201
|
D'Silva S, Chakraborty S, Kahali B. Concurrent outcomes from multiple approaches of epistasis analysis for human body mass index associated loci provide insights into obesity biology. Sci Rep 2022; 12:7306. [PMID: 35508500 PMCID: PMC9068779 DOI: 10.1038/s41598-022-11270-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/18/2022] [Indexed: 12/13/2022] Open
Abstract
Genome wide association studies (GWAS) have focused on elucidating the genetic architecture of complex traits by assessing single variant effects in additive genetic models, albeit explaining a fraction of the trait heritability. Epistasis has recently emerged as one of the intrinsic mechanisms that could explain part of this missing heritability. We conducted epistasis analysis for genome-wide body mass index (BMI) associated SNPs in Alzheimer's Disease Neuroimaging Initiative (ADNI) and followed up top significant interacting SNPs for replication in the UK Biobank imputed genotype dataset. We report two pairwise epistatic interactions, between rs2177596 (RHBDD1) and rs17759796 (MAPK1), rs1121980 (FTO) and rs6567160 (MC4R), obtained from a consensus of nine different epistatic approaches. Gene interaction maps and tissue expression profiles constructed for these interacting loci highlights co-expression, co-localisation, physical interaction, genetic interaction, and shared pathways emphasising the neuronal influence in obesity and implicating concerted expression of associated genes in liver, pancreas, and adipose tissues insinuating to metabolic abnormalities characterized by obesity. Detecting epistasis could thus be a promising approach to understand the effect of simultaneously interacting multiple genetic loci in disease aetiology, beyond single locus effects.
Collapse
Affiliation(s)
- Sheldon D'Silva
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Shreya Chakraborty
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
- Interdisciplinary Mathematical Sciences, Indian Institute of Science, Bangalore, 560012, India
| | - Bratati Kahali
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
202
|
Boutelle KN, Eichen DM, Peterson CB, Strong DR, Kang-Sim DJE, Rock CL, Marcus BH. Effect of a Novel Intervention Targeting Appetitive Traits on Body Mass Index Among Adults With Overweight or Obesity: A Randomized Clinical Trial. JAMA Netw Open 2022; 5:e2212354. [PMID: 35583870 PMCID: PMC9118075 DOI: 10.1001/jamanetworkopen.2022.12354] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/25/2022] [Indexed: 01/14/2023] Open
Abstract
Importance Behavioral weight loss (BWL) programs result in weight loss for some, but most individuals regain the weight. The behavioral susceptibility theory proposes that genetically determined appetitive traits, such as food responsiveness (FR) and satiety responsiveness (SR), interact with the environment and lead to overeating and weight gain; the regulation of cues (ROC) intervention was developed specifically to target FR and SR. Objective To evaluate the efficacy of ROC, ROC combined with BWL (ROC+), BWL, and an active comparator (AC) over 12 months of treatment and 12 months of follow-up. Design, Setting, and Participants This randomized clinical trial was conducted from December 2015 to December 2019 in a university clinic. A total of 1488 volunteers from the community inquired about the study; 1217 were excluded or declined to participate. Eligibility criteria included body mass index (BMI) of 25 to 45, age 18 to 65 years, and lack of comorbidities or other exclusionary criteria that would interfere with participation. Data were analyzed from September 2021 to January 2022. Interventions ROC uniquely targeted FR and SR. BWL included energy restriction, increasing physical activity, and behavior therapy techniques. ROC+ combined ROC with BWL. AC included mindfulness, social support, and nutrition education. Main Outcomes and Measures Change in body weight as measured by BMI. Results A total of 271 adults (mean [SD] age, 46.97 [11.80] years; 81.6% female [221 participants]; mean [SD] BMI, 34.59 [5.28]; 61.9% White [167 participants]) were assessed at baseline, midtreatment, posttreatment, and 6-month and 12-month follow-up. Sixty-six participants were randomized to AC, 69 to ROC, 67 to ROC+, and 69 to BWL. Results showed that ROC, ROC+, and BWL interventions resulted in significantly lower BMI at the end of treatment (BMI ROC, -1.18; 95% CI, -2.10 to -0.35; BMI ROC+, -1.56; 95% CI, -2.43 to -0.67; BMI BWL, -1.58; 95% CI, -2.45 to -0.71). Compared with BWL, BMI at the end of treatment was not significantly different from ROC or ROC+ (BMI ROC, 0.40; 95% CI, -0.55 to 1.36; BMI ROC+, 0.03; 95% CI, -0.88 to 0.93); however, the BMI of the AC group was substantially higher (BMI AC, 1.58; 95% CI, 0.72 to 2.45). BMI reductions at 24 months after randomization were similar for ROC, ROC+, and BWL. Importantly, FR was a moderator of treatment effects with more weight loss for participants who scored higher in FR in the ROC and ROC+ groups. Conclusions and Relevance These findings suggest that ROC and ROC+ provide alternative weight loss approaches for adults. These models could be particularly effective for individuals who struggle with FR and could be used as a precision approach for weight loss. Trial Registration ClinicalTrials.gov Identifier: NCT02516839.
