51
|
Asadi M, Amoli M, Ansari Y, Far I, Pashaie N, Noroozi N. Association study of Melanocortin-4 Receptor (rs17782313) and PKHD1 (rs2784243) variations and early incidence of obesity at the age of maturity. ADVANCES IN HUMAN BIOLOGY 2022. [DOI: 10.4103/aihb.aihb_160_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
52
|
Aurora A and AKT Kinase Signaling Associated with Primary Cilia. Cells 2021; 10:cells10123602. [PMID: 34944109 PMCID: PMC8699881 DOI: 10.3390/cells10123602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of kinase signaling is associated with various pathological conditions, including cancer, inflammation, and autoimmunity; consequently, the kinases involved have become major therapeutic targets. While kinase signaling pathways play crucial roles in multiple cellular processes, the precise manner in which their dysregulation contributes to disease is dependent on the context; for example, the cell/tissue type or subcellular localization of the kinase or substrate. Thus, context-selective targeting of dysregulated kinases may serve to increase the therapeutic specificity while reducing off-target adverse effects. Primary cilia are antenna-like structures that extend from the plasma membrane and function by detecting extracellular cues and transducing signals into the cell. Cilia formation and signaling are dynamically regulated through context-dependent mechanisms; as such, dysregulation of primary cilia contributes to disease in a variety of ways. Here, we review the involvement of primary cilia-associated signaling through aurora A and AKT kinases with respect to cancer, obesity, and other ciliopathies.
Collapse
|
53
|
Narjabadifam M, Bonyadi M, Rafat SA, Mahdavi R, Aliasghari F. Association study of rs17782313 polymorphism near MC4R gene with obesity/overweight, BMI, and hedonic hunger among women from Northwestern Iran. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2021. [DOI: 10.3233/mnm-200530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Obesity, as a medical condition, results from interactions between environmental and genetic factors. The rs17782313 polymorphism, located 188kb downstream of the Melanocortin 4 Receptor (MC4R) gene, is one of the essential candidate genetic markers that has shown the highest association with obesity in different populations. OBJECTIVE: This study aimed to investigate the possible associations of rs17782313 polymorphism near the MC4R gene with obesity/overweight, body mass index (BMI), and hedonic hunger among women from the Iranian Azeri ethnic group. METHODS: Five hundred sixty-three women, composed of 396 patients with obesity/overweight and 167 unrelated healthy controls, were genotyped for the rs17782313 polymorphism by applying the polymerase chain reaction-restriction fragment length polymorphism (PCR–RFLP) method. RESULTS: This population was in Hardy-Weinberg equilibrium (P = 0.878). The study confirmed a significant association of rs17782313 with obesity, where subjects carrying the C/C genotype had higher odds of obesity (OR = 2.681, P = 0.005, 95%CI:1.340–5.365). Also, C allele carriers have statistically significantly higher BMI scores than those carrying the T allele (P = 0.029). However, no significant associations were found among PFS scores and genotypic/allelic groups of rs17782313 polymorphism (P = 0.368). CONCLUSIONS: Our findings suggest that rs17782313 polymorphism is strongly associated with obesity and BMI but not with hedonic hunger among Northwest Iran women. Moreover, the sequencing data analysis in several homozygous and heterozygous carriers of the C allele led to identifying a novel frameshift variant with TCT deletion (rs534212081) in the 166 upstream of rs17782313, which has not been reported so far.
Collapse
Affiliation(s)
- Mahan Narjabadifam
- Center of Excellence for Biodiversity, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Morteza Bonyadi
- Center of Excellence for Biodiversity, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Seyed Abbas Rafat
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Reza Mahdavi
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Aliasghari
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
54
|
Ahluwalia MK. Nutrigenetics and nutrigenomics-A personalized approach to nutrition. ADVANCES IN GENETICS 2021; 108:277-340. [PMID: 34844714 DOI: 10.1016/bs.adgen.2021.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prevalence of non-communicable diseases has been on an upward trajectory for some time and this puts an enormous burden on the healthcare expenditure. Lifestyle modifications including dietary interventions hold an immense promise to manage and prevent these diseases. Recent advances in genomic research provide evidence that focussing these efforts on individual variations in abilities to metabolize nutrients (nutrigenetics) and exploring the role of dietary compounds on gene expression (nutrigenomics and nutri-epigenomics) can lead to more meaningful personalized dietary strategies to promote optimal health. This chapter aims to provide examples on these gene-diet interactions at multiple levels to support the need of embedding targeted dietary interventions as a way forward to prevent, avoid and manage diseases.
Collapse
|
55
|
Toumba M, Fanis P, Vlachakis D, Neocleous V, Phylactou LA, Skordis N, Mantzoros CS, Pantelidou M. Molecular modelling of novel ADCY3 variant predicts a molecular target for tackling obesity. Int J Mol Med 2021; 49:10. [PMID: 34821371 PMCID: PMC8651229 DOI: 10.3892/ijmm.2021.5065] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/26/2021] [Indexed: 12/27/2022] Open
Abstract
Severe early-onset obesity is mainly attributed to single gene variations of the hypothalamic leptin-melanocortin system, which is critical for controlling the balance between appetite and energy expenditure. Adenylate cyclase 3 (ADCY3), a transmembrane enzyme localized in primary neuronal cilia, is a key genetic candidate, which appears to have an essential role in regulating body weight. The present study aimed to identify ADCY3 genetic variants in severely obese young patients of Greek-Cypriot origin by genomic sequencing. Apart from previously reported variants, the novel and probably pathogenic variant c.349T>A, causing a p.Leu117Met substitution within one of the two pseudo-symmetric halves of the transmembrane part of the protein, was reported. Molecular modelling analysis used to delineate bonding interactions within the mutated protein structure strongly suggested a change in interactive forces and energy levels affecting the pseudo-twofold symmetry of the transmembrane domain of the protein and probably its catalytic function. These results support the involvement of ADCY3 in the pathology of the disease and point towards the requirement of defining protein function and evaluating the clinical significance of the detected variants.
Collapse
Affiliation(s)
- Meropi Toumba
- Pediatric Endocrinology Clinic, Department of Paediatrics, Aretaeio Hospital, 2024 Nicosia, Cyprus
| | - Pavlos Fanis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, 1683 Nicosia, Cyprus
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Vassos Neocleous
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, 1683 Nicosia, Cyprus
| | - Leonidas A Phylactou
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, 1683 Nicosia, Cyprus
| | - Nicos Skordis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, 1683 Nicosia, Cyprus
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Maria Pantelidou
- Department of Pharmacy, School of Health Sciences, Frederick University Cyprus, 1036 Nicosia, Cyprus
| |
Collapse
|
56
|
Nadkarni GN, Fei K, Galarneau G, Gao Y, Wilson JG, Cooper R, Madden EB, Denny JC, Richardson LD, Pollak M, Loos RJF, Horowitz CR. APOL1 renal risk variants are associated with obesity and body composition in African ancestry adults: An observational genotype-phenotype association study. Medicine (Baltimore) 2021; 100:e27785. [PMID: 34766590 PMCID: PMC8589256 DOI: 10.1097/md.0000000000027785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 01/02/2023] Open
Abstract
ABSTRACT While increased obesity prevalence among persons of African ancestry (AAs) compared to persons of European ancestry (EAs) is linked to social, environmental and behavioral factors, there are no gene variants that are common and significantly associated with obesity in AA populations. We sought to explore the association between ancestry specific renal risk variants in the apolipoprotein L1 (APOL1) gene with obesity related traits in AAs.We conducted a genotype-phenotype association study from 3 electronic medical record linked cohorts (BioMe Biobank, BioVU, nuGENE); randomized controlled trials (genetic testing to understand and address renal disease disparities) and prospective cohort study (Jackson Heart Study). We analyzed association of APOL1 renal risk variants with cross-sectional measures of obesity (average body mass index (BMI), and proportion of overweight and obesity) and with measures of body composition (in Jackson Heart Study).We had data on 11,930 self-reported AA adults. Across cohorts, mean age was from 42 to 49 years and percentage female from 58% to 75.3%. Individuals who have 2 APOL1 risk alleles (14% of AAs) have 30% higher obesity odds compared to others (recessive model adjusted odds ratio 1.30; 95% confidence interval 1.16-1.41; P = 2.75 × 10-6). An additive model better fit the association, in which each allele (47% of AAs) increases obesity odds by 1.13-fold (adjusted odds ratio 1.13; 95% confidence interval 1.07-1.19; P = 3.07 × 10-6) and increases BMI by 0.36 kg/m2 (∼1 kg, for 1.7 m height; P = 2 × 10-4). APOL1 alleles are not associated with refined body composition traits overall but are significantly associated with fat free mass index in women [0.30 kg/m2 increment per allele; P = .03].Thus, renal risk variants in the APOL1 gene, found in nearly half of AAs, are associated with BMI and obesity in an additive manner. These variants could, either on their own or interacting with environmental factors, explain a proportion of ethnic disparities in obesity.
Collapse
Affiliation(s)
- Girish N. Nadkarni
- Department of Medicine, The Charles Bronfman Institute for Personalized Medicine
| | - Kezhen Fei
- Department of Population Health Science and Policy, The Institute for Health Equity Research
| | - Genevieve Galarneau
- Department of Genetics and Genomic Sciences, Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yan Gao
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Richard Cooper
- Department of Public Health Sciences, Loyola University, Chicago, IL
| | - Ebony B. Madden
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Joshua C. Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Lynne D. Richardson
- Department of Emergency Medicine, Department of Population Health Science and Policy, The Institute for Health Equity Research, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Martin Pollak
- Department of Medicine, Division of Nephrology, Beth Israel Deaconess School of Medicine, Boston, MA
| | - Ruth J. F. Loos
- Department of Environmental Medicine & Public Health, The Charles Bronfman Institute for Personalized Medicine
| | - Carol R. Horowitz
- Department of Population Health Science and Policy, Department of Medicine, The Institute for Health Equity Research, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
57
|
Adamska-Patruno E, Bauer W, Bielska D, Fiedorczuk J, Moroz M, Krasowska U, Czajkowski P, Wielogorska M, Maliszewska K, Puckowska S, Szczerbinski L, Lipinska D, Gorska M, Kretowski A. An Association between Diet and MC4R Genetic Polymorphism, in Relation to Obesity and Metabolic Parameters-A Cross Sectional Population-Based Study. Int J Mol Sci 2021; 22:ijms222112044. [PMID: 34769477 PMCID: PMC8584592 DOI: 10.3390/ijms222112044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
The melanocortin-4 receptor (MC4R) gene harbours one of the strongest susceptibility loci for obesity and obesity-related metabolic consequences. We analysed whether dietary factors may attenuate the associations between MC4R genotypes and obesity and metabolic parameters. In 819 participants genotyped for common MC4R polymorphisms (rs17782313, rs12970134, rs633265, and rs135034), the anthropometric measurements, body fat content and distribution (visceral and subcutaneous adipose tissue, VAT and SAT, respectively), and blood glucose, insulin, total-, LDL-, HDL-cholesterol, triglycerides concentrations, and daily macronutrient intake were assessed. ANOVA or Kruskal–Wallis tests were used, and multivariate linear regression models were developed. We observed that the CC genotype carriers (rs17782313) presented higher VAT, VAT/SAT ratio, fasting blood glucose and triglyceride concentrations when they were stratified to the upper quantiles of protein intake. An increase in energy derived from proteins was associated with higher BMI (Est. 5.74, R2 = 0.12), body fat content (Est. 8.44, R2 = 0.82), VAT (Est. 32.59, R2 = 0.06), and VAT/SAT ratio (Est. 0.96, R2 = 0.05). The AA genotype carriers (rs12970134) presented higher BMI, body fat, SAT and VAT, fasting blood glucose, triglycerides and total cholesterol concentrations. An increase in energy derived from proteins by AA carriers was associated with higher VAT (Est.19.95, R2 = 0.06) and VAT/SAT ratio (Est. 0.64, R2 = 0.05). Our findings suggest that associations of the common MC4R SNPs with obesity and its metabolic complications may be dependent on the daily dietary intake, which may open new areas for developing personalised diets for preventing and treating obesity and obesity-related comorbidities.
