1
|
Azzam SK, Alsafar H, Sajini AA. FTO m6A Demethylase in Obesity and Cancer: Implications and Underlying Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms23073800. [PMID: 35409166 PMCID: PMC8998816 DOI: 10.3390/ijms23073800] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/20/2022] Open
Abstract
Fat mass and obesity-associated protein (FTO) is the first reported RNA N6-methyladenosine (m6A) demethylase in eukaryotic cells. m6A is considered as the most abundant mRNA internal modification, which modulates several cellular processes including alternative splicing, stability, and expression. Genome-wide association studies (GWAS) identified single-nucleotide polymorphisms (SNPs) within FTO to be associated with obesity, as well as cancer including endometrial cancer, breast cancer, pancreatic cancer, and melanoma. Since the initial classification of FTO as an m6A demethylase, various studies started to unravel a connection between FTO’s demethylase activity and the susceptibility to obesity on the molecular level. FTO was found to facilitate adipogenesis, by regulating adipogenic pathways and inducing pre-adipocyte differentiation. FTO has also been investigated in tumorigenesis, where emerging studies suggest m6A and FTO levels are dysregulated in various cancers, including acute myeloid leukemia (AML), glioblastoma, cervical squamous cell carcinoma (CSCC), breast cancer, and melanoma. Here we review the molecular bases of m6A in tumorigenesis and adipogenesis while highlighting the controversial role of FTO in obesity. We provide recent findings confirming FTO’s causative link to obesity and discuss novel approaches using RNA demethylase inhibitors as targeted oncotherapies. Our review aims to confirm m6A demethylation as a risk factor in obesity and provoke new research in FTO and human disorders.
Collapse
Affiliation(s)
- Sarah Kassem Azzam
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.K.A.); (H.A.)
- Healthcare Engineering Innovation Center (HEIC), Department of Biomedical Engineering, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Habiba Alsafar
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.K.A.); (H.A.)
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Emirates Bio-Research Center, Ministry of Interior, Abu Dhabi P.O. Box 389, United Arab Emirates
| | - Abdulrahim A. Sajini
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.K.A.); (H.A.)
- Healthcare Engineering Innovation Center (HEIC), Department of Biomedical Engineering, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Correspondence:
| |
Collapse
|
2
|
Structural characteristics of small-molecule inhibitors targeting FTO demethylase. Future Med Chem 2021; 13:1475-1489. [PMID: 34240624 DOI: 10.4155/fmc-2021-0132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Studies have shown that the FTO gene is closely related to obesity and weight gain in humans. FTO is an N6-methyladenosine demethylase and is linked to an increased risk of obesity and a variety of diseases, such as acute myeloid leukemia, type 2 diabetes, breast cancer, glioblastoma and cervical squamous cell carcinoma. In light of the significant role of FTO, the development of small-molecule inhibitors targeting the FTO protein provides not only a powerful tool for grasping the active site of FTO but also a theoretical basis for the design and synthesis of drugs targeting the FTO protein. This review focuses on the structural characteristics of FTO inhibitors and discusses the occurrence of obesity and cancer caused by FTO gene overexpression.
Collapse
|
3
|
SAINI SIMMI, WALIA GAGANDEEPKAUR, SACHDEVA MOHINDERPAL, GUPTA VIPIN. Genomics of body fat distribution. J Genet 2021. [DOI: 10.1007/s12041-021-01281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
4
|
Sun C, Kovacs P, Guiu-Jurado E. Genetics of Obesity in East Asians. Front Genet 2020; 11:575049. [PMID: 33193685 PMCID: PMC7606890 DOI: 10.3389/fgene.2020.575049] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022] Open
Abstract
Obesity has become a public health problem worldwide. Compared with Europe, people in Asia tend to suffer from type 2 diabetes with a lower body mass index (BMI). Genome-wide association studies (GWASs) have identified over 750 loci associated with obesity. Although the majority of GWAS results were conducted in individuals of European ancestry, a recent GWAS in individuals of Asian ancestry has made a significant contribution to the identification of obesity susceptibility loci. Indeed, owing to the multifactorial character of obesity with a strong environmental component, the revealed loci may have distinct contributions in different ancestral genetic backgrounds and in different environments as presented through diet and exercise among other factors. Uncovering novel, yet unrevealed genes in non-European ancestries may further contribute to explaining the missing heritability for BMI. In this review, we aimed to summarize recent advances in obesity genetics in individuals of Asian ancestry. We therefore compared proposed mechanisms underlying susceptibility loci for obesity associated with individuals of European and Asian ancestries and discussed whether known genetic variants might explain ethnic differences in obesity risk. We further acknowledged that GWAS implemented in individuals of Asian ancestries have not only validated the potential role of previously specified obesity susceptibility loci but also exposed novel ones, which have been missed in the initial genetic studies in individuals of European ancestries. Thus, multi-ethnic studies have a great potential not only to contribute to a better understanding of the complex etiology of human obesity but also potentially of ethnic differences in the prevalence of obesity, which may ultimately pave new avenues in more targeted and personalized obesity treatments.