Collapse
Affiliation(s)
- Kerri N. Boutelle
- Department of Pediatrics, University of California, San Diego, La Jolla
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla
- Department of Psychiatry, University of California, San Diego, La Jolla
| | - Dawn M. Eichen
- Department of Pediatrics, University of California, San Diego, La Jolla
| | - Carol B. Peterson
- Department of Psychiatry and Behavioral Health, University of Minnesota, Minneapolis
| | - David R. Strong
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla
| | | | - Cheryl L. Rock
- Department of Family Medicine, University of California, San Diego, La Jolla
| | - Bess H. Marcus
- Behavioral and Social Sciences, Brown University, Providence, Rhode Island
| |
Collapse
|
203
|
Ameroso D, Meng A, Chen S, Felsted J, Dulla CG, Rios M. Astrocytic BDNF signaling within the ventromedial hypothalamus regulates energy homeostasis. Nat Metab 2022; 4:627-643. [PMID: 35501599 PMCID: PMC9177635 DOI: 10.1038/s42255-022-00566-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for maintaining energy and glucose balance within the central nervous system. Because the study of its metabolic actions has been limited to effects in neuronal cells, its role in other cell types within the brain remains poorly understood. Here we show that astrocytic BDNF signaling within the ventromedial hypothalamus (VMH) modulates neuronal activity in response to changes in energy status. This occurs via the truncated TrkB.T1 receptor. Accordingly, either fasting or central BDNF depletion enhances astrocytic synaptic glutamate clearance, thereby decreasing neuronal activity in mice. Notably, selective depletion of TrkB.T1 in VMH astrocytes blunts the effects of energy status on excitatory transmission, as well as on responses to leptin, glucose and lipids. These effects are driven by increased astrocytic invasion of excitatory synapses, enhanced glutamate reuptake and decreased neuronal activity. We thus identify BDNF/TrkB.T1 signaling in VMH astrocytes as an essential mechanism that participates in energy and glucose homeostasis.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Stella Chen
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jennifer Felsted
- Graduate Program in Biochemical and Molecular Nutrition, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Chris G Dulla
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
204
|
Henning T, Kochlik B, Kusch P, Strauss M, Jurić V, Pignitter M, Marusch F, Grune T, Weber D. Pre-Operative Assessment of Micronutrients, Amino Acids, Phospholipids and Oxidative Stress in Bariatric Surgery Candidates. Antioxidants (Basel) 2022; 11:antiox11040774. [PMID: 35453460 PMCID: PMC9031169 DOI: 10.3390/antiox11040774] [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: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity has been linked to lower concentrations of fat-soluble micronutrients and higher concentrations of oxidative stress markers as well as an altered metabolism of branched chain amino acids and phospholipids. In the context of morbid obesity, the aim of this study was to investigate whether and to which extent plasma status of micronutrients, amino acids, phospholipids and oxidative stress differs between morbidly obese (n = 23) and non-obese patients (n = 13). In addition to plasma, malondialdehyde, retinol, cholesterol and triglycerides were assessed in visceral and subcutaneous adipose tissue in both groups. Plasma γ-tocopherol was significantly lower (p < 0.011) in the obese group while other fat-soluble micronutrients showed no statistically significant differences between both groups. Branched-chain amino acids (all p < 0.008) and lysine (p < 0.006) were significantly higher in morbidly obese patients compared to the control group. Malondialdehyde concentrations in both visceral (p < 0.016) and subcutaneous (p < 0.002) adipose tissue were significantly higher in the morbidly obese group while plasma markers of oxidative stress showed no significant differences between both groups. Significantly lower plasma concentrations of phosphatidylcholine, phosphatidylethanolamine, lyso-phosphatidylethanolamine (all p < 0.05) and their corresponding ether-linked analogs were observed, which were all reduced in obese participants compared to the control group. Pre-operative assessment of micronutrients in patients undergoing bariatric surgery is recommended for early identification of patients who might be at higher risk to develop a severe micronutrient deficiency post-surgery. Assessment of plasma BCAAs and phospholipids in obese patients might help to differentiate between metabolic healthy patients and those with metabolic disorders.
Collapse
Affiliation(s)
- Thorsten Henning
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (T.H.); (P.K.); (T.G.)
- Food4Future (F4F), c/o Leibniz Institute of Vegetable and Ornamental Crops (IGZ), 14979 Grossbeeren, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany;
| | - Bastian Kochlik
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany;
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, (DIfE), 14558 Nuthetal, Germany
| | - Paula Kusch
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (T.H.); (P.K.); (T.G.)
- Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Matthias Strauss
- Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (M.S.); (V.J.); (M.P.)
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, 1090 Vienna, Austria
| | - Viktorija Jurić
- Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (M.S.); (V.J.); (M.P.)
| | - Marc Pignitter
- Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (M.S.); (V.J.); (M.P.)
| | - Frank Marusch
- Department of General and Visceral Surgery, Klinikum Ernst von Bergmann, 14467 Potsdam, Germany;
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (T.H.); (P.K.); (T.G.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany;
- Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
- Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (M.S.); (V.J.); (M.P.)
- Deutsches Zentrum für Diabetesforschung (DZD), 85764 Neuherberg, Germany
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (T.H.); (P.K.); (T.G.)
- Food4Future (F4F), c/o Leibniz Institute of Vegetable and Ornamental Crops (IGZ), 14979 Grossbeeren, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany;
- Correspondence:
| |
Collapse
|
205
|
Catalán V, Avilés-Olmos I, Rodríguez A, Becerril S, Fernández-Formoso JA, Kiortsis D, Portincasa P, Gómez-Ambrosi J, Frühbeck G. Time to Consider the "Exposome Hypothesis" in the Development of the Obesity Pandemic. Nutrients 2022; 14:1597. [PMID: 35458158 PMCID: PMC9032727 DOI: 10.3390/nu14081597] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
The obesity epidemic shows no signs of abatement. Genetics and overnutrition together with a dramatic decline in physical activity are the alleged main causes for this pandemic. While they undoubtedly represent the main contributors to the obesity problem, they are not able to fully explain all cases and current trends. In this context, a body of knowledge related to exposure to as yet underappreciated obesogenic factors, which can be referred to as the "exposome", merits detailed analysis. Contrarily to the genome, the "exposome" is subject to a great dynamism and variability, which unfolds throughout the individual's lifetime. The development of precise ways of capturing the full exposure spectrum of a person is extraordinarily demanding. Data derived from epidemiological studies linking excess weight with elevated ambient temperatures, in utero, and intergenerational effects as well as epigenetics, microorganisms, microbiota, sleep curtailment, and endocrine disruptors, among others, suggests the possibility that they may work alone or synergistically as several alternative putative contributors to this global epidemic. This narrative review reports the available evidence on as yet underappreciated drivers of the obesity epidemic. Broadly based interventions are needed to better identify these drivers at the same time as stimulating reflection on the potential relevance of the "exposome" in the development and perpetuation of the obesity epidemic.
Collapse
Affiliation(s)
- Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Iciar Avilés-Olmos
- Department of Neurology, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | | | - Dimitrios Kiortsis
- Department of Nuclear Medicine, Medical School, University of Ioannina, 45110 Ioannina, Greece;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
206
|
Loh M, Zhang W, Ng HK, Schmid K, Lamri A, Tong L, Ahmad M, Lee JJ, Ng MCY, Petty LE, Spracklen CN, Takeuchi F, Islam MT, Jasmine F, Kasturiratne A, Kibriya M, Mohlke KL, Paré G, Prasad G, Shahriar M, Chee ML, de Silva HJ, Engert JC, Gerstein HC, Mani KR, Sabanayagam C, Vujkovic M, Wickremasinghe AR, Wong TY, Yajnik CS, Yusuf S, Ahsan H, Bharadwaj D, Anand SS, Below JE, Boehnke M, Bowden DW, Chandak GR, Cheng CY, Kato N, Mahajan A, Sim X, McCarthy MI, Morris AP, Kooner JS, Saleheen D, Chambers JC. Identification of genetic effects underlying type 2 diabetes in South Asian and European populations. Commun Biol 2022; 5:329. [PMID: 35393509 PMCID: PMC8991226 DOI: 10.1038/s42003-022-03248-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/08/2022] [Indexed: 02/08/2023] Open
Abstract
South Asians are at high risk of developing type 2 diabetes (T2D). We carried out a genome-wide association meta-analysis with South Asian T2D cases (n = 16,677) and controls (n = 33,856), followed by combined analyses with Europeans (neff = 231,420). We identify 21 novel genetic loci for significant association with T2D (P = 4.7 × 10-8 to 5.2 × 10-12), to the best of our knowledge at the point of analysis. The loci are enriched for regulatory features, including DNA methylation and gene expression in relevant tissues, and highlight CHMP4B, PDHB, LRIG1 and other genes linked to adiposity and glucose metabolism. A polygenic risk score based on South Asian-derived summary statistics shows ~4-fold higher risk for T2D between the top and bottom quartile. Our results provide further insights into the genetic mechanisms underlying T2D, and highlight the opportunities for discovery from joint analysis of data from across ancestral populations.