Collapse
Affiliation(s)
- Edyta Adamska-Patruno
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
- Correspondence:
| | - Witold Bauer
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Dorota Bielska
- Department of Family Medicine, Medical University of Bialystok, Mieszka I 4b, 15-054 Bialystok, Poland;
| | - Joanna Fiedorczuk
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
| | - Monika Moroz
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
| | - Urszula Krasowska
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Przemyslaw Czajkowski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Marta Wielogorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Sylwia Puckowska
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Lukasz Szczerbinski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Danuta Lipinska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| |
Collapse
|
58
|
Abstract
Obesity is one of the risk factors for the development and progression of chronic kidney disease (CKD). Several studies have shown the association between increased body mass index and kidney function decline. Obesity leads to CKD directly by acting as an independent risk factor and indirectly through increasing risks for diabetes, hypertension, and atherosclerosis, a group of well-established independent risk factors for CKD. Alterations in renal hemodynamics, inflammation, and in hormones and growth factors results in hyperfiltration injury and focal segmental glomerulosclerosis. In recent years, many studies have shown that the gut microbiome may play a role in the pathogenesis of obesity. Dysbiosis has been noted in obese subjects in both human and animal studies. Changes in the gut microbiome in obese patients promote weight gain by effectively extracting energy from diet, and induction of low-grade inflammation. Evidence also points to the role of inflammation within the adipose tissue in obesity as a key factor in the pathogenesis of obesity-related complications. Thus, obesity is the net result of complex interactions between behavioral, genetic, and environmental factors. In terms of management, conservative approaches are often the first option, but they often are unsuccessful in achieving and/or maintaining weight loss, particularly in severe obesity. Consequently, nonmedical management with bariatric surgery is the most effective treatment option for morbid obesity and has shown mitigation of multiple risk factors for the progression of CKD. The most frequently performed interventions are vertical sleeve gastrectomy and Roux-en-Y gastric bypass. Studies have shown that bariatric surgery is associated with beneficial effects on CKD by mitigating its risk factors by weight loss, reducing insulin resistance, hemoglobin A1c, and proteinuria, in addition to positive long-term outcomes. Because of the epidemic of obesity, the prevalence of obesity in kidney transplant recipients also is increasing. The maximal body mass index (BMI) threshold for kidney transplantation is not clear. The Organ Procurement Transplant Network/Scientific Registry of Transplant Recipients 2019 annual data report showed that the proportion of kidney transplant recipient candidates with a BMI of 30 kg/m2 or greater is increasing steadily. Morbid obesity is linked to adverse graft outcomes including delayed graft function, primary nonfunction, and decreased graft survival. Obesity is also an independent risk factor for cardiovascular death in kidney transplant recipients, suggesting that these patients should not be excluded from transplantation based on their BMI because transplantation is associated with lower mortality compared with dialysis. However, many centers exclude obese patients (with different BMI cut-off values) from transplantation to avoid postoperative complications. To minimize the surgical complications of kidney transplantation in obese patients, our center has adopted the robot-assisted kidney transplantation procedure. Our data show that this approach is comparable with historical nonobese controls in the United Network for Organ Sharing database in terms of patient and graft survival. Another surgical option for this group of patients at our center is a combined robotic sleeve gastrectomy and robotic-assisted kidney transplant. In a recent study, this approach showed promising results in terms of weight loss, patient survival, and graft survival, and might become more common in the future.
Collapse
|
59
|
Wang N, Tong R, Xu J, Tian Y, Pan J, Cui J, Chen H, Peng Y, Fei S, Yang S, Wang L, Yao J, Cui W. PDX1 and MC4R genetic polymorphisms are associated with type 2 diabetes mellitus risk in the Chinese Han population. BMC Med Genomics 2021; 14:249. [PMID: 34696776 PMCID: PMC8543917 DOI: 10.1186/s12920-021-01037-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/01/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a complex metabolic disease that is caused by a complex interplay between genetic and environmental factors. This research aimed to investigate the association of genetic polymorphisms in PDX1 and MC4R with T2DM risk. METHODS The genotypes of 10 selected SNPs in PDX1 and MC4R were identified using the Agena MassARRAY platform. We utilized odds ratio (OR) and 95% confidence intervals (CIs) to assess the correlation between genetic polymorphisms and T2DM risk. RESULTS We found that PDX1-rs9581943 decreased susceptibility to T2DM among in a Chinese Han population (OR = 0.76, p = 0.045). We also found that selected genetic polymorphisms in PDX1 and MC4R could modify the risk of T2DM, which might also be influenced by age, sex, BMI, smoking status, and drinking status (p < 0.05). CONCLUSIONS We concluded that PDX1 and MC4R genetic variants were significantly associated with T2DM risk in a Chinese Han population. These single polymorphic markers may be considered to be new targets in the assessment and prevention of T2DM among Chinese Han people.
Collapse
Affiliation(s)
- Ning Wang
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Rui Tong
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jing Xu
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yanni Tian
- Department of Oncology, East Branch of the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710089, Shaanxi, China
| | - Juan Pan
- Department of Endocrinology, Xianyang Central Hospital, Xianyang, 712000, Shaanxi, China
| | - Jiaqi Cui
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Huan Chen
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yanqi Peng
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Sijia Fei
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Shujun Yang
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Lu Wang
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Juanchuan Yao
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Wei Cui
- Department of Endocrinology and Second Department of Geriatrics, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
60
|
Aoun C, Hajj A, Hajj F, Papazian T, Rabbaa Khabbaz L. The interaction between genetic polymorphisms in FTO, MC4R and MTHFR genes and adherence to the Mediterranean Diet in relation to obesity. Gene 2021; 809:146037. [PMID: 34688820 DOI: 10.1016/j.gene.2021.146037] [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] [Received: 07/12/2021] [Revised: 10/04/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023]
Abstract
AIMS To investigate the potential interaction between genetic background and adherence to the Mediterranean Diet, macronutrient intake and physical activity with regard to obesity in a sample of healthy adults. DESIGN Cross-sectional epidemiological study including 392 adults living in the Mediterranean basin. Data including FFQ, IPAQ and sociodemographic questionnaires were collected via face-to-face interviews. Anthropometric measures were performed and saliva swab for DNA extraction. Two MD scores were calculated to assess the adherence of the population to this pattern. Three single nucleotid polymorphisms (SNPs) related to obesity were studied: FTO, MC4R, MTHFR. FINDINGS FTO rs9939609 is significantly associated with WHR, and MC4R with all phenotypic traits linked to obesity (BMI, WC and WHR). However, MTHFR polymorphism didn't show any significant correlation with anthropometric parameters. Adherence to the MD and high level of physical activity do not seem to protect against the occurrence of overweight and obesity in genetically predisposed subjects. CONCLUSION Classic lifestyle interventions are insufficient in addressing the challenging obesity pandemic. Identifying more genetic variants and understanding their interaction with lifestyle will improve the clinical outcome of these variants for risk prediction and personalized nutrition and medical therapy. Also, the MD should undergo a redefinition adapted to each country on the Mediterranean basin in order to organize public health measures for its comeback.
Collapse
Affiliation(s)
- Carla Aoun
- Department of Nutrition, Faculty of Pharmacy, Saint-Joseph University of Beirut, Beirut, Lebanon; Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments, Pôle Technologie- Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon.
| | - Aline Hajj
- Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments, Pôle Technologie- Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Fabienne Hajj
- Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments, Pôle Technologie- Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Tatiana Papazian
- Department of Nutrition, Faculty of Pharmacy, Saint-Joseph University of Beirut, Beirut, Lebanon; Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments, Pôle Technologie- Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | - Lydia Rabbaa Khabbaz
- Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments, Pôle Technologie- Santé (PTS), Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| |
Collapse
|
61
|
Polymorphisms on rs9939609 FTO and rs17782313 MC4R genes in children and adolescent obesity: A systematic review. Nutrition 2021; 91-92:111474. [PMID: 34628278 DOI: 10.1016/j.nut.2021.111474] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/18/2021] [Accepted: 08/24/2021] [Indexed: 11/23/2022]
Abstract
The aim of this review was to assess whether the presence of rs9939609 and rs17782313 polymorphisms increase the risk for obesity among children and adolescents. This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses checklist and it was registered in PROSPERO. The search was performed in the PubMed/Medline, The Cochrane Library, and Web of Science databases. The risk of bias of the studies was accessed using the Newcastle-Ottawa scale and JBI Critical Appraisal Checklist for Analytical. The search of the databases retrieved 859 references. Twelve studies were eligible to be included in this systematic review. Five studies founded a positive association between overweight and obesity in children and adolescents with the presence of the rs17783213 and four studies with rs9939609. Three studies did not find an association between overweight and obesity in children and adolescents with the presence of rs17782313 or rs9939609. One found a protective effect for obesity in individuals with risk A allele referring to rs9939609, one found a synergistic effect in relation to the presence of polymorphisms rs17782313 and rs9939609 for obese phenotype, and one observed that the presence together of the rs9939609, rs17782313, and rs12970134 MC4R were significant for the presence of obesity in children and adolescents. The results suggest that depending on the population evaluated and ethnicity, the polymorphisms rs17782313 and rs9939609 could be associated with overweight and obesity in children and adolescents.
Collapse
|
62
|
Burchat N, Sharma P, Ye H, Komakula SSB, Dobrzyn A, Vartanian V, Lloyd RS, Sampath H. Maternal Transmission of Human OGG1 Protects Mice Against Genetically- and Diet-Induced Obesity Through Increased Tissue Mitochondrial Content. Front Cell Dev Biol 2021; 9:718962. [PMID: 34604220 PMCID: PMC8480284 DOI: 10.3389/fcell.2021.718962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Obesity and related metabolic disorders are pressing public health concerns, raising the risk for a multitude of chronic diseases. Obesity is multi-factorial disease, with both diet and lifestyle, as well as genetic and developmental factors leading to alterations in energy balance. In this regard, a novel role for DNA repair glycosylases in modulating risk for obesity has been previously established. Global deletion of either of two different glycosylases with varying substrate specificities, Nei-like endonuclease 1 (NEIL1) or 8-oxoguanine DNA glycosylase-1 (OGG1), both predispose mice to diet-induced obesity (DIO). Conversely, enhanced expression of the human OGG1 gene renders mice resistant to obesity and adiposity. This resistance to DIO is mediated through increases in whole body energy expenditure and increased respiration in adipose tissue. Here, we report that hOGG1 expression also confers resistance to genetically-induced obesity. While Agouti obese (Ay/a) mice are hyperphagic and consequently develop obesity on a chow diet, hOGG1 expression in Ay/a mice (Ay/aTg) prevents increased body weight, without reducing food intake. Instead, obesity resistance in Ay/aTg mice is accompanied by increased whole body energy expenditure and tissue mitochondrial content. We also report for the first time that OGG1-mediated obesity resistance in both the Ay/a model and DIO model requires maternal transmission of the hOGG1 transgene. Maternal, but not paternal, transmission of the hOGG1 transgene is associated with obesity resistance and increased mitochondrial content in adipose tissue. These data demonstrate a critical role for OGG1 in modulating energy balance through changes in adipose tissue function. They also demonstrate the importance of OGG1 in modulating developmental programming of mitochondrial content and quality, thereby determining metabolic outcomes in offspring.
