs, 479 missense SNPs, 399 untranslated region SNPs which might affect microRNA binding, 262 promoter and 5,492 enhancer SNPs which might affect transcription factor binding, 7 splicing sites, 76 SNPs which might affect gene methylation levels, 1,839 SNPs under natural selection and 17,351 SNPs which might modify histone binding. Expression quantitative trait loci analyses for functional SNPs identified 98 target genes, including 69 protein coding genes, 27 long non-coding RNAs and 3 processed transcripts. The percentage of protein coding genes that could be correlated with obesity-related pathways directly or through gene-gene interaction is 75.36 (52/69). Our results may serve as an encyclopaedia of obesity susceptibility SNPs and offer guide for functional experiments.
Collapse
Affiliation(s)
- S-S Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y-J Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y-X Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - S Yao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - R-H Hao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y Rong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - H-M Niu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - J-B Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - T-L Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
46
|
Zhang K, Cheng BH, Yang LL, Wang ZP, Zhang HL, Xu SS, Wang SZ, Wang YX, Zhang H, Li H. Identification of a potential functional single nucleotide polymorphism for fatness and growth traits in the 3'-untranslated region of the PCSK1 gene in chickens. J Anim Sci 2018; 95:4776-4786. [PMID: 29293721 DOI: 10.2527/jas2017.1706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Prohormone convertase 1/3 is a serine endoprotease belonging to the subtilisin-like proprotein convertase family that is encoded by the () gene, and its major function is the processing and bioactivation of the proproteins of many kinds of neuroendocrine hormones, including insulin, cholecystokinin, and adrenocorticotropic hormone. The results of our previous genomewide association study indicated that the gene might be an important candidate gene for fatness traits in chickens. The objectives of this study were to investigate the tissue expression profiles of gene and to identify functional variants associated with fatness and growth traits in the chicken. The results indicated that mRNA was widely expressed in various tissues, especially neuroendocrine and intestinal tissues. Of these 2 tissue types, mRNA expression in lean males was significantly higher than in fat males. A SNP in the 3' untranslated region of (c.*900G > A) was identified. Association analysis in the Arbor Acres commercial broiler population and Northeast Agricultural University broiler lines divergently selected for abdominal fat content (NEAUHLF) population showed that the SNP c.*900G > A was associated with abdominal fat weight, abdominal fat percentage, BW, metatarsus length, and metatarsal circumference. In the 5th to 19th generation (G to G) of NEAUHLF, the allele frequency of c.*900G > A changed along with selection for abdominal fat content. At G, allele G of c.*900G > A was predominate in the lean line, whereas allele A was predominate in the fat line. Functional analysis demonstrated that allele A of c.*900G > A reduced mRNA stability and consequently downregulated gene expression. These results suggested that c.*900G > A was a functional SNP for fatness and growth traits in the chicken. The results of this study provide basic molecular information for the role of gene in avian growth and development, especially obesity.
Collapse
|
47
|
Stryjecki C, Alyass A, Meyre D. Ethnic and population differences in the genetic predisposition to human obesity. Obes Rev 2018; 19:62-80. [PMID: 29024387 DOI: 10.1111/obr.12604] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/17/2017] [Accepted: 08/02/2017] [Indexed: 12/22/2022]
Abstract
Obesity rates have escalated to the point of a global pandemic with varying prevalence across ethnic groups. These differences are partially explained by lifestyle factors in addition to genetic predisposition to obesity. This review provides a comprehensive examination of the ethnic differences in the genetic architecture of obesity. Using examples from evolution, heritability, admixture, monogenic and polygenic studies of obesity, we provide explanations for ethnic differences in the prevalence of obesity. The debate over definitions of race and ethnicity, the advantages and limitations of multi-ethnic studies and future directions of research are also discussed. Multi-ethnic studies have great potential to provide a better understanding of ethnic differences in the prevalence of obesity that may result in more targeted and personalized obesity treatments.