Collapse
Affiliation(s)
- Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Middlesex, UB1 3HW, UK
| | - Hong Kiat Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Katharina Schmid
- Institute of Computational Biology, Deutsches Forschungszentrum für Gesundheit und Umwelt, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Department of Informatics, Technical University of Munich, 85748, Garching bei München, Neuherberg, Germany
| | - Amel Lamri
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Lin Tong
- The University of Chicago, Biological Sciences Division, Public Health Sciences, 5841 South Maryland Avenue, MC2000, Chicago, IL, 60637, USA
| | - Meraj Ahmad
- Genomic Research on Complex diseases, CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Jung-Jin Lee
- Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Mayo Hospital, Lahore, Pakistan
| | - Maggie C Y Ng
- Center for Genomics and Personalized Medicine Research, Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, 37215, USA
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Lauren E Petty
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA, 01003, USA
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Md Tariqul Islam
- U Chicago Research Bangladesh, House#4, Road#2b, Sector#4, Uttara, Dhaka, 1230, Bangladesh
| | - Farzana Jasmine
- The University of Chicago, Biological Sciences Division, Public Health Sciences, 5841 South Maryland Avenue, MC2000, Chicago, IL, 60637, USA
| | - Anuradhani Kasturiratne
- Department of Public Health, Faculty of Medicine, University of Kelaniya, Kelaniya, Sri Lanka
| | - Muhammad Kibriya
- The University of Chicago, Biological Sciences Division, Public Health Sciences, 5841 South Maryland Avenue, MC2000, Chicago, IL, 60637, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Guillaume Paré
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Gauri Prasad
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi, 110020, India
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohammad Shahriar
- The University of Chicago, Biological Sciences Division, Public Health Sciences, 5841 South Maryland Avenue, MC2000, Chicago, IL, 60637, USA
| | - Miao Ling Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - H Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Kelaniya, Sri Lanka
| | - James C Engert
- Department of Medicine, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Hertzel C Gerstein
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - K Radha Mani
- Genomic Research on Complex diseases, CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | - Marijana Vujkovic
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Ananda R Wickremasinghe
- Department of Public Health, Faculty of Medicine, University of Kelaniya, Kelaniya, Sri Lanka
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Salim Yusuf
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Habibul Ahsan
- The University of Chicago, Biological Sciences Division, Public Health Sciences, 5841 South Maryland Avenue, MC2000, Chicago, IL, 60637, USA
| | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi, 110020, India
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sonia S Anand
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Donald W Bowden
- Department of Medicine, Mayo Hospital, Lahore, Pakistan
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, 37215, USA
| | - Giriraj R Chandak
- Genomic Research on Complex diseases, CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
- JSS Academy of Health Education of Research, Mysuru, India
- Science and Engineering Research Board, Department of Science and Technology, Ministry of Science and technology, Government of India, New Delhi, India
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hosptial, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, L69 3GL, UK
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Middlesex, UB1 3HW, UK.
- Imperial College Healthcare NHS Trust, Imperial College London, London, W12 0HS, UK.
- MRC-PHE Centre for Enviroment and Health, Imperial College London, London, W2 1PG, UK.
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK.
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, Pakistan.
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Cardiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - John C Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK.
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Middlesex, UB1 3HW, UK.
- Imperial College Healthcare NHS Trust, Imperial College London, London, W12 0HS, UK.
- MRC-PHE Centre for Enviroment and Health, Imperial College London, London, W2 1PG, UK.
| |
Collapse
|
207
|
Rapid and Lasting Effects of Activating BDNF-Expressing PVH Neurons on Energy Balance. eNeuro 2022; 9:ENEURO.0009-22.2022. [PMID: 35338053 PMCID: PMC8994543 DOI: 10.1523/eneuro.0009-22.2022] [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: 01/07/2022] [Revised: 02/15/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrkB), are implicit in causing obesity. Mutations that reduce BDNF and TrkB expression are associated with obesity in humans and mice. Recently, it was reported that Bdnf gene deletion in the neurons of the paraventricular hypothalamus (PVH) caused positive energy balance and severe obesity in the form of hyperphagia, impaired adaptive thermogenesis, and decreased energy expenditure. Thus, we hypothesize that activation of these neurons will have the opposite effect and provide an opportunity for long-lasting obesity treatment. To specifically activate BDNF-expressing PVH (PVHBDNF) neurons, we injected Cre-dependent adeno-associated virus (AAV) expressing the excitatory DREADD hM3Dq bilaterally into the PVH of Bdnf2A-Cre/+ knock-in mice and then administered clozapine-N-oxide (CNO). Using this technique, we demonstrated that acute activation of these neurons rapidly decreased normal nocturnal feeding and fasting-induced feeding in male and female mice. At thermoneutral temperatures, acute activation also rapidly increased adaptive thermogenesis, increased core body temperature, increased locomotion, increased energy expenditure, and decreased respiratory exchange ratio (RER) in male and female mice. These observations indicate that acute stimulation of PVHBDNF neurons promotes negative energy balance and weight loss. However, the rapid decrease in RER after activation of PVHBDNF neurons was followed by a delayed and prolonged increase in RER that remained elevated for 3 d in female mice. Thus, although acute activation of PVHBDNF neurons promotes negative energy balance in the short term, long-term effects of activation include sexually dimorphic overcompensatory mechanisms that may promote positive energy balance in female mice.