Collapse
Affiliation(s)
| | - Peter Kovacs
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | | |
Collapse
|
5
|
Javanrouh N, Soltanian AR, Tapak L, Azizi F, Ott J, Daneshpour MS. A novel association of rs13334070 in the RPGRIP1L gene with adiposity factors discovered by joint linkage and linkage disequilibrium analysis in Iranian pedigrees: Tehran Cardiometabolic Genetic Study (TCGS). Genet Epidemiol 2018; 43:342-351. [PMID: 30597647 DOI: 10.1002/gepi.22179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/15/2018] [Accepted: 11/26/2018] [Indexed: 02/01/2023]
Abstract
Understanding the genetic and metabolic bases of obesity is helpful in planning and developing health strategies. Therefore, the first family-based joint linkage and linkage disequilibrium study was conducted in Iranian pedigrees to assess the relationship between obesity and single-nucleotide polymorphisms (SNPs) located in the 16q12.2 region. In the present study, a total of 13,344 individuals were included, of whom 12,502 individuals were within 3,109 pedigrees and 842 were unrelated singletons. To investigate the relationship between obesity and genetic variants, a joint model of linkage and linkage disequilibrium was applied. Moreover, a sequence kernel association test (SKAT) was used to evaluate the association of the SNP set with body size and lipid profile measurements. The joint model showed that rs13334070, in the intron 4 of the RPGRIP1L gene, has a significant association with obesity. According to the 4-gamete rule, which is a procedure for constructing SNP sets by considering recombination occurrence between SNPs, this polymorphism has a high correlation with six nearby SNPs that make an SNP set. SKAT showed that this SNP set has a significant association with body size factors, but almost no association with most of the lipid profile measurements. In conclusion, from the result of this study, it might be reasonable to consider RPGRIP1L as an important gene whose variations could be associated with obesity risk factors.
Collapse
Affiliation(s)
- Niloufar Javanrouh
- Department of Biostatistics and Epidemiology, Modeling of Non-Communicable Diseases Research Center, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Cellular and Molecular, Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali R Soltanian
- Department of Biostatistics and Epidemiology, Modeling of Non-Communicable Diseases Research Center, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leili Tapak
- Department of Biostatistics and Epidemiology, Modeling of Non-Communicable Diseases Research Center, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fereidoun Azizi
- Department of Thyroid, Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jurg Ott
- Department of Statistical Genomics Methodology, Laboratory of Statistical Genetics, Rockefeller University, New York, New York
| | - Maryam S Daneshpour
- Department of Cellular and Molecular, Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Chang JY, Park JH, Park SE, Shon J, Park YJ. The Fat Mass- and Obesity-Associated (FTO) Gene to Obesity: Lessons from Mouse Models. Obesity (Silver Spring) 2018; 26:1674-1686. [PMID: 30311736 DOI: 10.1002/oby.22301] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/10/2018] [Accepted: 08/10/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Genetic variants at the fat mass- and obesity-associated (FTO) locus are strongly associated with obesity-related traits by regulating neighboring genes. Nevertheless, it is possible that FTO protein is directly involved in mechanisms regulating body composition and adiposity. Here, the in vivo biological functions of FTO in the risk for obesity were studied by reviewing murine models. METHODS The effects of the locus-specific manipulations of the murine Fto gene on metabolic-related phenotypes in genetically modified mouse models were reviewed and summarized into the following three categories: growth and body composition, eating behaviors, and metabolic homeostasis. RESULTS The mouse models showed different phenotypes depending on target tissues and methods for gene manipulation. Mice harboring deletions or point mutations at the Fto locus had high metabolic rates, while FTO-overexpressing mice showed dyslipidemia. Both deletion and overexpression of the Fto gene led to abnormal eating behaviors. Intriguingly, several phenotypes were differently expressed depending on developmental timing of the genetic manipulations. For instance, a germ line deletion decreased total body fat mass, while the deletion in adult mice increased it. CONCLUSIONS The results highlight that FTO is critical not only for body composition but also normal development, and its function might differ depending on the stage of development.