Collapse
Affiliation(s)
- C Stryjecki
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - A Alyass
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - D Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
48
|
Skaaby T, Taylor AE, Thuesen BH, Jacobsen RK, Friedrich N, Møllehave LT, Hansen S, Larsen SC, Völker U, Nauck M, Völzke H, Hansen T, Pedersen O, Jørgensen T, Paternoster L, Munafò M, Grarup N, Linneberg A. Estimating the causal effect of body mass index on hay fever, asthma and lung function using Mendelian randomization. Allergy 2018; 73:153-164. [PMID: 28675761 DOI: 10.1111/all.13242] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND Observational studies have shown that body mass index (BMI) is positively associated with asthma. However, observational data are prone to confounding and reverse causation. In Mendelian randomization, genetic variants are used as unconfounded markers of exposures to examine causal effects. We examined the causal effect of BMI on asthma, hay fever, allergic sensitization, serum total immunoglobulin E (IgE), forced expiratory volume in one-second (FEV1) and forced vital capacity (FVC). METHODS We included 490 497 participants in the observational and 162 124 participants in the genetic analyses. A genetic risk score (GRS) was created using 26 BMI-associated single nucleotide polymorphisms (SNPs). Results were pooled in meta-analyses and expressed as odds ratios (ORs) or β-estimates with 95% confidence interval (CI). RESULTS The GRS was significantly associated with asthma (OR=1.009; 95% CI: 1.004, 1.013), but not with hay fever (OR= 0.998; 95% CI: 0.994, 1.002) or allergic sensitization (OR=0.999; 95% CI: 0.986, 1.012) per BMI-increasing allele. The GRS was significantly associated with decrease in FEV1: β=-0.0012 (95% CI: -0.0019, -0.0006) and FVC: β=-0.0022 (95% CI: -0.0031, -0.0014) per BMI-increasing allele. Effect sizes estimated by instrumental variable analyses were OR=1.07 (95% CI: 1.03, 1.10) for asthma, a 9 ml decrease in FEV1 (95% CI: 2.0-15 mL decrease) and a 16 ml decrease in FVC (95% CI: 7.0-24 mL decrease) per 1 kg/m2 higher BMI. CONCLUSIONS The results support the conclusion that increasing BMI is causally related to higher prevalence of asthma and decreased lung function, but not with hay fever or biomarkers of allergy.
Collapse
Affiliation(s)
- T. Skaaby
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
| | - A. E. Taylor
- MRC Integrative Epidemiology Unit (IEU) The University of Bristol Bristol UK
- UK Centre for Tobacco and Alcohol Studies School of Experimental Psychology University of Bristol Bristol UK
| | - B. H. Thuesen
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
| | - R. K. Jacobsen
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
| | - N. Friedrich
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
- Institute of Clinical Chemistry and Laboratory Medicine University Medicine Greifswald Greifswald Germany
| | - L. T. Møllehave
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
| | - S. Hansen
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
| | - S. C. Larsen
- Research unit for Dietary Studies The Parker Institute Frederiksberg and Bispebjerg Hospitals The Capital Region Frederiksberg Denmark
| | - U. Völker
- Interfaculty Institute for Genetics and Functional Genomics University Medicine and Ernst‐Moritz‐Arndt University Greifswald Greifswald Germany
| | - M. Nauck
- Institute of Clinical Chemistry and Laboratory Medicine University Medicine Greifswald Greifswald Germany
| | - H. Völzke
- Institute for Community Medicine University Medicine Greifswald Greifswald Germany
| | - T. Hansen
- Section on Metabolic Genetics Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - O. Pedersen
- Section on Metabolic Genetics Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - T. Jørgensen
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
- Department of Public Health Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Faculty of Medicine Aalborg University Aalborg Denmark
| | - L. Paternoster
- MRC Integrative Epidemiology Unit (IEU) The University of Bristol Bristol UK
| | - M. Munafò
- MRC Integrative Epidemiology Unit (IEU) The University of Bristol Bristol UK
- UK Centre for Tobacco and Alcohol Studies School of Experimental Psychology University of Bristol Bristol UK
| | - N. Grarup
- Section on Metabolic Genetics Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - A. Linneberg
- Research Centre for Prevention and Health Centre for Health Capital Region of Denmark Copenhagen Denmark
- Department of Clinical Experimental Research Rigshospitalet Glostrup Denmark
- Department of Clinical Medicine Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
49
|