Collapse
Affiliation(s)
- Natalie Burchat
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Priyanka Sharma
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Hong Ye
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Sai Santosh Babu Komakula
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States.,Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Agnieszka Dobrzyn
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States.,Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Vladimir Vartanian
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| | - R Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Harini Sampath
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States.,Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States.,Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| |
Collapse
|
63
|
Dashti HS, Levy DE, Hivert MF, Alimenti K, McCurley JL, Saxena R, Thorndike AN. Genetic risk for obesity and the effectiveness of the ChooseWell 365 workplace intervention to prevent weight gain and improve dietary choices. Am J Clin Nutr 2021; 115:180-188. [PMID: 34581769 PMCID: PMC8755032 DOI: 10.1093/ajcn/nqab303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/26/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND It is unknown whether behavioral interventions to improve diet are effective in people with a genetic predisposition to obesity. OBJECTIVES To examine associations between BMI genetic risk and changes in weight and workplace purchases by employees participating in a randomized controlled trial of an automated behavioral workplace intervention to promote healthy food choices. METHODS Participants were hospital employees enrolled in a 12-mo intervention followed by a 12-mo follow-up. Hospital cafeterias utilized a traffic-light labeling system (e.g., green = healthy, red = unhealthy) that was used to calculate a validated Healthy Purchasing Score (HPS; higher = healthier). A weighted genome-wide BMI genetic score was generated by summing BMI-increasing alleles. RESULTS The study included 397 adults of European ancestry (mean age, 44.9 y; 80.9% female). Participants in the highest genetic quartile (Q4) had a lower HPS and higher purchases of red-labeled items relative to participants in the lowest quartile (Q1) at baseline [Q4-Q1 Beta HPS, -4.66 (95% CI, -8.01 to -1.32); red-labeled items, 4.26% (95% CI, 1.45%-7.07%)] and at the 12-mo [HPS, -3.96 (95% CI, -7.5 to -0.41); red-labeled items, 3.20% (95% CI, 0.31%-6.09%)] and 24-mo [HPS, -3.70 (95% CI, -7.40 to 0.00); red-labeled items, 3.48% (95% CI, 0.54%-6.41%)] follow-up periods. In the intervention group, increases in HPS were similar in Q4 and Q1 at 12 mo (Q4-Q1 Beta, 1.04; 95% CI, -2.42 to 4.50). At the 24-mo follow-up, the change in BMI from baseline was similar between Q4 and Q1 (0.17 kg/m2; 95% CI, -0.55 to 0.89 kg/m2) in the intervention group, but higher in Q4 than Q1 (1.20 kg/m2; 95% CI, 0.26-2.13 kg/m2) in the control group. No interaction was evident between the treatment arm and genetic score for BMI or HPS. CONCLUSIONS Having a high BMI genetic risk was associated with greater increases in BMI and lower quality purchases over 2 y. The 12-mo behavioral intervention improved employees' food choices, regardless of the genetic burden, and may have attenuated weight gain conferred by having the genetic risk.
Collapse
Affiliation(s)
| | - Douglas E Levy
- Mongan Institute Health Policy Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA,Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Kaitlyn Alimenti
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica L McCurley
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA,Department of Medical and Population Genetics, Broad Institute, Cambridge, MA, USA,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anne N Thorndike
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
64
|
Goulet D, O'Loughlin J, Sylvestre MP. Association of Genetic Variants With Body-Mass Index and Blood Pressure in Adolescents: A Replication Study. Front Genet 2021; 12:690335. [PMID: 34539733 PMCID: PMC8440872 DOI: 10.3389/fgene.2021.690335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
The strong correlation between adiposity and blood pressure (BP) might be explained in part by shared genetic risk factors. A recent study identified three nucleotide variants [rs16933812 (PAX5), rs7638110 (MRPS22), and rs9930333 (FTO)] associated with both body mass index (BMI) and systolic blood pressure (SBP) in adolescents age 12-18years. We attempted to replicate these findings in a sample of adolescents of similar age. A total of 713 adolescents were genotyped and had anthropometric indicators and blood pressure measured at age 13, 15, 17, and 24years. Using linear mixed models, we assessed associations of these variants with BMI and SBP. In our data, rs9930333 (FTO) was associated with body mass index, but not systolic blood pressure. Neither rs16933812 (PAX5) nor rs7638110 (MRPS22) were associated with body mass index or systolic blood pressure. Although, differences in phenotypic definitions and in genetic architecture across populations may explain some of the discrepancy across studies, nucleotide variant selection in the initial study may have led to false-positive results that could not be replicated.
Collapse
Affiliation(s)
- Danick Goulet
- École de santé publique, Université de Montréal, Montréal, QC, Canada
| | - Jennifer O'Loughlin
- École de santé publique, Université de Montréal, Montréal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Marie-Pierre Sylvestre
- École de santé publique, Université de Montréal, Montréal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| |
Collapse
|
65
|
Dastgheib SA, Bahrami R, Setayesh S, Salari S, Mirjalili SR, Noorishadkam M, Sadeghizadeh-Yazdi J, Akbarian E, Neamatzadeh H. Evidence from a meta-analysis for association of MC4R rs17782313 and FTO rs9939609 polymorphisms with susceptibility to obesity in children. Diabetes Metab Syndr 2021; 15:102234. [PMID: 34364300 DOI: 10.1016/j.dsx.2021.102234] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIM The aim of this study was to evaluate the association of MC4R rs17782313 and FTO rs9939609 polymorphisms with childhood obesity. METHODS A universal search was performed up to May 2021. RESULTS A total of 31 studies including 13 studies with 9565 cases and 11956 controls on MC4R rs17782313 and 18 studies with 4789 cases and 15918 controls on FTO rs9939609 were selected. CONCLUSIONS Pooled data showed that FTO rs9930506 and MC4R rs17782313 polymorphisms were significantly associated with obesity in children. Stratified analyses revealed that these genetic variants were associated with childhood obesity in Caucasian and Asian children.
Collapse
Affiliation(s)
- Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sepideh Setayesh
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedali Salari
- Department of Biology, Science and Arts University, Yazd, Iran
| | - Seyed Reza Mirjalili
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahmood Noorishadkam
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Jalal Sadeghizadeh-Yazdi
- Department of Food Science and Technology, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elahe Akbarian
- Children Growth Disorder Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
66
|
|
67
|
Analysis of the Potential Genetic Links between Psoriasis and Cardiovascular Risk Factors. Int J Mol Sci 2021; 22:ijms22169063. [PMID: 34445769 PMCID: PMC8396451 DOI: 10.3390/ijms22169063] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/20/2021] [Accepted: 08/21/2021] [Indexed: 01/07/2023] Open
Abstract
Cardiovascular risk factors are one of the most common comorbidities in psoriasis. A higher prevalence of hypertension, insulin resistance and type 2 diabetes, dyslipidemia, obesity, metabolic syndrome, depression, as well as cardiovascular disease was confirmed in psoriatic patients in comparison to the general population. Data suggest that psoriasis and systemic inflammatory disorders may originate from the pleiotropic interactions with many genetic pathways. In this review, the authors present the current state of knowledge on the potential genetic links between psoriasis and cardiovascular risk factors. The understanding of the processes linking psoriasis with cardiovascular risk factors can lead to improvement of psoriasis management in the future.
Collapse
|
68
|
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
|
69
|
Mardones L, Parra-Valencia E, Petermann-Rocha F, Martínez-Sanguinetti MA, Leiva-Ordoñez AM, Lasserre-Laso N, Martorell M, Ulloa N, Sanhueza E, Pérez-Bravo F, Celis-Morales C, Villagrán M. The rs483145 polymorphism of MC4R gene is not associated with obesity in the Chilean population: Results of GENADIO study. ENDOCRINOL DIAB NUTR 2021; 69:S2530-0164(21)00156-7. [PMID: 34340956 DOI: 10.1016/j.endinu.2021.06.001] [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: 02/05/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The melanocortin receptor 4 (MC4R) participates in the control of appetite at the level of the central nervous system, through the leptin-melanocortin pathway. An association between different polymorphisms of the MC4R gene and obesity has been reported. However, there are few studies of the rs483145 single nucleotide polymorphism (SNP) of this gene. OBJECTIVE To investigate its prevalence and association with adiposity markers in Chilean adults. METHODS The prevalence of SNP rs483145, of the MC4R gene, was determined in 259 participants of the GENADIO study (genes, environment, diabetes and obesity) by means of real-time polymerase chain reaction (PCR). The association between the risk allele of MC4R (A) and adiposity markers (body weight, body mass index, fat mass percentage, hip circumference, waist circumference, waist-to-hip ratio) was performed by linear regression analysis and adjusted for confusion variables (socio-demographic and physic activity) using three statistical models. RESULTS It was determined that the prevalence of the risk allele (A) of the SNP rs483145 of the MC4R gene is 24.5% in the Chilean adult population included in this study, without finding an association with any of the adiposity markers studied, both in adjusted and unadjusted models. CONCLUSION The presence of the risk allele of SNP rs483145 of the MC4R gene is not associated with adiposity markers in the Chilean adult population studied. New studies with a bigger sample size will be necessary to confirm these results.
Collapse
Affiliation(s)
- Lorena Mardones
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Esteban Parra-Valencia
- Departamento de Ciencias Clínicas y Pre-Clínicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Fanny Petermann-Rocha
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Ana María Leiva-Ordoñez
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Nicole Lasserre-Laso
- Escuela de Nutrición y Dietética, Facultad de Salud, Universidad Santo Tomas, Región Metropolitana, Chile
| | - Miquel Martorell
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile; Departamento de Nutrición y Dietética, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Natalia Ulloa
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile; Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Eduardo Sanhueza
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisco Pérez-Bravo
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Carlos Celis-Morales
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Centro de Investigación en Fisiología del Ejercicio (CIFE), Universidad Mayor, Santiago, Chile; Laboratorio de Rendimiento Humano, Grupo de Estudio en Educación, Actividad Física y Salud (GEEAFyS), Universidad Católica del Maule, Talca, Chile
| | - Marcelo Villagrán
- Laboratorio de Investigación en Ciencias Biomédicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
70
|
Akbari P, Gilani A, Sosina O, Kosmicki JA, Khrimian L, Fang YY, Persaud T, Garcia V, Sun D, Li A, Mbatchou J, Locke AE, Benner C, Verweij N, Lin N, Hossain S, Agostinucci K, Pascale JV, Dirice E, Dunn M, Kraus WE, Shah SH, Chen YDI, Rotter JI, Rader DJ, Melander O, Still CD, Mirshahi T, Carey DJ, Berumen-Campos J, Kuri-Morales P, Alegre-Díaz J, Torres JM, Emberson JR, Collins R, Balasubramanian S, Hawes A, Jones M, Zambrowicz B, Murphy AJ, Paulding C, Coppola G, Overton JD, Reid JG, Shuldiner AR, Cantor M, Kang HM, Abecasis GR, Karalis K, Economides AN, Marchini J, Yancopoulos GD, Sleeman MW, Altarejos J, Della Gatta G, Tapia-Conyer R, Schwartzman ML, Baras A, Ferreira MAR, Lotta LA. Sequencing of 640,000 exomes identifies GPR75 variants associated with protection from obesity. Science 2021; 373:373/6550/eabf8683. [PMID: 34210852 DOI: 10.1126/science.abf8683] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/17/2021] [Indexed: 12/11/2022]
Abstract
Large-scale human exome sequencing can identify rare protein-coding variants with a large impact on complex traits such as body adiposity. We sequenced the exomes of 645,626 individuals from the United Kingdom, the United States, and Mexico and estimated associations of rare coding variants with body mass index (BMI). We identified 16 genes with an exome-wide significant association with BMI, including those encoding five brain-expressed G protein-coupled receptors (CALCR, MC4R, GIPR, GPR151, and GPR75). Protein-truncating variants in GPR75 were observed in ~4/10,000 sequenced individuals and were associated with 1.8 kilograms per square meter lower BMI and 54% lower odds of obesity in the heterozygous state. Knock out of Gpr75 in mice resulted in resistance to weight gain and improved glycemic control in a high-fat diet model. Inhibition of GPR75 may provide a therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Parsa Akbari
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Ankit Gilani
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Olukayode Sosina
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Jack A Kosmicki
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Lori Khrimian
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Yi-Ya Fang
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Trikaldarshi Persaud
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Victor Garcia
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Dylan Sun
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Alexander Li
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Joelle Mbatchou
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Adam E Locke
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Christian Benner
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Niek Verweij
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Nan Lin
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Sakib Hossain
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Kevin Agostinucci
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Jonathan V Pascale
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Ercument Dirice
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Michael Dunn
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | | | - William E Kraus
- Division of Cardiology, Duke University Medical Center, Durham, NC 27710, USA.,Duke Center for Living, Duke University Medical Center, Durham, NC 27705, USA
| | - Svati H Shah
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Yii-Der I Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, 221 00 Malmö, Sweden.,Department of Emergency and Internal Medicine, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Christopher D Still
- Geisinger Obesity Institute, Geisinger Health System, Danville, PA 17882, USA
| | - Tooraj Mirshahi
- Geisinger Obesity Institute, Geisinger Health System, Danville, PA 17882, USA
| | - David J Carey
- Geisinger Obesity Institute, Geisinger Health System, Danville, PA 17882, USA
| | - Jaime Berumen-Campos
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, 4360 Ciudad de México, Mexico
| | - Pablo Kuri-Morales
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, 4360 Ciudad de México, Mexico
| | - Jesus Alegre-Díaz
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, 4360 Ciudad de México, Mexico
| | - Jason M Torres
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, England, UK
| | - Jonathan R Emberson
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, England, UK
| | - Rory Collins
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, England, UK
| | | | - Alicia Hawes
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Marcus Jones
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | | | - Charles Paulding
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Giovanni Coppola
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - John D Overton
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Jeffrey G Reid
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Alan R Shuldiner
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Michael Cantor
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Hyun M Kang
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Goncalo R Abecasis
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Katia Karalis
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Aris N Economides
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA.,Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Jonathan Marchini
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | - Mark W Sleeman
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | - Giusy Della Gatta
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Roberto Tapia-Conyer
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, 4360 Ciudad de México, Mexico
| | - Michal L Schwartzman
- Department of Pharmacology and Medicine, New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Aris Baras
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA.