Collapse
|
208
|
Convergence of case-specific epigenetic alterations identify a confluence of genetic vulnerabilities tied to opioid overdose. Mol Psychiatry 2022; 27:2158-2170. [PMID: 35301427 PMCID: PMC9133127 DOI: 10.1038/s41380-022-01477-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/19/2022] [Accepted: 02/08/2022] [Indexed: 11/08/2022]
Abstract
Opioid use disorder is a highly heterogeneous disease driven by a variety of genetic and environmental risk factors which have yet to be fully elucidated. Opioid overdose, the most severe outcome of opioid use disorder, remains the leading cause of accidental death in the United States. We interrogated the effects of opioid overdose on the brain using ChIP-seq to quantify patterns of H3K27 acetylation in dorsolateral prefrontal cortical neurons isolated from 51 opioid-overdose cases and 51 accidental death controls. Among opioid cases, we observed global hypoacetylation and identified 388 putative enhancers consistently depleted for H3K27ac. Machine learning on H3K27ac patterns predicted case-control status with high accuracy. We focused on case-specific regulatory alterations, revealing 81,399 hypoacetylation events, uncovering vast inter-patient heterogeneity. We developed a strategy to decode this heterogeneity based on convergence analysis, which leveraged promoter-capture Hi-C to identify five genes over-burdened by alterations in their regulatory network or "plexus": ASTN2, KCNMA1, DUSP4, GABBR2, ENOX1. These convergent loci are enriched for opioid use disorder risk genes and heritability for generalized anxiety, number of sexual partners, and years of education. Overall, our multi-pronged approach uncovers neurobiological aspects of opioid use disorder and captures genetic and environmental factors perpetuating the opioid epidemic.
Collapse
|
209
|
Associations between KCNQ1 and ITIH4 gene polymorphisms and infant weight gain in early life. Pediatr Res 2022; 91:1290-1295. [PMID: 34247200 DOI: 10.1038/s41390-021-01601-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/17/2021] [Accepted: 05/20/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND An earlier meta-analysis of genome-wide association studies in Asian populations detected five novel body mass index-associated single-nucleotide polymorphisms (SNPs), including potassium voltage-gated channel subfamily Q member 1 (KCNQ1) (rs2237892), ALDH2/MYL2 (rs671, rs12229654), ITIH4 (rs2535633), and NT5C2 (rs11191580). Whether these SNPs take effect in early life, for example, affect infant rapid weight gain (RWG), is unclear. METHODS We obtained genomic DNA from 460 term infants with normal birth weight. RWG was defined as the change of weight-for-age standardized Z-score, calculated according to the Children Growth Standard released by the World Health Organization, from birth to 3 months of age >0.67. Using genetic models, associations between the candidate SNPs and infant RWG were examined, along with the interaction between the SNPs and the potential risk factors. RESULTS RWG was presented in 225 of 460 infants. SNP rs2535633 and rs2237892 were associated with the risk of RWG. Both additive and multiplicative interaction effects were found between infant delivery mode and rs2237892. The negative association between the rs2237892 T allele and infant RWG was only observed in vaginally delivered infants. CONCLUSIONS Obesity-related loci rs2535633 and rs2237892 are associated with infant RWG in the first 3 months of infancy. The relationship between rs2237892 and infant RGW might be moderated by cesarean delivery. IMPACT Genetic predisposition is an essential aspect to understand infant weight gain. Obesity-related SNPs, rs2535633 and rs2237892, are associated with RWG in very early years of life. The negative association between rs2237892 T allele and RWG is only observed in infants delivered vaginally instead of cesarean section.