Collapse
Affiliation(s)
- Jeong Yoon Chang
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Joo Hyun Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Sung Eun Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Jinyoung Shon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Yoon Jung Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Deng X, Su R, Stanford S, Chen J. Critical Enzymatic Functions of FTO in Obesity and Cancer. Front Endocrinol (Lausanne) 2018; 9:396. [PMID: 30105001 PMCID: PMC6077364 DOI: 10.3389/fendo.2018.00396] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/27/2018] [Indexed: 01/14/2023] Open
Abstract
Fat mass and obesity-associated protein (FTO) single-nucleotide polymorphisms (SNPs) have been linked to increased body mass and obesity in humans by genome-wide association studies (GWAS) since 2007. Although some recent studies suggest that the obesity-related SNPs in FTO influence obesity susceptibility likely through altering the expression of the adjacent genes such as IRX3 and RPGRIP1L, rather than FTO itself, a solid link between the SNP risk genotype and the increased FTO expression in both human blood cells and fibroblasts has been reported. Moreover, multiple lines of evidence have demonstrated that FTO does play a critical role in the regulation of fat mass, adipogenesis, and body weight. Epidemiology studies also showed a strong association of FTO SNPs and overweight/obesity with increased risk of various types of cancers. As the first identified messenger RNA N6-methyladenosine (m6A) demethylase, FTO has been shown recently to play m6A-dependent roles in adipogenesis and tumorigenesis (especially in the development of leukemia and glioblastoma). Given the critical roles of FTO in cancers, the development of selective and effective inhibitors targeting FTO holds potential to treat cancers. This mini review discusses the roles and underlying molecular mechanisms of FTO in both obesity and cancers, and also summarizes recent advances in the development of FTO inhibitors.
Collapse
Affiliation(s)
- Xiaolan Deng
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, Beckman Research Institute of City of HopeMonrovia, CA, United States
- School of PharmacyChina Medical University, Shenyang, China
| | - Rui Su
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, Beckman Research Institute of City of HopeMonrovia, CA, United States
| | - Savanna Stanford
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, Beckman Research Institute of City of HopeMonrovia, CA, United States
| | - Jianjun Chen
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, Beckman Research Institute of City of HopeMonrovia, CA, United States
- *Correspondence: Jianjun Chen
| |
Collapse
|
8
|
Park C, Kim JI, Hong SN, Jung HM, Kim TJ, Lee S, Kim SJ, Kim HC, Kim DH, Cho B, Park JH, Sung J, Lee DS, Kang M, Son HJ, Kim YH. A copy number variation in PKD1L2 is associated with colorectal cancer predisposition in korean population. Int J Cancer 2016; 140:86-94. [PMID: 27605020 DOI: 10.1002/ijc.30421] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 08/15/2016] [Indexed: 12/30/2022]
Abstract
Recently reported genome-wide association studies have identified more than 20 common low-penetrance colorectal cancer (CRC) susceptibility loci. Recent studies have reported that copy number variations (CNVs) are considered important human genomic variants related to cancer, while the contribution of CNVs remains unclear. We performed array comparative genomic hybridization (aCGH) in 36 CRC patients and 47 controls. Using breakpoint PCR, we confirmed the breakpoint of the PKD1L2 deletion region. High frequency of PKD1L2 CNV was observed in CRC cases. We validated the association between PKD1L2 variation and CRC risk in 1,874 cases and 2,088 controls (OR = 1.44, 95% CI = 1.04-1.98, p = 0.028). Additionally, PKD1L2 CNV is associated with increased CRC risk in patients younger than 50 years (OR = 2.14, 95% CI 1.39-3.30, p = 5.8 × 10-4 ). In subgroup analysis according to body mass index (BMI), we found that the CN loss of PKD1L2 with BMI above or equal to 25 exhibited a significant increase in CRC risk (OR = 2.29, 95% CI 1.29-4.05, p = 0.005). PKD1L2 CNV with BMI above or equal to 25 and age below 50 is associated with a remarkably increased risk of colorectal cancer (OR = 5.24, 95% CI 2.36-11.64, p= 4.8 × 10-5 ). Moreover, we found that PKD1L2 variation in obese patients (BMI ≥ 25) was associated with poor survival rate (p = 0.026). Our results suggest that the common PKD1L2 CNV is associated with CRC, and PKD1L2 CNV with high BMI and/or age below 50 exhibited a significant increased risk of CRC. In obese patients, PKD1L2 variation was associated with poor survival.