| | - Manuel A R Ferreira
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Luca A Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA.
| |
Collapse
|
71
|
Pharmacological potential of ferulic acid for the treatment of metabolic diseases and its mechanism of action: A review. PHYSIOLOGY AND PHARMACOLOGY 2021. [DOI: 10.52547/phypha.26.4.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
72
|
Nono Nankam PA, Blüher M. Retinol-binding protein 4 in obesity and metabolic dysfunctions. Mol Cell Endocrinol 2021; 531:111312. [PMID: 33957191 DOI: 10.1016/j.mce.2021.111312] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022]
Abstract
Excessive increased adipose tissue mass in obesity is associated with numerous co-morbid disorders including increased risk of type 2 diabetes, fatty liver disease, hypertension, dyslipidemia, cardiovascular diseases, dementia, airway disease and some cancers. The causal mechanisms explaining these associations are not fully understood. Adipose tissue is an active endocrine organ that secretes many adipokines, cytokines and releases metabolites. These biomolecules referred to as adipocytokines play a significant role in the regulation of whole-body energy homeostasis and metabolism by influencing and altering target tissues function. Understanding the mechanisms of adipocytokine actions represents a hot topic in obesity research. Among several secreted bioactive signalling molecules from adipose tissue and liver, retinol-binding protein 4 (RBP4) has been associated with systemic insulin resistance, dyslipidemia, type 2 diabetes and other metabolic diseases. Here, we aim to review and discuss the current knowledge on RBP4 with a focus on its role in the pathogenesis of obesity comorbid diseases.
Collapse
Affiliation(s)
- Pamela A Nono Nankam
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Germany.
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Germany; Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Germany
| |
Collapse
|
73
|
Rana S, Sultana A, Bhatti AA. Effect of interaction between obesity-promoting genetic variants and behavioral factors on the risk of obese phenotypes. Mol Genet Genomics 2021; 296:919-938. [PMID: 33966103 DOI: 10.1007/s00438-021-01793-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/22/2021] [Indexed: 01/28/2023]
Abstract
The studies investigating gene-gene and gene-environment (or gene-behavior) interactions provide valuable insight into the pathomechanisms underlying obese phenotypes. The Pakistani population due to its unique characteristics offers numerous advantages for conducting such studies. In this view, the current study was undertaken to examine the effects of gene-gene and gene-environment/behavior interactions on the risk of obesity in a sample of Pakistani population. A total of 578 adult participants including 290 overweight/obese cases and 288 normal-weight controls were involved. The five key obesity-associated genetic variants namely MC4R rs17782313, BDNF rs6265, FTO rs1421085, TMEM18 rs7561317, and NEGR1 rs2815752 were genotyped using the TaqMan allelic discrimination assays. The data related to behavioral factors, such as eating pattern, diet consciousness, the tendency toward fat-dense food (TFDF), sleep duration, sleep-wake cycle (SWC), shift work (SW), and physical activity levels were collected via a questionnaire. Gene-gene and gene-behavior interactions were analyzed by multifactor dimensionality reduction and linear regression, respectively. In our study, only TMEM18 rs7561317 was found to be significantly associated with anthropometric traits with no significant effect of gene-gene interactions were observed on obesity-related phenotypes. However, the genetic variants were found to interact with the behavioral factors to significantly influence various obesity-related anthropometric traits including BMI, waist circumference, hip circumference, waist-to-hip ratio, waist-to-height ratio, and percentage of body fat. In conclusion, the interaction between genetic architecture and behavior/environment determines the outcome of obesity-related anthropometric phenotypes. Thus, gene-environment/behavior interaction studies should be promoted to explore the risk of complex and multifactorial disorders, such as obesity.
Collapse
Affiliation(s)
- Sobia Rana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Ayesha Sultana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Adil Anwar Bhatti
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
74
|
Dashti HS, Ordovás JM. Genetics of Sleep and Insights into Its Relationship with Obesity. Annu Rev Nutr 2021; 41:223-252. [PMID: 34102077 DOI: 10.1146/annurev-nutr-082018-124258] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Considerable recent advancements in elucidating the genetic architecture of sleep traits and sleep disorders may provide insight into the relationship between sleep and obesity. Despite the considerable involvement of the circadian clock in sleep and metabolism, few shared genes, including FTO, were implicated in genome-wide association studies (GWASs) of sleep and obesity. Polygenic scores composed of signals from GWASs of sleep traits show largely null associations with obesity, suggesting lead variants are unique to sleep. Modest genome-wide genetic correlations are observed between many sleep traits and obesity and are largest for snoring.Notably, U-shaped positive genetic correlations with body mass index (BMI) exist for both short and long sleep durations. Findings from Mendelian randomization suggest robust causal effects of insomnia on higher BMI and, conversely, of higher BMI on snoring and daytime sleepiness. Bidirectional effects between sleep duration and daytime napping with obesity may also exist. Limited gene-sleep interaction studies suggest that achieving favorable sleep, as part of a healthy lifestyle, may attenuate genetic predisposition to obesity, but whether these improvements produce clinically meaningful reductions in obesity risk remains unclear. Investigations of the genetic link between sleep and obesity for sleep disorders other than insomnia and in populations of non-European ancestry are currently limited. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Hassan S Dashti
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; .,Broad Institute, Cambridge, Massachusetts 02142, USA
| | - José M Ordovás
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA.,Precision Nutrition and Obesity Program, IMDEA Alimentación, 28049 Madrid, Spain
| |
Collapse
|
75
|
Yeo GSH, Chao DHM, Siegert AM, Koerperich ZM, Ericson MD, Simonds SE, Larson CM, Luquet S, Clarke I, Sharma S, Clément K, Cowley MA, Haskell-Luevano C, Van Der Ploeg L, Adan RAH. The melanocortin pathway and energy homeostasis: From discovery to obesity therapy. Mol Metab 2021; 48:101206. [PMID: 33684608 PMCID: PMC8050006 DOI: 10.1016/j.molmet.2021.101206] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Over the past 20 years, insights from human and mouse genetics have illuminated the central role of the brain leptin-melanocortin pathway in controlling mammalian food intake, with genetic disruption resulting in extreme obesity, and more subtle polymorphic variations influencing the population distribution of body weight. At the end of 2020, the U.S. Food and Drug Administration (FDA) approved setmelanotide, a melanocortin 4 receptor agonist, for use in individuals with severe obesity due to either pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency. SCOPE OF REVIEW Herein, we chart the melanocortin pathway's history, explore its pharmacology, genetics, and physiology, and describe how a neuropeptidergic circuit became an important druggable obesity target. MAJOR CONCLUSIONS Unravelling the genetics of the subset of severe obesity has revealed the importance of the melanocortin pathway in appetitive control; coupling this with studying the molecular pharmacology of compounds that bind melanocortin receptors has brought a new obesity drug to the market. This process provides a drug discovery template for complex disorders, which for setmelanotide took 25 years to transform from a single gene into an approved drug.
Collapse
Affiliation(s)
- Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | | | - Anna-Maria Siegert
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Zoe M Koerperich
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | - Mark D Ericson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | - Stephanie E Simonds
- Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute, and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Courtney M Larson
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France.
| | - Iain Clarke
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3010, Australia.
| | | | - Karine Clément
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France, Sorbonne Université, INSERM, Nutrition and Obesity: Systemic Approaches (NutriOmics) Research Unit, Paris, France.
| | - Michael A Cowley
- Metabolism, Diabetes, and Obesity Programme, Monash Biomedicine Discovery Institute, and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA 55455.
| | | | - Roger A H Adan
- Department of Translational Neuroscience, UMCU Brain Centre, University Medical Centre Utrecht, Utrecht University, the Netherlands; Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sweden.
| |
Collapse
|
76
|
Farooqi S. Putting a brake on hunger. Science 2021; 372:792-793. [PMID: 34016769 DOI: 10.1126/science.abi8942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
77
|
Birth weight, childhood obesity and risk of hypertension: a Mendelian randomization study. J Hypertens 2021; 39:1876-1883. [PMID: 34001814 DOI: 10.1097/hjh.0000000000002871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSES Observational studies indicate that birth weight and childhood obesity are associated with essential hypertension, but their causal effect on essential hypertension remains unclear. The aim of our study is to elucidate the causal relationship between birth weight, childhood obesity, and essential hypertension by Mendelian randomization (MR) with genetic variants as instrumental variables (IVs). METHODS We identified IVs based on single nucleotide polymorphisms (SNPs) associated with birth weight (n = 160 295) and childhood obesity (n = 6889, 1509 cases and 5380 controls) from the meta-analysis of a genome-wide association study. Summary level data from the UK Biobank essential hypertension consortium (n = 463 010, 54 358 cases and 408 652 controls) was used to analyze the relationship between IVs and essential hypertension. Two MR analysis methods, two threshold values of selecting IVs, and leave-one-out analysis were used to ensure the robustness of the results. RESULTS Genetic predisposition to higher birth weight did not increase the risk of essential hypertension. In contrast, per one standard deviation increase in childhood body mass index was significantly associated with an increased risk of essential hypertension (odds ratio = 1.0075, 95% confidence interval: 1.0035-1.0116) when using seven SNPs that achieved genome-wide significance (P < 5 × 10-8). Sensitivity analysis and MR-Egger regression indicated that the results were robust and not influenced by pleiotropy. CONCLUSIONS No evidence of an association between birth weight and essential hypertension was found. Childhood obesity, however, showed a causal relationship with the risk of essential hypertension, which was helpful to understand the mechanisms of essential hypertension and develop strategies for its prevention.
Collapse
|
78
|
Wang Y, Bernard A, Comblain F, Yue X, Paillart C, Zhang S, Reiter JF, Vaisse C. Melanocortin 4 receptor signals at the neuronal primary cilium to control food intake and body weight. J Clin Invest 2021; 131:142064. [PMID: 33938449 DOI: 10.1172/jci142064] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 03/18/2021] [Indexed: 01/02/2023] Open
Abstract
The melanocortin 4 receptor (MC4R) plays a critical role in the long-term regulation of energy homeostasis, and mutations in the MC4R are the most common cause of monogenic obesity. However, the precise molecular and cellular mechanisms underlying the maintenance of energy balance within MC4R-expressing neurons are unknown. We recently reported that the MC4R localizes to the primary cilium, a cellular organelle that allows for partitioning of incoming cellular signals, raising the question of whether the MC4R functions in this organelle. Here, using mouse genetic approaches, we found that cilia were required specifically on MC4R-expressing neurons for the control of energy homeostasis. Moreover, these cilia were critical for pharmacological activators of the MC4R to exert an anorexigenic effect. The MC4R is expressed in multiple brain regions. Using targeted deletion of primary cilia, we found that cilia in the paraventricular nucleus of the hypothalamus (PVN) were essential to restrict food intake. MC4R activation increased adenylyl cyclase (AC) activity. As with the removal of cilia, inhibition of AC activity in the cilia of MC4R-expressing neurons of the PVN caused hyperphagia and obesity. Thus, the MC4R signaled via PVN neuron cilia to control food intake and body weight. We propose that defects in ciliary localization of the MC4R cause obesity in human inherited obesity syndromes and ciliopathies.