Collapse
|
210
|
Saeed S, Janjua QM, Haseeb A, Khanam R, Durand E, Vaillant E, Ning L, Badreddine A, Berberian L, Boissel M, Amanzougarene S, Canouil M, Derhourhi M, Bonnefond A, Arslan M, Froguel P. Rare Variant Analysis of Obesity-Associated Genes in Young Adults With Severe Obesity From a Consanguineous Population of Pakistan. Diabetes 2022; 71:694-705. [PMID: 35061034 DOI: 10.2337/db21-0373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022]
Abstract
Recent advances in genetic analysis have significantly helped in progressively attenuating the heritability gap of obesity and have brought into focus monogenic variants that disrupt the melanocortin signaling. In a previous study, next-generation sequencing revealed a monogenic etiology in ∼50% of the children with severe obesity from a consanguineous population in Pakistan. Here we assess rare variants in obesity-causing genes in young adults with severe obesity from the same region. Genomic DNA from 126 randomly selected young adult obese subjects (BMI 37.2 ± 0.3 kg/m2; age 18.4 ± 0.3 years) was screened by conventional or augmented whole-exome analysis for point mutations and copy number variants (CNVs). Leptin, insulin, and cortisol levels were measured by ELISA. We identified 13 subjects carrying 13 different pathogenic or likely pathogenic variants in LEPR, PCSK1, MC4R, NTRK2, POMC, SH2B1, and SIM1. We also identified for the first time in the human, two homozygous stop-gain mutations in ASNSD1 and IFI16 genes. Inactivation of these genes in mouse models has been shown to result in obesity. Additionally, we describe nine homozygous mutations (seven missense, one stop-gain, and one stop-loss) and four copy-loss CNVs in genes or genomic regions previously linked to obesity-associated traits by genome-wide association studies. Unexpectedly, in contrast to obese children, pathogenic mutations in LEP and LEPR were either absent or rare in this cohort of young adults. High morbidity and mortality risks and social disadvantage of children with LEP or LEPR deficiency may in part explain this difference between the two cohorts.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Qasim M Janjua
- Department of Physiology and Biophysics, National University of Science and Technology, Sohar, Oman
| | - Attiya Haseeb
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Roohia Khanam
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Emmanuelle Durand
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Emmanuel Vaillant
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Lijiao Ning
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Alaa Badreddine
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Lionel Berberian
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Mathilde Boissel
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Souhila Amanzougarene
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Mickaël Canouil
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Mehdi Derhourhi
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Amélie Bonnefond
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Muhammad Arslan
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Philippe Froguel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| |
Collapse
|
211
|
Schlauch KA, Read RW, Neveux I, Lipp B, Slonim A, Grzymski JJ. The Impact of ACEs on BMI: An Investigation of the Genotype-Environment Effects of BMI. Front Genet 2022; 13:816660. [PMID: 35342390 PMCID: PMC8942770 DOI: 10.3389/fgene.2022.816660] [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: 11/17/2021] [Accepted: 02/04/2022] [Indexed: 12/31/2022] Open
Abstract
Adverse Childhood Experiences are stressful and traumatic events occurring before the age of eighteen shown to cause mental and physical health problems, including increased risk of obesity. Obesity remains an ongoing national challenge with no predicted solution. We examine a subset of the Healthy Nevada Project, focusing on a multi-ethnic cohort of 15,886 sequenced participants with recalled adverse childhood events, to study how ACEs and their genotype-environment interactions affect BMI. Specifically, the Healthy Nevada Project participants sequenced by the Helix Exome+ platform were cross-referenced to their electronic medical records and social health determinants questionnaire to identify: 1) the effect of ACEs on BMI in the absence of genetics; 2) the effect of genotype-environment interactions on BMI; 3) how these gene-environment interactions differ from standard genetic associations of BMI. The study found very strong significant associations between the number of adverse childhood experiences and adult obesity. Additionally, we identified fifty-five common and rare variants that exhibited gene-interaction effects including three variants in the CAMK1D gene and four variants in LHPP; both genes are linked to schizophrenia. Surprisingly, none of the variants identified with interactive effects were in canonical obesity-related genes. Here we show the delicate balance between genes and environment, and how the two strongly influence each other.