Collapse
Affiliation(s)
- Changho Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hey Mi Jung
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Jun Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seungbok Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Seong Jin Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk-Hwan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Belong Cho
- Department of Family Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jin-Ho Park
- Department of Family Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Joohon Sung
- Complex Disease and Genome Epidemiology Branch, Department of Epidemiology, School of Public Health, Seoul National University, Seoul, Korea
| | - Dong-Sung Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Mingon Kang
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Jung Son
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Zhang YP, Zhang YY, Duan DD. From Genome-Wide Association Study to Phenome-Wide Association Study: New Paradigms in Obesity Research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:185-231. [PMID: 27288830 DOI: 10.1016/bs.pmbts.2016.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity is a condition in which excess body fat has accumulated over an extent that increases the risk of many chronic diseases. The current clinical classification of obesity is based on measurement of body mass index (BMI), waist-hip ratio, and body fat percentage. However, these measurements do not account for the wide individual variations in fat distribution, degree of fatness or health risks, and genetic variants identified in the genome-wide association studies (GWAS). In this review, we will address this important issue with the introduction of phenome, phenomics, and phenome-wide association study (PheWAS). We will discuss the new paradigm shift from GWAS to PheWAS in obesity research. In the era of precision medicine, phenomics and PheWAS provide the required approaches to better definition and classification of obesity according to the association of obese phenome with their unique molecular makeup, lifestyle, and environmental impact.
Collapse
Affiliation(s)
- Y-P Zhang
- Pediatric Heart Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Y-Y Zhang
- Department of Cardiology, Changzhou Second People's Hospital, Changzhou, Jiangsu, China
| | - D D Duan
- Laboratory of Cardiovascular Phenomics, Center for Cardiovascular Research, Department of Pharmacology, and Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV, United States.
| |
Collapse
|
10
|
Aissani B, Wiener HW, Zhang K. Fine Mapping of the Body Fat QTL on Human Chromosome 1q43. PLoS One 2016; 11:e0153794. [PMID: 27111224 PMCID: PMC4844098 DOI: 10.1371/journal.pone.0153794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/04/2016] [Indexed: 11/20/2022] Open
Abstract
Introduction Evidence for linkage and association of obesity-related quantitative traits to chromosome 1q43 has been reported in the Quebec Family Study (QFS) and in populations of Caribbean Hispanic ancestries yet no specific candidate locus has been replicated to date. Methods Using a set of 1,902 single nucleotide polymorphisms (SNPs) genotyped in 525 African American (AA) and 391 European American (EA) women enrolled in the NIEHS uterine fibroid study (NIEHS-UFS), we generated a fine association map for the body mass index (BMI) across a 2.3 megabase-long interval delimited by RGS7 (regulator of G-protein signaling 7) and PLD5 (Phospholipase D, member 5). Multivariable-adjusted linear regression models were fitted to the data to evaluate the association in race-stratified analyses and meta-analysis. Results The strongest associations were observed in a recessive genetic model and peaked in the 3’ end of RGS7 at intronic rs261802 variant in the AA group (p = 1.0 x 10−4) and in meta-analysis of AA and EA samples (p = 9.0 x 10−5). In the EA group, moderate associations peaked at rs6429264 (p = 2.0 x 10−3) in the 2 Kb upstream sequence of RGS7. In the reference populations for the European ancestry in the 1,000 genomes project, rs6429264 occurs in strong linkage disequilibrium (D’ = 0.94) with rs1341467, the strongest candidate SNP for total body fat in QFS that failed genotyping in the present study. Additionally we report moderate associations at the 3’ end of PLD5 in meta-analysis (3.2 x 10−4 ≤ p ≤ 5.8 x 10−4). Conclusion We report replication data suggesting that RGS7, a gene abundantly expressed in the brain, might be a putative body fat QTL on human chromosome 1q43. Future genetic and functional studies are required to substantiate our observations and to potentially link them to the neurobehavioral phenotypes associated with the RGS7 region.