Collapse
Affiliation(s)
- Yi Wang
- Department of Medicine and The Diabetes Center and
| | | | | | - Xinyu Yue
- Department of Medicine and The Diabetes Center and
| | | | - Sumei Zhang
- Department of Medicine and The Diabetes Center and
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, UCSF, San Francisco, California, USA.,Chan Zuckerberg Biohub, San Francisco, California, USA
| | | |
Collapse
|
79
|
Association of the MC4R rs17782313 polymorphism with plasma ghrelin, leptin, IL6 and TNFα concentrations, food intake and eating behaviors in morbidly obese women. Eat Weight Disord 2021; 26:1079-1087. [PMID: 32918257 DOI: 10.1007/s40519-020-01003-5] [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: 04/13/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022] Open
Abstract
PURPOSE The rs17782313 variant of the MC4R gene plays an important role in the obesity phenotype. Studies that evaluate environmental factors and genetic variants associated with obesity may represent a great advance in understanding the development of this disease. This work seeks to assess the association of the polymorphism of MC4R rs17782313 on plasma parameters, including leptin, ghrelin, tumor necrosis factor (TNFα) and interleukin 6 (IL6), and on the eating behaviors of morbidly obese women. METHODS 70 adult women with BMI between 40 and 60 kg/m2 were recruited. Laboratory and anthropometric data were recorded. Using a visual analog scale (VAS), the feelings of hunger and satiety were evaluated. The presence or absence of binge eating was evaluated through the Binge Eating Scale (BES) questionnaire. Habitual food intake was analyzed using 3-day dietary records. TaqMan® assays were conducted using real-time PCR to assess genotype polymorphism variants from peripheral blood DNA. RESULTS This study found that female patients with the MC4R rs17782313 polymorphism had high levels of ghrelin and reduced levels of IL6 in the postprandial period. We observed a higher prevalence of severe binge eating in more than 50% of women with at least one risk allele. CONCLUSION Our hypothesis is that the MC4R rs17782313 polymorphism may influence the release of ghrelin, even without being associated with feelings of hunger and satiety. More than half of women with this polymorphism exhibited severe binge eating. LEVEL OF EVIDENCE Level III: case-control analytic study.
Collapse
|
80
|
Bouchard C. Genetics of Obesity: What We Have Learned Over Decades of Research. Obesity (Silver Spring) 2021; 29:802-820. [PMID: 33899337 DOI: 10.1002/oby.23116] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
There is a genetic component to human obesity that accounts for 40% to 50% of the variability in body weight status but that is lower among normal weight individuals (about 30%) and substantially higher in the subpopulation of individuals with obesity and severe obesity (about 60%-80%). The appreciation that heritability varies across classes of BMI represents an important advance. After controlling for BMI, ectopic fat and fat distribution traits are characterized by heritability levels ranging from 30% to 55%. Defects in at least 15 genes are the cause of monogenic obesity cases, resulting mostly from deficiencies in the leptin-melanocortin signaling pathway. Approximately two-thirds of the BMI heritability can be imputed to common DNA variants, whereas low-frequency and rare variants explain the remaining fraction. Diminishing allele effect size is observed as the number of obesity-associated variants expands, with most BMI-increasing or -decreasing alleles contributing only a few grams or less to body weight. Obesity-promoting alleles exert minimal effects in normal weight individuals but have larger effects in individuals with a proneness to obesity, suggesting a higher penetrance; however, it is not known whether these larger effect sizes precede obesity or are caused by an obese state. The obesity genetic risk is conditioned by thousands of DNA variants that make genetically based obesity prevention and treatment a major challenge.
Collapse
Affiliation(s)
- Claude Bouchard
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
81
|
Jenkins DA, Wade KH, Carslake D, Bowden J, Sattar N, Loos RJF, Timpson NJ, Sperrin M, Rutter MK. Estimating the causal effect of BMI on mortality risk in people with heart disease, diabetes and cancer using Mendelian randomization. Int J Cardiol 2021; 330:214-220. [PMID: 33592239 DOI: 10.1016/j.ijcard.2021.02.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/02/2021] [Accepted: 02/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Observational data have reported that being overweight or obese, compared to being normal weight, is associated with a lower risk for death - the "obesity paradox". We used Mendelian randomization (MR) to estimate causal effects of body mass index (BMI) on mortality risks in people with coronary heart disease (CHD), type 2 diabetes mellitus (T2DM) or malignancy in whom this paradox has been often reported. METHODS We studied 457,746 White British UK Biobank participants including three subgroups with T2DM (n = 19,737), CHD (n = 21,925) or cancer (n = 42,612) at baseline and used multivariable-adjusted Cox models and MR approaches to describe relationships between BMI and mortality risk. RESULTS Observational Cox models showed J-shaped relationships between BMI and mortality risk including within disease subgroups in which the BMI values associated with minimum mortality risk were within overweight/obese ranges (26.5-32.5 kg/m2). In all participants, MR analyses showed a positive linear causal effect of BMI on mortality risk (HR for mortality per unit higher BMI: 1.05; 95% CI: 1.03-1.08), also evident in people with CHD (HR: 1.08; 95% CI: 1.01-1.14). Point estimates for hazard ratios across all BMI values in participants with T2DM and cancer were consistent with overall positive linear effects but confidence intervals included the null. CONCLUSION These data support the idea that population efforts to promote intentional weight loss towards the normal BMI range would reduce, not enhance, mortality risk in the general population including, importantly, individuals with CHD.
Collapse
Affiliation(s)
- David A Jenkins
- NIHR Greater Manchester Patient Safety Translational Research Centre, The University of Manchester, Manchester, UK; School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PL, UK.
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, Population Health Sciences, Oakfield House, Oakfield Grove, University of Bristol, Bristol BS8 2BN, UK
| | - David Carslake
- MRC Integrative Epidemiology Unit, Population Health Sciences, Oakfield House, Oakfield Grove, University of Bristol, Bristol BS8 2BN, UK
| | - Jack Bowden
- MRC Integrative Epidemiology Unit, Population Health Sciences, Oakfield House, Oakfield Grove, University of Bristol, Bristol BS8 2BN, UK; Exeter Diabetes Group (ExCEED), College of Medicine and Health, University of Exeter, Exeter, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, Population Health Sciences, Oakfield House, Oakfield Grove, University of Bristol, Bristol BS8 2BN, UK
| | - Matthew Sperrin
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Martin K Rutter
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Rd, Manchester M13 9PL, UK; Diabetes, Endocrinology and Metabolism Centre, Peter Mount Building, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 0HY, UK
| |
Collapse
|
82
|
Walia GK, Saini S, Vimal P, Bhatia K, Kumar A, Singh R, Prabhakaran D, Gupta V. Association of MC4R (rs17782313) gene polymorphism with obesity measures in Western India. Diabetes Metab Syndr 2021; 15:661-665. [PMID: 33813238 DOI: 10.1016/j.dsx.2021.03.013] [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: 07/07/2020] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS The association of melanocortin receptor 4 (MC4R) gene with adiposity measures is widely studied in European populations. Only six studies have investigated the role of MC4R gene with adiposity measures among Indian populations. We have evaluated the role of MC4R (rs17782313) gene polymorphism in influencing adiposity measures in India among children and adults. MATERIALS AND METHODS The present population based cross sectional study was conducted among 303 individuals (208 children and 95 adults) of age group 10-30 years, belonging to Rajasthan. Somatometric measurements (standing height, weight, and waist and hip girths) and blood samples were taken after obtaining written informed consent. Genotyping of MC4R rs17782313 single nucleotide polymorphism was done using restriction fragment length polymorphism method for polymerase chain reaction amplified fragments. We examined association between rs17782313 and different adiposity measures (height, weight, BMI, WHR, and waist and hip girths) using linear regression models. RESULTS The MC4R variant (rs17782313) predicted increased body weight (0.15 kg, S.E ± 0.076, P = 0.043) among children. In combined population, the rs17782313 variant was moderately associated with body weight (0.13 kg, S.E ± 0.070, P = 0.057). This variant was not found to be associated with any other adiposity measure. CONCLUSION Further studies are needed to evaluate the association of MC4R variants through sequencing and functional genomics with different adiposity measures in Indian populations for understanding the genetic underpinnings of adiposity in India.
Collapse
Affiliation(s)
| | - Simmi Saini
- Department of Anthropology, University of Delhi, India.
| | - Pradeep Vimal
- Indian Institute of Public Health- Delhi, Public Health Foundation of India, Gurugram, India.
| | | | - Arun Kumar
- Department of Anthropology, University of Delhi, India.
| | - Ranjana Singh
- Indian Institute of Public Health- Delhi, Public Health Foundation of India, Gurugram, India.
| | | | - Vipin Gupta
- Department of Anthropology, University of Delhi, India.
| |
Collapse
|
83
|
Gordon-Larsen P, French JE, Moustaid-Moussa N, Voruganti VS, Mayer-Davis EJ, Bizon CA, Cheng Z, Stewart DA, Easterbrook JW, Shaikh SR. Synergizing Mouse and Human Studies to Understand the Heterogeneity of Obesity. Adv Nutr 2021; 12:2023-2034. [PMID: 33885739 PMCID: PMC8483969 DOI: 10.1093/advances/nmab040] [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] [Received: 12/15/2020] [Revised: 02/17/2021] [Accepted: 03/13/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is routinely considered as a single disease state, which drives a "one-size-fits-all" approach to treatment. We recently convened the first annual University of North Carolina Interdisciplinary Nutrition Sciences Symposium to discuss the heterogeneity of obesity and the need for translational science to advance understanding of this heterogeneity. The symposium aimed to advance scientific rigor in translational studies from animal to human models with the goal of identifying underlying mechanisms and treatments. In this review, we discuss fundamental gaps in knowledge of the heterogeneity of obesity ranging from cellular to population perspectives. We also advocate approaches to overcoming limitations in the field. Examples include the use of contemporary mouse genetic reference population models such as the Collaborative Cross and Diversity Outbred mice that effectively model human genetic diversity and the use of translational models that integrate -omics and computational approaches from pre-clinical to clinical models of obesity. Finally, we suggest best scientific practices to ensure strong rigor that will allow investigators to delineate the sources of heterogeneity in the population with obesity. Collectively, we propose that it is critical to think of obesity as a heterogeneous disease with complex mechanisms and etiologies, requiring unique prevention and treatment strategies tailored to the individual.
Collapse
Affiliation(s)
| | - John E French
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| | - Naima Moustaid-Moussa
- Obesity Research Institute and Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Venkata S Voruganti
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher A Bizon
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, NC, USA
| | - Zhiyong Cheng
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Delisha A Stewart
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| | - John W Easterbrook
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
84
|
Rana S, Bhatti AA. Predicting anthropometric and metabolic traits with a genetic risk score for obesity in a sample of Pakistanis. Sci Rep 2021; 11:8320. [PMID: 33859285 PMCID: PMC8050295 DOI: 10.1038/s41598-021-87702-0] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity is an outcome of multiple factors including environmental and genetic influences. Common obesity is a polygenic trait indicating that multiple genetic variants act synergistically to influence its expression. We constructed a genetic risk score (GRS) based on five genetic variants (MC4R rs17782313, BDNF rs6265, FTO rs1421085, TMEM18 rs7561317, and NEGR1 rs2815752) and examined its association with obesity-related traits in a sample of Pakistanis. The study involved 306 overweight/obese (OW/OB) and 300 normal-weight (NW) individuals. The age range of the study participants was 12-63 years. All anthropometric and metabolic parameters were measured for each participant via standard procedures and biochemical assays, respectively. The genetic variants were genotyped by allelic discrimination assays. The age- and gender-adjusted associations between the GRS and obesity-related anthropometric and metabolic measures were determined using linear regression analyses. The results showed that OW/OB individuals had significantly higher mean ranks of GRS than NW individuals. Moreover, a significant association of the GRS with obesity-related anthropometric traits was seen. However, the GRS did not appear to affect any obesity-related metabolic parameter. In conclusion, our findings indicate the combined effect of multiple genetic variants on the obesity-related anthropometric phenotypes in Pakistanis.