Collapse
Affiliation(s)
- Karen A Schlauch
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Robert W Read
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Iva Neveux
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Bruce Lipp
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | | | - Joseph J Grzymski
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States.,Renown Health, Reno, NV, United States
| |
Collapse
|
212
|
Fu L, Wang Y, Li T, Yang S, Hu YQ. A Novel Hierarchical Clustering Approach for Joint Analysis of Multiple Phenotypes Uncovers Obesity Variants Based on ARIC. Front Genet 2022; 13:791920. [PMID: 35391794 PMCID: PMC8981031 DOI: 10.3389/fgene.2022.791920] [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: 10/21/2021] [Accepted: 01/27/2022] [Indexed: 12/02/2022] Open
Abstract
Genome-wide association studies (GWASs) have successfully discovered numerous variants underlying various diseases. Generally, one-phenotype one-variant association study in GWASs is not efficient in identifying variants with weak effects, indicating that more signals have not been identified yet. Nowadays, jointly analyzing multiple phenotypes has been recognized as an important approach to elevate the statistical power for identifying weak genetic variants on complex diseases, shedding new light on potential biological mechanisms. Therefore, hierarchical clustering based on different methods for calculating correlation coefficients (HCDC) is developed to synchronously analyze multiple phenotypes in association studies. There are two steps involved in HCDC. First, a clustering approach based on the similarity matrix between two groups of phenotypes is applied to choose a representative phenotype in each cluster. Then, we use existing methods to estimate the genetic associations with the representative phenotypes rather than the individual phenotypes in every cluster. A variety of simulations are conducted to demonstrate the capacity of HCDC for boosting power. As a consequence, existing methods embedding HCDC are either more powerful or comparable with those of without embedding HCDC in most scenarios. Additionally, the application of obesity-related phenotypes from Atherosclerosis Risk in Communities via existing methods with HCDC uncovered several associated variants. Among these, UQCC1-rs1570004 is reported as a significant obesity signal for the first time, whose differential expression in subcutaneous fat, visceral fat, and muscle tissue is worthy of further functional studies.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-communicable Disease Management, National Center for Children’s Health, Beijing Children’s Hospital, Capital Medical University, Beijing, China
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuquan Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Tingting Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Siqian Yang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yue-Qing Hu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
- Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
213
|
Lu C, Wang HJ, Song JY, Wang S, Li XY, Huang T, Wang H. Fine Mapping of the MAP2K5 Region Identified rs7175517 as a Causal Variant Related to BMI in China and the United Kingdom Populations. Front Genet 2022; 13:838685. [PMID: 35368675 PMCID: PMC8967323 DOI: 10.3389/fgene.2022.838685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/10/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Genome-wide association studies (GWASs) have consistently identified MAP2K5 as an obesity susceptibility gene. To deepen our understanding of the potential causal genetic variants of this region, a fine-mapping study of MAP2K5 was conducted. Methods and Results: SNPs rs7175517 (G > A) and rs4776970 (T > A) were identified as the leading SNPs associated with BMI in both Chinese and the United Kingdom populations. Second, colocalization of GWAS and expression quantitative trait loci (eQTL) analyses and bioinformatic analyses indicated that rs7175517 is the functionally leading variant in the MAP2K5 gene region. Dual-luciferase assays indicated that the G allele of rs7175517 reduced the mRNA expression of MAP2K5 in HEK293T cells. The possible mechanism was that the G allele interacted with more RNA repressors from nuclei extracts, which was evidenced by electrophoretic mobility shift assays (EMSAs). Furthermore, the pathway enrichment analyses of the products from DNA pull-down and protein mass spectrometry demonstrated that the G allele of rs7175517 might interact with RNA catabolic or splicing transcription factors, which consequentially increased adiposity deposition. Conclusion: SNP rs7175517 of the MAP2K5 gene was the putative causal variant associated with BMI. More precisely designed in vitro or animal experiments are warranted to further delineate the function of MAP2K5 in adipogenesis.
Collapse
Affiliation(s)
- Ce Lu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hai-Jun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| | - Jie-Yun Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Shuo Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing, China
| | - Xue-Ying Li
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Hui Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
- *Correspondence: Hui Wang,
| |
Collapse
|
214
|
Wang R, Snieder H, Hartman CA. Familial co-aggregation and shared heritability between depression, anxiety, obesity and substance use. Transl Psychiatry 2022; 12:108. [PMID: 35296640 PMCID: PMC8927111 DOI: 10.1038/s41398-022-01868-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/09/2022] Open
Abstract
Depression, anxiety, obesity and substance use are heritable and often co-occur. However, the mechanisms underlying this co-occurrence are not fully understood. We estimated their familial aggregation and co-aggregation as well as heritabilities and genetic correlations to improve etiological understanding. Data came from the multi-generational population-based Lifelines Cohort Study (n = 162,439). Current depression and anxiety were determined using the MINI International Neuropsychiatric Interview. Smoking, alcohol and drug use were assessed by self-report questionnaires. Body mass index (BMI) and obesity were calculated by measured height and weight. Modified Cox proportional hazards models estimated recurrence risk ratios (λR), and restricted maximum likelihood variance decomposition methods estimated heritabilities (h2) and genetic correlations (rG). All analyses were adjusted for age, age2, and sex. Depression, anxiety, obesity and substance use aggregated within families (λR first-degree relative = 1.08-2.74) as well as between spouses (λR = 1.11-6.60). All phenotypes were moderately heritable (from h2depression = 0.25 to h2BMI = 0.53). Depression, anxiety, obesity and smoking showed positive familial co-aggregation. That is, each of these traits confers increased risk on the other ones within families, consistent with the positive genetic correlations between these phenotypes (rG = 0.16-0.94). The exception was obesity, which showed a negative co-aggregation with alcohol and drug use and vice versa, consistent with the negative genetic correlations of BMI with alcohol (rG = -0.14) and soft drug use (rG = -0.10). Patterns of cross-phenotype recurrence risk highlight the co-occurrence among depression, anxiety, obesity and substance use within families. Patterns of genetic overlap between these phenotypes provide clues to uncovering the mechanisms underlying familial co-aggregation.