Collapse
Affiliation(s)
- Brahim Aissani
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama, United States of America
- * E-mail:
| | - Howard W. Wiener
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama, United States of America
| | - Kui Zhang
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| |
Collapse
|
11
|
Young KL, Graff M, North KE, Richardson AS, Mohlke KL, Lange LA, Lange EM, Harris KM, Gordon-Larsen P. Interaction of smoking and obesity susceptibility loci on adolescent BMI: The National Longitudinal Study of Adolescent to Adult Health. BMC Genet 2015; 16:131. [PMID: 26537541 PMCID: PMC4634717 DOI: 10.1186/s12863-015-0289-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/29/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Adolescence is a sensitive period for weight gain and risky health behaviors, such as smoking. Genome-wide association studies (GWAS) have identified loci contributing to adult body mass index (BMI). Evidence suggests that many of these loci have a larger influence on adolescent BMI. However, few studies have examined interactions between smoking and obesity susceptibility loci on BMI. This study investigates the interaction of current smoking and established BMI SNPs on adolescent BMI. Using data from the National Longitudinal Study of Adolescent to Adult Health, a nationally-representative, prospective cohort of the US school-based population in grades 7 to 12 (12-20 years of age) in 1994-95 who have been followed into adulthood (Wave II 1996; ages 12-21, Wave III; ages 18-27), we assessed (in 2014) interactions of 40 BMI-related SNPs and smoking status with percent of the CDC/NCHS 2000 median BMI (%MBMI) in European Americans (n = 5075), African Americans (n = 1744) and Hispanic Americans (n = 1294). RESULTS Two SNPs showed nominal significance for interaction (p < 0.05) between smoking and genotype with %MBMI in European Americans (EA) (rs2112347 (POC5): β = 1.98 (0.06, 3.90), p = 0.04 and near rs571312 (MC4R): β 2.15 (-0.03, 4.33) p = 0.05); and one SNP showed a significant interaction effect after stringent correction for multiple testing in Hispanic Americans (HA) (rs1514175 (TNNI3K): β 8.46 (4.32, 12.60), p = 5.9E-05). Stratifying by sex, these interactions suggest a stronger effect in female smokers. CONCLUSIONS Our study highlights potentially important sex differences in obesity risk by smoking status in adolescents, with those who may be most likely to initiate smoking (i.e., adolescent females), being at greatest risk for exacerbating genetic obesity susceptibility.
Collapse
Affiliation(s)
- Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- , 137 East Franklin Street, Suite 306, Chapel Hill, NC, 27514, USA.
| | - Misa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Andrea S Richardson
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Karen L Mohlke
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Leslie A Lange
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Ethan M Lange
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Kathleen M Harris
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Sociology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Penny Gordon-Larsen
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Merkestein M, Sellayah D. Role of FTO in Adipocyte Development and Function: Recent Insights. Int J Endocrinol 2015; 2015:521381. [PMID: 26788058 PMCID: PMC4695642 DOI: 10.1155/2015/521381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/24/2015] [Accepted: 11/25/2015] [Indexed: 01/11/2023] Open
Abstract
In 2007, FTO was identified as the first genome-wide association study (GWAS) gene associated with obesity in humans. Since then, various animal models have served to establish the mechanistic basis behind this association. Many earlier studies focussed on FTO's effects on food intake via central mechanisms. Emerging evidence, however, implicates adipose tissue development and function in the causal relationship between perturbations in FTO expression and obesity. The purpose of this mini review is to shed light on these new studies of FTO function in adipose tissue and present a clearer picture of its impact on obesity susceptibility.
Collapse
Affiliation(s)
- Myrte Merkestein
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, Oxfordshire OX1 3PT, UK
| | - Dyan Sellayah
- School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6AS, UK
- *Dyan Sellayah:
| |
Collapse
|
13
|
Scott O, Pugh J, Kiddoo D, Sonnenberg LK, Bamforth S, Goez HR. Global developmental delay, progressive relapsing-remitting parkinsonism, and spinal syrinx in a child with SOX6 mutation. J Child Neurol 2014; 29:NP164-7. [PMID: 24453155 DOI: 10.1177/0883073813514134] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
SOX6, a member of the SOX gene family, plays a key role in the development of several mammalian tissues and organs, including the central nervous system. Specifically, this gene modulates the differentiation and proliferation of interneurons in the medial ganglionic eminence, as well as oligodendrocytes in the spinal cord. We describe the case of a 4-year-old girl with global developmental delay and a spinal cord syrinx who presented with recurrent episodes of parkinsonian symptoms subsequent to febrile illnesses. The symptoms included gait instability, tremor, and dysarthria, with a progressive relapsing-remitting course over the span of 2 years. The patient was later found to have a large deletion-type mutation in the SOX6 gene. This case is the first report in humans implying a role for SOX6 in basal ganglia function, as well as spinal cord development.