Collapse
Affiliation(s)
- Sobia Rana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Adil Anwar Bhatti
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
85
|
Association of metabolites with obesity based on two gene variants, MC4R rs17782313 and BDNF rs6265. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166144. [PMID: 33862146 DOI: 10.1016/j.bbadis.2021.166144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Previous genome-wide association analyses for obesity related genes demonstrated the association of BDNF gene variant rs6265 and MC4R gene variant rs17782313 with body mass index (BMI). However, the associated metabolite pathways are still behind the curtain. The aim of the current study is to investigate the associations of metabolic changes in obesity with MC4R gene variant rs17782313 and BDNF variant rs6265. Gas chromatography-mass spectrometry based untargeted metabolomics approach was used and 42 identified serum metabolites were selected for statistical analyses. Significant association of seven metabolites with MC4R gene variant rs17782313 based on obesity and thirty metabolites with obesity dependent BDNF variant rs6265 using additive model (adjusted p < 0.05) was observed. This study highlights the importance of alteration of fatty acid biosynthesis, probably due to high consumption of fats may cause to develop obesity. But obesity is a complex disorder and the full clarification of this complex machinery is still distant. To understand the obesity in a better way, more studies are required to identify remaining metabolites and also mechanism of these metabolic entities.
Collapse
|
86
|
Miller RG, McGurnaghan SJ, Onengut-Gumuscu S, Chen WM, Colhoun HM, Rich SS, Orchard TJ, Costacou T. Insulin resistance-associated genetic variants in type 1 diabetes. J Diabetes Complications 2021; 35:107842. [PMID: 33468396 PMCID: PMC7936951 DOI: 10.1016/j.jdiacomp.2020.107842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 12/22/2022]
Abstract
AIMS To examine candidate insulin resistance single nucleotide polymorphisms (SNPs) for associations with glycemic control, insulin resistance, BMI, and complications in an observational type 1 diabetes (T1D) cohort: the Pittsburgh Epidemiology of Diabetes Complications (EDC) study. METHODS In 422 European-ancestry participants, we assessed associations using additive models between 15 candidate SNPs and 25-year mortality, cardiovascular disease, microalbuminuria, overt nephropathy and proliferative retinopathy, and 25-year mean HbA1c, estimated glucose disposal rate (eGDR, inverse measure of insulin resistance), and BMI. RESULTS The A allele of rs12970134 was associated with higher mean HbA1c (β = +0.34 ± 0.09, p = 0.00009) and nominally associated with worse eGDR (p = 0.02). Further analyses suggest the HbA1c association may be modified by diabetes therapy regimen: rs12970134 AA genotype was associated with higher HbA1c under non-intensive therapy conditions (<3 insulin injections/day or monitoring blood glucose<3 times/day [p = 0.004]), but not under intensive therapy (≥3 injections/day or insulin pump and monitoring glucose≥3 times/day [p = 0.71]). There were no significant associations between any SNPs and BMI or complications. CONCLUSIONS rs12970134, near MC4R, is strongly associated with HbA1c in this cohort. Further exploration of this genomic region is warranted, as it may hold promise for discovering new therapeutic targets to improve glycemic control in T1D.
Collapse
Affiliation(s)
- Rachel G Miller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 4200 Fifth Avenue, Pittsburgh, PA 15260, USA.
| | - Stuart J McGurnaghan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, Scotland, United Kingdom of Great Britain and Northern Ireland
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA 22903, USA
| | - Wei-Min Chen
- Center for Public Health Genomics, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA 22903, USA
| | - Helen M Colhoun
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, Scotland, United Kingdom of Great Britain and Northern Ireland
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA 22903, USA
| | - Trevor J Orchard
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 4200 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Tina Costacou
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 4200 Fifth Avenue, Pittsburgh, PA 15260, USA
| |
Collapse
|
87
|
Rebelos E, Honka MJ, Ekblad L, Bucci M, Hannukainen JC, Fernandes Silva L, Virtanen KA, Nummenmaa L, Nuutila P. The Obesity Risk SNP (rs17782313) near the MC4R Gene Is Not Associated with Brain Glucose Uptake during Insulin Clamp-A Study in Finns. J Clin Med 2021; 10:jcm10061312. [PMID: 33806715 PMCID: PMC8004974 DOI: 10.3390/jcm10061312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 12/03/2022] Open
Abstract
The melanocortin system is involved in the control of adiposity through modulation of food intake and energy expenditure. The single nucleotide polymorphism (SNP) rs17782313 near the MC4R gene has been linked to obesity, and a previous study using magnetoencephalography has shown that carriers of the mutant allele have decreased cerebrocortical response to insulin. Thus, in this study, we addressed whether rs17782313 associates with brain glucose uptake (BGU). Here, [18F]-fluorodeoxyglucose positron emission tomography (PET) data from 113 Finnish subjects scanned under insulin clamp conditions who also had the rs17782313 determined were compiled from a single-center cohort. BGU was quantified by the fractional uptake rate. Statistical analysis was performed with statistical parametric mapping. There was no difference in age, BMI, and insulin sensitivity as indexed by the M value between the rs17782313-C allele carriers and non-carriers. Brain glucose uptake during insulin clamp was not different by gene allele, and it correlated with the M value, in both the rs17782313-C allele carriers and non-carriers. The obesity risk SNP rs17782313 near the MC4R gene is not associated with brain glucose uptake during insulin clamp in humans, and this frequent mutation cannot explain the enhanced brain glucose metabolic rates in insulin resistance.
Collapse
Affiliation(s)
- Eleni Rebelos
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
- Correspondence: (E.R.); (P.N.); Tel.: +39-3488454140 (E.R.); +358-2313-1868 (P.N.)
| | | | - Laura Ekblad
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
| | - Marco Bucci
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Jarna C. Hannukainen
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Kirsi A. Virtanen
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
| | - Lauri Nummenmaa
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
- Department of Psychology, University of Turku, 20520 Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, 20520 Turku, Finland; (L.E.); (M.B.); (J.C.H.); (K.A.V.); (L.N.)
- Department of Endocrinology, Turku University Hospital, 20521 Turku, Finland
- Correspondence: (E.R.); (P.N.); Tel.: +39-3488454140 (E.R.); +358-2313-1868 (P.N.)
| |
Collapse
|
88
|
Chen Y, Chen XY, Dong XL, Wang YZ, Wang N, Zhu JF, Chen Y, Jiang QW, Fu CW. Investigation of the Association between 45 Tag SNPs and Type 2 Diabetes Mellitus in Han Chinese Adults: A Prospective Cohort Study. Public Health Genomics 2021; 24:123-130. [PMID: 33706321 DOI: 10.1159/000513891] [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] [Received: 06/13/2020] [Accepted: 12/17/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The objective of this study was to examine the association between type 2 diabetes mellitus (T2DM) and genes identified in previous genome-wide association studies (GWASs) in rural Han Chinese adults. METHODS This prospective study included 1,832 adults aged ≥18 years in Deqing without diabetes at baseline. The subjects were followed up for 8.7 years on average. We selected 45 susceptible tag single-nucleotide polymorphisms (SNPs) for T2DM that have been identified in GWASs and genotyped. A Cox model was constructed to calculate the adjusted hazard ratios (aHRs) for the association between SNPs and incident T2DM. RESULTS The incidence rate of T2DM was 12.0 per 1,000 person-years. After adjustment for covariates and a Bonferroni correction, rs17584499 of protein tyrosine phosphatase, receptor-type D (PTPRD), rs11257655 and rs10906115 of cell division cycle 123 gene (CDC123), and rs12970134 of melanocortin-4 receptor (MC4R) were significantly associated with incident T2DM. The aHRs for incident T2DM were 1.75 (95% confidence interval [CI]: 1.28-2.40) and 1.61 (95% CI: 1.27-2.04) in association with an increasing number of T alleles in rs17584499 and rs11257655 under an additive genetic model, and the aHR was 1.72 (95% CI: 1.33-2.22) with an increasing number of A alleles in rs10906115. The aHRs under the dominant model were 1.82 (95% CI: 1.25-2.66) for TT + CT versus CC of rs17584499 and 2.04 (95% CI: 1.47-2.86) for AA + AG versus GG of rs10966115. The aHRs under the recessive model were 2.99 (95% CI: 1.30-6.89) for TT versus CT + CC of rs17584499, 1.92 (95% CI: 1.39-2.70) for TT versus CT + CC of rs11257655, and 2.54 (95% CI:1.22-5.29) for AA versus AG + GG of rs12970134. In addition, an increased incidence of T2DM was significantly associated with the TA haplotype of rs11257655 and rs10906115 (aHR = 1.81, 95% CI: 1.12-2.92), while a decreased incidence was associated with the CG haplotype (aHR = 0.49, 95% CI: 0.35-0.68) and the CT haplotype of rs1111875 and rs5015480 (aHR = 0.61, 95% CI: 0.37-0.98). CONCLUSION Variants of the PTPRD, CDC123, and MC4R genes were associated with the T2DM incidence in a rural Han Chinese population.
Collapse
Affiliation(s)
- Yun Chen
- School of Public Health, Key Laboratory of Public Health Safety, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Xiao-Ying Chen
- School of Public Health, Key Laboratory of Public Health Safety, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Xiao-Lian Dong
- Deqing County Center for Disease Prevention and Control, Deqing County, China
| | - Yu-Zhuo Wang
- School of Public Health, Key Laboratory of Public Health Safety, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Na Wang
- School of Public Health, Key Laboratory of Public Health Safety, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Jian-Fu Zhu
- Deqing County Center for Disease Prevention and Control, Deqing County, China
| | - Yue Chen
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Qing-Wu Jiang
- School of Public Health, Key Laboratory of Public Health Safety, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Chao-Wei Fu
- School of Public Health, Key Laboratory of Public Health Safety, NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China,
| |
Collapse
|
89
|
Iwase M, Matsuo K, Nakatochi M, Oze I, Ito H, Koyanagi Y, Ugai T, Kasugai Y, Hishida A, Takeuchi K, Okada R, Kubo Y, Shimanoe C, Tanaka K, Ikezaki H, Murata M, Takezaki T, Nishimoto D, Kuriyama N, Ozaki E, Suzuki S, Watanabe M, Mikami H, Nakamura Y, Uemura H, Katsuura-Kamano S, Kuriki K, Kita Y, Takashima N, Nagino M, Momozawa Y, Kubo M, Wakai K. Differential Effect of Polymorphisms on Body Mass Index Across the Life Course of Japanese: The Japan Multi-Institutional Collaborative Cohort Study. J Epidemiol 2021; 31:172-179. [PMID: 32147644 PMCID: PMC7878711 DOI: 10.2188/jea.je20190296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/24/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Obesity is a reported risk factor for various health problems. Genome-wide association studies (GWASs) have identified numerous independent loci associated with body mass index (BMI). However, most of these have been focused on Europeans, and little evidence is available on the genetic effects across the life course of other ethnicities. METHODS We conducted a cross-sectional study to examine the associations of 282 GWAS-identified single nucleotide polymorphisms with three BMI-related traits, current BMI, BMI at 20 years old (BMI at 20), and change in BMI (BMI change), among 11,586 Japanese individuals enrolled in the Japan Multi-Institutional Collaborative Cohort study. Associations were examined using multivariable linear regression models. RESULTS We found a significant association (P < 0.05/282 = 1.77 × 10-4) between BMI and 11 polymorphisms in or near FTO, BDNF, TMEM18, HS6ST3, and BORCS7. The trend was similar between current BMI and BMI change, but differed from that of the BMI at 20. Among the significant variants, those on FTO were associated with all BMI traits, whereas those on TMEM18 and HS6SR3 were only associated with BMI at 20. The association of FTO loci with BMI remained, even after additional adjustment for dietary energy intake. CONCLUSIONS Previously reported BMI-associated loci discovered in Europeans were also identified in the Japanese population. Additionally, our results suggest that the effects of each loci on BMI may vary across the life course and that this variation may be caused by the differential effects of individual genes on BMI via different pathways.