Abadi A, Alyass A, Robiou du Pont S, Bolker B, Singh P, Mohan V, Diaz R, Engert JC, Yusuf S, Gerstein HC, Anand SS, Meyre D. Penetrance of Polygenic Obesity Susceptibility Loci across the Body Mass Index Distribution. Am J Hum Genet 2017; 101:925-938. [PMID: 29220676 PMCID: PMC5812888 DOI: 10.1016/j.ajhg.2017.10.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022] Open
Abstract
A growing number of single-nucleotide polymorphisms (SNPs) have been associated with body mass index (BMI) and obesity, but whether the effects of these obesity-susceptibility loci are uniform across the BMI distribution remains unclear. We studied the effects of 37 BMI-associated SNPs in 75,230 adults of European ancestry across BMI percentiles by using conditional quantile regression (CQR) and meta-regression (MR) models. The effects of nine SNPs (24%)-rs1421085 (FTO; p = 8.69 × 10-15), rs6235 (PCSK1; p = 7.11 × 10-6), rs7903146 (TCF7L2; p = 9.60 × 10-6), rs11873305 (MC4R; p = 5.08 × 10-5), rs12617233 (FANCL; p = 5.30 × 10-5), rs11672660 (GIPR; p = 1.64 × 10-4), rs997295 (MAP2K5; p = 3.25 × 10-4), rs6499653 (FTO; p = 6.23 × 10-4), and rs3824755 (NT5C2; p = 7.90 × 10-4)-increased significantly across the sample BMI distribution. We showed that such increases stemmed from unadjusted gene interactions that enhanced the effects of SNPs in persons with a high BMI. When 125 height-associated SNPs were analyzed for comparison, only one (<1%), rs6219 (IGF1, p = 1.80 × 10-4), showed effects that varied significantly across height percentiles. Cumulative gene scores of these SNPs (GS-BMI and GS-height) showed that only GS-BMI had effects that increased significantly across the sample distribution (BMI: p = 7.03 × 10-37; height: p = 0.499). Overall, these findings underscore the importance of gene-gene and gene-environment interactions in shaping the genetic architecture of BMI and advance a method for detecting such interactions by using only the sample outcome distribution.
Collapse
Affiliation(s)
- Arkan Abadi
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Akram Alyass
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sebastien Robiou du Pont
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ben Bolker
- Department of Mathematics and Statistics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Pardeep Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation, Gopalapuram, Chennai 600086, India
| | - Rafael Diaz
- Estudios Clínicos Latino America, Paraguay 160, S2000CVD Rosario, Santa Fe, Argentina
| | | | - Salim Yusuf
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Hertzel C Gerstein
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sonia S Anand
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton General Hospital, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
50
|
Abstract
Zusammenfassung
Autosomal-rezessiv vererbte Mutationen in den Genen für Leptin, Leptinrezeptor, Proopiomelanocortin (POMC) und Prohormon-Convertase (PC1) führen zu einer ausgeprägten frühkindlichen Adipositas. Patienten mit biologisch inaktivem Leptin oder Leptinmangel können mit humanem rekombinanten Leptin erfolgreich behandelt werden. Für die anderen Patienten hat sich die Behandlung mit einem α‑MSH-Analogon als erfolgreich erwiesen (POMC-Patienten) bzw. befindet sich derzeit in Erprobung.
Kodominant vererbte Mutationen im MC4R-Gen stellen die häufigste Form der monogenen Adipositas dar. Eine kausale Therapie ist hier allerdings nicht möglich.
Es sind inzwischen noch weitere, autosomal-rezessiv vererbte Genmutationen identifiziert worden, die ebenfalls mit einer ausgeprägten Adipositas assoziiert sind. Die meisten dieser Mutationen liegen in Genen, die in die Signaltransduktion von MC4R oder dem Leptinrezeptor involviert sind. Auch für diese Patienten gibt es aktuell noch keine kausale Therapie.
Schlussfolgerung: Bei Patienten mit extremer frühkindlicher Adipositas sollte eine molekulargenetische Diagnostik eingeleitet werden, da die Diagnosestellung für die Betroffenen und ihre Familie eine enorme Erleichterung bedeuten kann. Außerdem gewinnen die Familien Klarheit über das Wiederholungsrisiko und eventuell ist sogar eine kausale oder zumindest optimierte Therapie möglich.
Collapse
Affiliation(s)
- Julia von Schnurbein
- Aff1 grid.410712.1 Klinik für Kinder- und Jugendmedizin, Zentrum für Seltene Erkrankungen (ZSE) Ulm, Sektion Pädiatrische Endokrinologie und Diabetologie Universitätsklinik für Kinder- und Jugendmedizin Eythstr. 24 89075 Ulm Deutschland
| | - Martin Wabitsch
- Aff1 grid.410712.1 Klinik für Kinder- und Jugendmedizin, Zentrum für Seltene Erkrankungen (ZSE) Ulm, Sektion Pädiatrische Endokrinologie und Diabetologie Universitätsklinik für Kinder- und Jugendmedizin Eythstr. 24 89075 Ulm Deutschland
| |
Collapse
|