Collapse
Affiliation(s)
- Ori Scott
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jeffrey Pugh
- Division of Neurosurgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Darcie Kiddoo
- Division of Urology and Pediatric Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Lyn K Sonnenberg
- Glenrose Rehabilitation Hospital, Stollery Children's Hospital, Division of Developmental Pediatrics, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Steven Bamforth
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Helly R Goez
- Division of Pediatric Neurology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
14
|
Zhang W, He C. WITHDRAWN: Regulation of plasma membrane receptors by a new autophagy-related BECN/Beclin family member. Autophagy 2014; 10:938-41. [PMID: 24569622 DOI: 10.4161/auto.28149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Weiran Zhang
- Department of Cell and Molecular Biology; Feinberg School of Medicine; Northwestern University; Chicago, IL USA
| | - Congcong He
- Department of Cell and Molecular Biology; Feinberg School of Medicine; Northwestern University; Chicago, IL USA
| |
Collapse
|
15
|
Affiliation(s)
- Brahim Aissani
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
16
|
Abstract
Single nucleotide polymorphisms (SNPs) that cluster in the first intron of fat mass and obesity associated (FTO) gene are associated obesity traits in genome-wide association studies. The minor allele increases BMI by 0.39 kg/m(2) (or 1,130 g in body weight) and risk of obesity by 1.20-fold. This association has been confirmed across age groups and populations of diverse ancestry; the largest effect is seen in young adulthood. The effect of FTO SNPs on obesity traits in populations of African and Asian ancestry is similar or somewhat smaller than in European ancestry populations. However, the BMI-increasing allele in FTO is substantially less prevalent in populations with non-European ancestry. FTO SNPs do not influence physical activity levels; yet, in physically active individuals, FTO's effect on obesity susceptibility is attenuated by approximately 30%. Evidence from epidemiological and functional studies suggests that FTO confers an increased risk of obesity by subtly changing food intake and preference. Moreover, emerging data suggest a role for FTO in nutrient sensing, regulation of mRNA translation and general growth. In this Review, we discuss the genetic epidemiology of FTO and discuss how its complex biology might link to the regulation of body weight.
Collapse
Affiliation(s)
- Ruth J F Loos
- The Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1003, New York, NY 10029-6574, USA
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| |
Collapse
|
17
|
Abu-Farha M, Tiss A, Abubaker J, Khadir A, Al-Ghimlas F, Al-Khairi I, Baturcam E, Cherian P, Elkum N, Hammad M, John J, Kavalakatt S, Warsame S, Behbehani K, Dermime S, Dehbi M. Proteomics analysis of human obesity reveals the epigenetic factor HDAC4 as a potential target for obesity. PLoS One 2013; 8:e75342. [PMID: 24086512 PMCID: PMC3782461 DOI: 10.1371/journal.pone.0075342] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/13/2013] [Indexed: 02/07/2023] Open
Abstract
Sedentary lifestyle and excessive energy intake are prominent contributors to obesity; a major risk factors for the development of insulin resistance, type 2 diabetes and cardiovascular diseases. Elucidating the molecular mechanisms underlying these chronic conditions is of relevant importance as it might lead to the identification of novel anti-obesity targets. The purpose of the current study is to investigate differentially expressed proteins between lean and obese subjects through a shot-gun quantitative proteomics approach using peripheral blood mononuclear cells (PBMCs) extracts as well as potential modulation of those proteins by physical exercise. Using this approach, a total of 47 proteins showed at least 1.5 fold change between lean and obese subjects. In obese, the proteomic profiling before and after 3 months of physical exercise showed differential expression of 38 proteins. Thrombospondin 1 (TSP1) was among the proteins that were upregulated in obese subjects and then decreased by physical exercise. Conversely, the histone deacetylase 4 (HDAC4) was downregulated in obese subjects and then induced by physical exercise. The proteomic data was further validated by qRT-PCR, Western blot and immunohistochemistry in both PBMCs and adipose tissue. We also showed that HDAC4 levels correlated positively with maximum oxygen consumption (VO2 Max) but negatively with body mass index, percent body fat, and the inflammatory chemokine RANTES. In functional assays, our data indicated that ectopic expression of HDAC4 significantly impaired TNF-α-dependent activation of NF-κB, establishing thus a link between HDAC4 and regulation of the immune system. Together, the expression pattern of HDAC4 in obese subjects before and after physical exercise, its correlation with various physical, clinical and metabolic parameters along with its inhibitory effect on NF-κB are suggestive of a protective role of HDAC4 against obesity. HDAC4 could therefore represent a potential therapeutic target for the control and management of obesity and presumably insulin resistance.