Collapse
Affiliation(s)
- Madoka Iwase
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Surgical Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Nakatochi
- Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hidemi Ito
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuriko Koyanagi
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Tomotaka Ugai
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yumiko Kasugai
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Takeuchi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Kubo
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Keitaro Tanaka
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiroaki Ikezaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Masayuki Murata
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Toshiro Takezaki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Daisaku Nishimoto
- School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Nagato Kuriyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Etsuko Ozaki
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Miki Watanabe
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Haruo Mikami
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yohko Nakamura
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Hirokazu Uemura
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Sakurako Katsuura-Kamano
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, University of Shizuoka, Shizuoka, Japan
| | - Yoshikuni Kita
- Faculty of Nursing Science, Tsuruga Nursing University, Fukui, Japan
| | - Naoyuki Takashima
- Department of Public Health, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Masato Nagino
- Department of Surgical Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kenji Wakai
- Division of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
90
|
Blauw LL, Noordam R, van der Laan SW, Trompet S, Kooijman S, van Heemst D, Jukema JW, van Setten J, de Borst GJ, Tybjærg-Hansen A, Pasterkamp G, Berbée JFP, Rensen PCN. Common Genetic Variation in MC4R Does Not Affect Atherosclerotic Plaque Phenotypes and Cardiovascular Disease Outcomes. J Clin Med 2021; 10:jcm10050932. [PMID: 33804309 PMCID: PMC7957774 DOI: 10.3390/jcm10050932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/11/2021] [Accepted: 02/23/2021] [Indexed: 12/01/2022] Open
Abstract
We analyzed the effects of the common BMI-increasing melanocortin 4 receptor (MC4R) rs17782313-C allele with a minor allele frequency of 0.22–0.25 on (1) cardiovascular disease outcomes in two large population-based cohorts (Copenhagen City Heart Study and Copenhagen General Population Study, n = 106,018; and UK Biobank, n = 357,426) and additionally in an elderly population at risk for cardiovascular disease (n = 5241), and on (2) atherosclerotic plaque phenotypes in samples of patients who underwent endarterectomy (n = 1439). Using regression models, we additionally analyzed whether potential associations were modified by sex or explained by changes in body mass index. We confirmed the BMI-increasing effects of +0.22 kg/m2 per additional copy of the C allele (p < 0.001). However, we found no evidence for an association of common MC4R genetic variation with coronary artery disease (HR 1.03; 95% CI 0.99, 1.07), ischemic vascular disease (HR 1.00; 95% CI 0.98, 1.03), myocardial infarction (HR 1.01; 95% CI 0.94, 1.08 and 1.02; 0.98, 1.07) or stroke (HR 0.93; 95% CI 0.85, 1.01), nor with any atherosclerotic plaque phenotype. Thus, common MC4R genetic variation, despite increasing BMI, does not affect cardiovascular disease risk in the general population or in populations at risk for cardiovascular disease.
Collapse
Affiliation(s)
- Lisanne L. Blauw
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Raymond Noordam
- Department Medicine, Division Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.T.); (D.v.H.)
- Correspondence: ; Tel.: +31-71-52-66640
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (S.W.v.d.L.); (G.P.)
| | - Stella Trompet
- Department Medicine, Division Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.T.); (D.v.H.)
| | - Sander Kooijman
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Diana van Heemst
- Department Medicine, Division Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.T.); (D.v.H.)
| | - Johan Wouter Jukema
- Department Cardiology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
| | - Jessica van Setten
- Surgery Specialties, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Gert J. de Borst
- Department Cardiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Anne Tybjærg-Hansen
- Department Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark;
- The Copenhagen City Heart Study, Frederiksberg Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg, Denmark
- The Copenhagen General Population Study and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
- Copenhagen University Hospitals and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (S.W.v.d.L.); (G.P.)
| | - Jimmy F. P. Berbée
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
91
|
Feng GJ, Wei XT, Zhang H, Yang XL, Shen H, Tian Q, Deng HW, Zhang L, Pei YF. Identification of pleiotropic loci underlying hip bone mineral density and trunk lean mass. J Hum Genet 2021; 66:251-260. [PMID: 32929176 PMCID: PMC7880826 DOI: 10.1038/s10038-020-00835-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 11/09/2022]
Abstract
Bone mineral density (BMD) and lean body mass (LBM) not only have a considerable heritability each, but also are genetically correlated. However, common genetic determinants shared by both traits are largely unknown. In the present study, we performed a bivariate genome-wide association study (GWAS) meta-analysis of hip BMD and trunk lean mass (TLM) in 11,335 subjects from 6 samples, and performed replication in estimated heel BMD and TLM in 215,234 UK Biobank (UKB) participants. We identified 2 loci that nearly attained the genome-wide significance (GWS, p < 5.0 × 10-8) level in the discovery GWAS meta-analysis and that were successfully replicated in the UKB sample: 11p15.2 (lead SNP rs12800228, discovery p = 2.88 × 10-7, replication p = 1.95 × 10-4) and 18q21.32 (rs489693, discovery p = 1.67 × 10-7, replication p = 1.17 × 10-3). The above 2 pleiotropic loci may play a pleiotropic role for hip BMD and TLM development. So our findings provide useful insights that further enhance our understanding of genetic interplay between BMD and LBM.
Collapse
Affiliation(s)
- Gui-Juan Feng
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Xin-Tong Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Hong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Xiao-Lin Yang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Hui Shen
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Qing Tian
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hong-Wen Deng
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China.
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China.
| | - Yu-Fang Pei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China.
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China.
| |
Collapse
|
92
|
Yam P, Albright J, VerHague M, Gertz ER, Pardo-Manuel de Villena F, Bennett BJ. Genetic Background Shapes Phenotypic Response to Diet for Adiposity in the Collaborative Cross. Front Genet 2021; 11:615012. [PMID: 33643372 PMCID: PMC7905354 DOI: 10.3389/fgene.2020.615012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Defined as chronic excessive accumulation of adiposity, obesity results from long-term imbalance between energy intake and expenditure. The mechanisms behind how caloric imbalance occurs are complex and influenced by numerous biological and environmental factors, especially genetics, and diet. Population-based diet recommendations have had limited success partly due to the wide variation in physiological responses across individuals when they consume the same diet. Thus, it is necessary to broaden our understanding of how individual genetics and diet interact relative to the development of obesity for improving weight loss treatment. To determine how consumption of diets with different macronutrient composition alter adiposity and other obesity-related traits in a genetically diverse population, we analyzed body composition, metabolic rate, clinical blood chemistries, and circulating metabolites in 22 strains of mice from the Collaborative Cross (CC), a highly diverse recombinant inbred mouse population, before and after 8 weeks of feeding either a high protein or high fat high sucrose diet. At both baseline and post-diet, adiposity and other obesity-related traits exhibited a broad range of phenotypic variation based on CC strain; diet-induced changes in adiposity and other traits also depended largely on CC strain. In addition to estimating heritability at baseline, we also quantified the effect size of diet for each trait, which varied by trait and experimental diet. Our findings identified CC strains prone to developing obesity, demonstrate the genotypic and phenotypic diversity of the CC for studying complex traits, and highlight the importance of accounting for genetic differences when making dietary recommendations.
Collapse
Affiliation(s)
- Phoebe Yam
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, Davis, CA, United States
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, CA, United States
| | - Jody Albright
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Melissa VerHague
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Erik R. Gertz
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, CA, United States
| | | | - Brian J. Bennett
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, Davis, CA, United States
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, CA, United States
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
93
|
Namjou B, Stanaway IB, Lingren T, Mentch FD, Benoit B, Dikilitas O, Niu X, Shang N, Shoemaker AH, Carey DJ, Mirshahi T, Singh R, Nestor JG, Hakonarson H, Denny JC, Crosslin DR, Jarvik GP, Kullo IJ, Williams MS, Harley JB. Evaluation of the MC4R gene across eMERGE network identifies many unreported obesity-associated variants. Int J Obes (Lond) 2021; 45:155-169. [PMID: 32952152 PMCID: PMC7752751 DOI: 10.1038/s41366-020-00675-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/07/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND/OBJECTIVES Melanocortin-4 receptor (MC4R) plays an essential role in food intake and energy homeostasis. More than 170 MC4R variants have been described over the past two decades, with conflicting reports regarding the prevalence and phenotypic effects of these variants in diverse cohorts. To determine the frequency of MC4R variants in large cohort of different ancestries, we evaluated the MC4R coding region for 20,537 eMERGE participants with sequencing data plus additional 77,454 independent individuals with genome-wide genotyping data at this locus. SUBJECTS/METHODS The sequencing data were obtained from the eMERGE phase III study, in which multisample variant call format calls have been generated, curated, and annotated. In addition to penetrance estimation using body mass index (BMI) as a binary outcome, GWAS and PheWAS were performed using median BMI in linear regression analyses. All results were adjusted for principal components, age, sex, and sites of genotyping. RESULTS Targeted sequencing data of MC4R revealed 125 coding variants in 1839 eMERGE participants including 30 unreported coding variants that were predicted to be functionally damaging. Highly penetrant unreported variants included (L325I, E308K, D298N, S270F, F261L, T248A, D111V, and Y80F) in which seven participants had obesity class III defined as BMI ≥ 40 kg/m2. In GWAS analysis, in addition to known risk haplotype upstream of MC4R (best variant rs6567160 (P = 5.36 × 10-25, Beta = 0.37), a novel rare haplotype was detected which was protective against obesity and encompassed the V103I variant with known gain-of-function properties (P = 6.23 × 10-08, Beta = -0.62). PheWAS analyses extended this protective effect of V103I to type 2 diabetes, diabetic nephropathy, and chronic renal failure independent of BMI. CONCLUSIONS MC4R screening in a large eMERGE cohort confirmed many previous findings, extend the MC4R pleotropic effects, and discovered additional MC4R rare alleles that probably contribute to obesity.
Collapse
Affiliation(s)
- Bahram Namjou
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA.
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Ian B Stanaway
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, WA, USA
| | - Todd Lingren
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Frank D Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Barbara Benoit
- Research Information Science and Computing, Partners HealthCare, Somerville, MA, USA
| | - Ozan Dikilitas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xinnan Niu
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, Nashville, TN, USA
| | - Ning Shang
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Ashley H Shoemaker
- Department of Pediatrics, Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David J Carey
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Tooraj Mirshahi
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | | | - Jordan G Nestor
- Department of Medicine, Division of Nephrology, Columbia University, New York, NY, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, Nashville, TN, USA
| | - David R Crosslin
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, WA, USA
| | - Gail P Jarvik
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, USA
- Department Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Marc S Williams
- Genomic Medicine Institute (M.S.W.), Geisinger, Danville, PA, USA
| | - John B Harley
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- U.S. Department of Veterans Affairs Medical Center, Cincinnati, OH, USA
| |
Collapse
|
94
|
Soret PA, Magusto J, Housset C, Gautheron J. In Vitro and In Vivo Models of Non-Alcoholic Fatty Liver Disease: A Critical Appraisal. J Clin Med 2020; 10:jcm10010036. [PMID: 33374435 PMCID: PMC7794936 DOI: 10.3390/jcm10010036] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), including non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH), represents the hepatic manifestation of obesity and metabolic syndrome. Due to the spread of the obesity epidemic, NAFLD is becoming the most common chronic liver disease and one of the principal indications for liver transplantation. However, no pharmacological treatment is currently approved to prevent the outbreak of NASH, which leads to fibrosis and cirrhosis. Preclinical research is required to improve our knowledge of NAFLD physiopathology and to identify new therapeutic targets. In the present review, we summarize advances in NAFLD preclinical models from cellular models, including new bioengineered platforms, to in vivo models, with a particular focus on genetic and dietary mouse models. We aim to discuss the advantages and limits of these different models.