Collapse
Affiliation(s)
- Mohamed Abu-Farha
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Ali Tiss
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Jehad Abubaker
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Abdelkrim Khadir
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Fahad Al-Ghimlas
- Fitness and Rehabilitation Centre, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Irina Al-Khairi
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Engin Baturcam
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Preethi Cherian
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Naser Elkum
- Department of Biostatistics & Epidemiology, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Maha Hammad
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Jeena John
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Sina Kavalakatt
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Samia Warsame
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Kazem Behbehani
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
- Fitness and Rehabilitation Centre, Dasman Diabetes Institute, Kuwait, Kuwait
- Department of Biostatistics & Epidemiology, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Said Dermime
- Biomedical Research Facility, King Fahad Specialist Hospital Dammam, Dammam, Kingdom of Saudi Arabia
| | - Mohammed Dehbi
- Department of Biomedical Research, Dasman Diabetes Institute, Kuwait, Kuwait
- Genomic Medicine and Systems Biology Research Center, Qatar Biomedical Research Institute, Education City, Doha, Qatar
| |
Collapse
|
18
|
He C, Wei Y, Sun K, Li B, Dong X, Zou Z, Liu Y, Kinch LN, Khan S, Sinha S, Xavier RJ, Grishin NV, Xiao G, Eskelinen EL, Scherer PE, Whistler JL, Levine B. Beclin 2 functions in autophagy, degradation of G protein-coupled receptors, and metabolism. Cell 2013; 154:1085-1099. [PMID: 23954414 PMCID: PMC4231430 DOI: 10.1016/j.cell.2013.07.035] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 05/14/2013] [Accepted: 07/10/2013] [Indexed: 11/26/2022]
Abstract
The molecular mechanism of autophagy and its relationship to other lysosomal degradation pathways remain incompletely understood. Here, we identified a previously uncharacterized mammalian-specific protein, Beclin 2, which, like Beclin 1, functions in autophagy and interacts with class III PI3K complex components and Bcl-2. However, Beclin 2, but not Beclin 1, functions in an additional lysosomal degradation pathway. Beclin 2 is required for ligand-induced endolysosomal degradation of several G protein-coupled receptors (GPCRs) through its interaction with GASP1. Beclin 2 homozygous knockout mice have decreased embryonic viability, and heterozygous knockout mice have defective autophagy, increased levels of brain cannabinoid 1 receptor, elevated food intake, and obesity and insulin resistance. Our findings identify Beclin 2 as a converging regulator of autophagy and GPCR turnover and highlight the functional and mechanistic diversity of Beclin family members in autophagy, endolysosomal trafficking, and metabolism.
Collapse
Affiliation(s)
- Congcong He
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yongjie Wei
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kai Sun
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Binghua Li
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaonan Dong
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhongju Zou
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yang Liu
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lisa N Kinch
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shaheen Khan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sangita Sinha
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND 58102, USA
| | - Ramnik J Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Nick V Grishin
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guanghua Xiao
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eeva-Liisa Eskelinen
- Department of Biosciences, Division of Biochemistry and Biotechnology, University of Helsinki, Helsinki, FI-00014 Finland
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jennifer L Whistler
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, CA 94608, USA
| | - Beth Levine
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
19
|
Lu Y, Loos RJ. Obesity genomics: assessing the transferability of susceptibility loci across diverse populations. Genome Med 2013; 5:55. [PMID: 23806069 PMCID: PMC3706771 DOI: 10.1186/gm459] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The prevalence of obesity has nearly doubled worldwide over the past three decades, but substantial differences exist between nations. Although these differences are partly due to the degree of westernization, genetic factors also contribute. To date, little is known about whether the same genes contribute to obesity-susceptibility in populations of different ancestry. We review the transferability of obesity-susceptibility loci (identified by genome-wide association studies) using both single nucleotide polymorphism (SNP) and locus-wide comparisons. SNPs in FTO and near MC4R, obesity-susceptibility loci first identified in Europeans, replicate widely across other ancestries. SNP-to-SNP comparisons suggest that more than half of the 36 body mass index-associated loci are shared across European and East Asian ancestry populations, whereas locus-wide analyses suggest that the transferability might be even more extensive. Furthermore, by taking advantage of differences in haplotype structure, populations of different ancestries can help to narrow down loci, thereby pinpointing causal genes for functional follow-up. Larger-scale genetic association studies in ancestrally diverse populations will be needed for in-depth and locus-wide analyses aimed at determining, with greater confidence, the transferability of loci and allowing fine-mapping. Understanding similarities and differences in genetic susceptibility across populations of diverse ancestries might eventually contribute to a more targeted prevention and customized treatment of obesity.
Collapse
Affiliation(s)
- Yingchang Lu
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA ; The Charles Bronfman Institute of Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ruth Jf Loos
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA ; The Charles Bronfman Institute of Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA ; The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA ; The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
20
|
Wu C, Gong Y, Yuan J, Gong H, Zou Y, Ge J. Identification of shared genetic susceptibility locus for coronary artery disease, type 2 diabetes and obesity: a meta-analysis of genome-wide studies. Cardiovasc Diabetol 2012; 11:68. [PMID: 22697793 PMCID: PMC3481354 DOI: 10.1186/1475-2840-11-68] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/28/2012] [Indexed: 01/10/2023] Open
Abstract
Type 2 diabetes (2DM), obesity, and coronary artery disease (CAD) are frequently coexisted being as key components of metabolic syndrome. Whether there is shared genetic background underlying these diseases remained unclear. We performed a meta-analysis of 35 genome screens for 2DM, 36 for obesity or body mass index (BMI)-defined obesity, and 21 for CAD using genome search meta-analysis (GSMA), which combines linkage results to identify regions with only weak evidence and provide genetic interactions among different diseases. For each study, 120 genomic bins of approximately 30 cM were defined and ranked according to the best linkage evidence within each bin. For each disease, bin 6.2 achieved genomic significanct evidence, and bin 9.3, 10.5, 16.3 reached suggestive level for 2DM. Bin 11.2 and 16.3, and bin 10.5 and 9.3, reached suggestive evidence for obesity and CAD respectively. In pooled all three diseases, bin 9.3 and 6.5 reached genomic significant and suggestive evidence respectively, being relatively much weaker for 2DM/CAD or 2DM/obesity or CAD/obesity. Further, genomewide significant evidence was observed of bin 16.3 and 4.5 for 2DM/obesity, which is decreased when CAD was added. These findings indicated that bin 9.3 and 6.5 are most likely to be shared by 2DM, obesity and CAD. And bin 16.3 and 4.5 are potentially common regions to 2DM and obesity only. The observed shared susceptibility regions imply a partly overlapping genetic aspects of disease development. Fine scanning of these regions will definitely identify more susceptibility genes and causal variants.
Collapse
Affiliation(s)
- Chaoneng Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
21
|
Parikh M, Hetherington J, Sheth S, Seiler J, Ostrer H, Gerhard G, Wood C, Still C. Frequencies of obesity susceptibility alleles among ethnically and racially diverse bariatric patient populations. Surg Obes Relat Dis 2012; 9:436-41. [PMID: 22695173 DOI: 10.1016/j.soard.2012.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 04/23/2012] [Accepted: 04/24/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genetic factors likely play a role in obesity and the outcomes after bariatric surgery. Single nucleotide polymorphisms in or near the insulin-induced gene 2 (INSIG-2), fat mass and obesity-associated gene (FTO), melanocortin 4 receptor gene (MC4R), and proprotein convertase subtilisn/kexin type 1 gene (PCSK-1) have been associated with class III obesity in whites. Minimal data are available regarding the genetic susceptibility to obesity in class III obese nonwhites, especially Hispanics. Our objective was to perform a comparative analysis of 4 common genetic variants (INSIG-2, FTO, MC4R, and PCSK-1) associated with obesity in a diverse population of bariatric surgery patients to determine whether a difference exists by ethnicity (white versus Hispanic). The setting of the study was 2 university hospitals in the United States. METHODS Bariatric surgery patients from 2 different institutions were enrolled prospectively, and genotyping was performed. Differences in the distribution of INSIG-2, FTO, MC4R, and PCSK-1 single nucleotide polymorphisms among the different ethnicities (whites and Hispanics) were compared using an additive model (0, 1, or 2 risk alleles). A propensity-matched analysis was used to account for cohort differences. RESULTS A total of 1276 bariatric patients were genotyped for the INSIG-2, FTO, MC4R, and PCSK-1 obesity single nucleotide polymorphisms. Statistically significant differences in FTO, INSIG-2, MC4R, and PCSK-1 were seen using an additive model. FTO, PCSK-1, and MC4R (test for trend) remained significantly different in the propensity analysis. CONCLUSION Significant differences in the frequencies of several common obesity susceptibility variants in or near FTO, PCSK-1, and MC4R were found in white and Hispanic patients with class III obesity undergoing bariatric surgery. Larger studies in more class III obese Hispanics of different nationalities are needed.
Collapse
Affiliation(s)
- Manish Parikh
- Department of Surgery, New York University Medical Center/Bellevue Hospital Center, New York, NY 10016, USA.
| | | | | | | | | | | | | | | |
Collapse
|