Collapse
Affiliation(s)
- Pierre-Antoine Soret
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hepatology Department, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, 75012 Paris, France
| | - Julie Magusto
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
| | - Chantal Housset
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hepatology Department, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, 75012 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
| | - Jérémie Gautheron
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
- Correspondence:
| |
Collapse
|
95
|
Lee WJ, Lim JE, Jung HU, Kang JO, Park T, Won S, Rhee SY, Kim MK, Kim YJ, Oh B. Analysis of the Interaction between Polygenic Risk Score and Calorie Intake in Obesity in the Korean Population. Lifestyle Genom 2020; 14:20-29. [PMID: 33302275 DOI: 10.1159/000511333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/31/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Obesity results from an imbalance in the intake and expenditure of calories that leads to lifestyle-related diseases. Although genome-wide association studies (GWAS) have revealed many obesity-related genetic factors, the interactions of these factors and calorie intake remain unknown. This study aimed to investigate interactions between calorie intake and the polygenic risk score (PRS) of BMI. METHODS Three cohorts, i.e., from the Korea Association REsource (KARE; n = 8,736), CArdioVAscular Disease Association Study (CAVAS; n = 9,334), and Health EXAminee (HEXA; n = 28,445), were used for this study. BMI-related genetic loci were selected from previous GWAS. Two scores, PRS, and association (a)PRS, were used; the former was determined from 193 single-nucleotide polymorphisms (SNPs) from 5 GWAS datasets, and the latter from 62 SNPs (potentially associated) from 3 Korean cohorts (meta-analysis, p < 0.01). RESULTS PRS and aPRS were significantly associated with BMI in all 3 cohorts but did not exhibit a significant interaction with total calorie intake. Similar results were obtained for obesity. PRS and aPRS were significantly associated with obesity but did not show a significant interaction with total calorie intake. We further analyzed the interaction with protein, fat, and carbohydrate intake. The results were similar to those for total calorie intake, with PRS and aPRS found to not be associated with the interaction of any of the 3 nutrition components for either BMI or obesity. DISCUSSION The interaction of BMI PRS with calorie intake was investigated in 3 independent Korean cohorts (total n = 35,094) and no interactions were found between PRS and calorie intake for obesity.
Collapse
Affiliation(s)
- Won-Jun Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hae Un Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ji-One Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea.,Department of Statistics, Seoul National University, Seoul, Republic of Korea
| | - Sungho Won
- Department of Public Health Science, Seoul National University, Seoul, Republic of Korea
| | - Sang Youl Rhee
- Department of Endocrinology and Metabolism, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Mi Kyung Kim
- Department of Preventive Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea.,Institute for Health and Society, Hanyang University, Seoul, Republic of Korea
| | - Yeon-Jung Kim
- Division of Biobank for Health Science, Center for Genome Science, Korea National Institute of Health, Chungcheongbuk-do, Republic of Korea
| | - Bermseok Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea,
| |
Collapse
|
96
|
Yarizadeh H, Bahiraee A, Asadi S, Maddahi NS, Setayesh L, Casazza K, Mirzaei K. The interaction between dietary approaches to stop hypertension and MC4R gene variant in predicting cardiovascular risk factors. INT J VITAM NUTR RES 2020; 92:376-384. [PMID: 33284034 DOI: 10.1024/0300-9831/a000690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Objective: The genetic variants near the melanocortin-4 receptor gene (MC4R), a key protein regulating energy balance and adiposity, have been related to obesity and cardiovascular risk factors. However, qualitative and quantitative aspects of diet may modulate the association of this polymorphism with obesity and cardiovascular diseases (CVDs). The aim of this study was to evaluate interactions among MC4R rs17782313, the Dietary Approaches to Stop Hypertension (DASH) diet and risk factors for CVDs. Method: This cross-sectional study was conducted on 266 Iranian women categorized by body mass index (BMI) range of 25-40 kg/m2 as overweight or obese. CVD risk factors included waist circumference (WC), lipid profile, blood pressure, insulin circulation and fasting blood sugar (FBS). Insulin and FBS were used to calculate homeostatic model assessment insulin resistance (HOMA-IR) Body composition was assessed by a multi-frequency bioelectrical impedance analyzer, InBody 770 scanner. Results: The findings of this study show that high adherence to the DASH diet in the CC groups were associated with decreased SBP and DBP compared to the TT group. In addition, a significant difference between women with high adherence to the DASH diet compared to low adherence was observed for body weight (p < 0.001), fat free mass (FFM) (p = 0.01) and BMI (p = 0.02). Women with the CC genotype had higher insulin (mg/dl) (mean and SD, for TT: 14.6 ± 4.6, TC: 17.3 ± 9.2, CC: 15.3 ± 4.8, p = 0.04) and HOMA-IR (mean for and SD, TT: 3.1 ± 1.07, TC: 3.9 ± 2.4, CC: 3.2 ± 1.1, p = 0.01) than TT group. Inclusion of potential confounding variables (age, physical activity, BMI and daily caloric intake) did not attenuate the difference. Conclusion: Among overweight/obese Iranian women with the CC genotype, incorporating the DASH diet may serve as a dietary prescription to decrease CVD risk. A dietary intervention trial is warranted.
Collapse
Affiliation(s)
- Habib Yarizadeh
- Students' Scientific Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sara Asadi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Niloofar Sadat Maddahi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Leila Setayesh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Krista Casazza
- Marieb College of Health and Human Services, Florida Gulf Coast University, Fort Myers, Florida, USA
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
97
|
Rojo D, McCarthy C, Raingo J, Rubinstein M. Mouse models for V103I and I251L gain of function variants of the human MC4R display decreased adiposity but are not protected against a hypercaloric diet. Mol Metab 2020; 42:101077. [PMID: 32916307 PMCID: PMC7559519 DOI: 10.1016/j.molmet.2020.101077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 09/06/2020] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor that plays major roles in the central control of energy balance. Loss-of-function mutations of MC4R constitute the most common monogenic cause of early-onset extreme obesity in humans, whereas gain-of-function mutations appear to be protective. In particular, two relatively frequent alleles carrying the non-synonymous coding mutations V103I or I251L are associated with lower risks of obesity and type-2 diabetes. Although V103I and I251L MC4Rs showed more efficient signalling in transfected cells, their specific effects in live animals remain unexplored. Here, we investigated whether the introduction of V103I and I251L mutations into the mouse MC4R leads to a lean phenotype and provides protection against an obesogenic diet. METHODS Using CRISPR/Cas9, we generated two novel strains of mice carrying single-nucleotide mutations into the mouse Mc4r which are identical to those present in V103I and I251L MCR4 human alleles, and studied their phenotypic outcomes in mice fed with normal chow or a high-fat diet. In particular, we measured body weight progression, food intake and adiposity. In addition, we analysed glucose homeostasis through glucose and insulin tolerance tests. RESULTS We found that homozygous V103I females displayed shorter longitudinal length and decreased abdominal white fat, whereas homozygous I251L females were also shorter and leaner due to decreased weight in all white fat pads examined. Homozygous Mc4rV103I/V103I and Mc4rI251L/I251L mice of both sexes showed improved glucose homeostasis when challenged in a glucose tolerance test, whereas Mc4rI251L/I251L females showed improved responses to insulin. Despite being leaner and metabolically more efficient, V103I and I251L mutants fed with a hypercaloric diet increased their fasting glucose levels and adiposity similar to their wild-type littermates. CONCLUSIONS Our results demonstrate that mice carrying V103I and I251L MC4R mutations displayed gain-of-function phenotypes that were more evident in females. However, hypermorphic MC4R mutants were as susceptible as their control littermates to the obesogenic and diabetogenic effects elicited by a long-term hypercaloric diet, highlighting the importance of healthy feeding habits even under favourable genetic conditions.
Collapse
Affiliation(s)
- Daniela Rojo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Buenos Aires, Argentina
| | - Clara McCarthy
- Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de La Plata and Comisión de Investigaciones de la Provincia de Buenos Aires, La Plata, Province of Buenos Aires, Argentina
| | - Jesica Raingo
- Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de La Plata and Comisión de Investigaciones de la Provincia de Buenos Aires, La Plata, Province of Buenos Aires, Argentina
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Buenos Aires, Argentina; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, 1428 Buenos Aires, Argentina; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
98
|
Wallis N, Raffan E. The Genetic Basis of Obesity and Related Metabolic Diseases in Humans and Companion Animals. Genes (Basel) 2020; 11:E1378. [PMID: 33233816 PMCID: PMC7699880 DOI: 10.3390/genes11111378] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is one of the most prevalent health conditions in humans and companion animals globally. It is associated with premature mortality, metabolic dysfunction, and multiple health conditions across species. Obesity is, therefore, of importance in the fields of medicine and veterinary medicine. The regulation of adiposity is a homeostatic process vulnerable to disruption by a multitude of genetic and environmental factors. It is well established that the heritability of obesity is high in humans and laboratory animals, with ample evidence that the same is true in companion animals. In this review, we provide an overview of how genes link to obesity in humans, drawing on a wealth of information from laboratory animal models, and summarise the mechanisms by which obesity causes related disease. Throughout, we focus on how large-scale human studies and niche investigations of rare mutations in severely affected patients have improved our understanding of obesity biology and can inform our ability to interpret results of animal studies. For dogs, cats, and horses, we compare the similarities in obesity pathophysiology to humans and review the genetic studies that have been previously reported in those species. Finally, we discuss how veterinary genetics may learn from humans about studying precise, nuanced phenotypes and implementing large-scale studies, but also how veterinary studies may be able to look past clinical findings to mechanistic ones and demonstrate translational benefits to human research.
Collapse
Affiliation(s)
- Natalie Wallis
- Anatomy Building, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Eleanor Raffan
- Anatomy Building, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
99
|
Micioni Di Bonaventura E, Botticelli L, Tomassoni D, Tayebati SK, Micioni Di Bonaventura MV, Cifani C. The Melanocortin System behind the Dysfunctional Eating Behaviors. Nutrients 2020; 12:E3502. [PMID: 33202557 PMCID: PMC7696960 DOI: 10.3390/nu12113502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| |
Collapse
|
100
|
Naureen Z, Miggiano GAD, Aquilanti B, Velluti V, Matera G, Gagliardi L, Zulian A, Romanelli R, Bertelli M. Genetic test for the prescription of diets in support of physical activity. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:e2020011. [PMID: 33170161 PMCID: PMC8023120 DOI: 10.23750/abm.v91i13-s.10584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/17/2020] [Indexed: 01/03/2023]
Abstract
Owing to the fields of nutrigenetics and nutrigenomics today we can think of devising approaches to optimize health, delay onset of diseases and reduce its severity according to our genetic blue print. However this requires a deep understanding of nutritional impact on expression of genes that may result in a specific phenotype. The extensive research and observational studies during last two decades reporting interactions between genes, diet and physical activity suggest a cross talk between various genetic and environmental factors and lifestyle interventions. Although considerable efforts have been made in unraveling the mechanisms of gene-diet interactions the scientific evidences behind developing commercial genetic tests for providing personalized nutrition recommendations are still scarce. In this scenario the current mini-review aims to provide useful insights into salient feature of nutrition based genetic research and its commercial application and the ethical issue and concerns related to its outcome.
Collapse
Affiliation(s)
- Zakira Naureen
- Department of Biological Sciences and Chemistry, College of Arts and Sciences, University of Nizwa, Nizwa, Oman.
| | | | - Barbara Aquilanti
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Valeria Velluti
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Giuseppina Matera
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Lucilla Gagliardi
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | | | | | - Matteo Bertelli
- MAGI'S LAB, Rovereto (TN), Italy; MAGI EUREGIO, Bolzano, Italy; EBTNA-LAB, Rovereto (TN), Italy.
| |
Collapse
|