Collapse
Affiliation(s)
- Rujia Wang
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| | - Harold Snieder
- grid.4494.d0000 0000 9558 4598Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Catharina A. Hartman
- grid.4494.d0000 0000 9558 4598Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
215
|
Williams MJ, Alsehli AM, Gartner SN, Clemensson LE, Liao S, Eriksson A, Isgrove K, Thelander L, Khan Z, Itskov PM, Moulin TC, Ambrosi V, Al-Sabri MH, Lagunas-Rangel FA, Olszewski PK, Schiöth HB. The Statin Target Hmgcr Regulates Energy Metabolism and Food Intake through Central Mechanisms. Cells 2022; 11:cells11060970. [PMID: 35326421 PMCID: PMC8946516 DOI: 10.3390/cells11060970] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
The statin drug target, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), is strongly linked to body mass index (BMI), yet how HMGCR influences BMI is not understood. In mammals, studies of peripheral HMGCR have not clearly identified a role in BMI maintenance and, despite considerable central nervous system expression, a function for central HMGCR has not been determined. Similar to mammals, Hmgcr is highly expressed in the Drosophila melanogaster brain. Therefore, genetic and pharmacological studies were performed to identify how central Hmgcr regulates Drosophila energy metabolism and feeding behavior. We found that inhibiting Hmgcr, in insulin-producing cells of the Drosophila pars intercerebralis (PI), the fly hypothalamic equivalent, significantly reduces the expression of insulin-like peptides, severely decreasing insulin signaling. In fact, reducing Hmgcr expression throughout development causes decreased body size, increased lipid storage, hyperglycemia, and hyperphagia. Furthermore, the Hmgcr induced hyperphagia phenotype requires a conserved insulin-regulated α-glucosidase, target of brain insulin (tobi). In rats and mice, acute inhibition of hypothalamic Hmgcr activity stimulates food intake. This study presents evidence of how central Hmgcr regulation of metabolism and food intake could influence BMI.
Collapse
Affiliation(s)
- Michael J. Williams
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Ahmed M. Alsehli
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
- Faculty of Medicine, King Abdulaziz University and Hospital, Al Ehtifalat St., Jeddah 21589, Saudi Arabia
| | - Sarah N. Gartner
- Department of Biological Sciences, University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand; (S.N.G.); (K.I.); (P.K.O.)
| | - Laura E. Clemensson
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Sifang Liao
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Anders Eriksson
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Kiriana Isgrove
- Department of Biological Sciences, University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand; (S.N.G.); (K.I.); (P.K.O.)
| | - Lina Thelander
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Zaid Khan
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
- Department of Plant Protection Biology, Swedish University of Agricultural Sciences (SLU), Sundsvägen 14, 230 53 Alnarp, Sweden
| | - Pavel M. Itskov
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Thiago C. Moulin
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Valerie Ambrosi
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Mohamed H. Al-Sabri
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Francisco Alejandro Lagunas-Rangel
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
| | - Pawel K. Olszewski
- Department of Biological Sciences, University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand; (S.N.G.); (K.I.); (P.K.O.)
| | - Helgi B. Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, 751 24 Uppsala, Sweden; (M.J.W.); (A.M.A.); (L.E.C.); (S.L.); (A.E.); (L.T.); (Z.K.); (P.M.I.); (T.C.M.); (V.A.); (M.H.A.-S.); (F.A.L.-R.)
- Correspondence:
| |
Collapse
|
216
|
Meliț LE, Mărginean CO, Săsăran MO. The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota. Biomedicines 2022; 10:biomedicines10030645. [PMID: 35327446 PMCID: PMC8945275 DOI: 10.3390/biomedicines10030645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 02/04/2023] Open
Abstract
The era of pediatric obesity is no longer a myth. Unfortunately, pediatric obesity has reached alarming incidence levels worldwide and the factors that contribute to its development have been intensely studied in multiple recent and emerging studies. Gut microbiota was recently included in the wide spectrum of factors implicated in the determination of obesity, but its role in pediatric obese patients is far from being fully understood. In terms of the infant gut microbiome, multiple factors have been demonstrated to shape its content, including maternal diet and health, type of delivery, feeding patterns, weaning and dietary habits. Nevertheless, the role of the intrauterine environment, such as the placental microbial community, cannot be completely excluded. Most studies have identified Firmicutes and Bacteroidetes as the most important players related to obesity risk in gut microbiota reflecting an increase of Firmicutes and a decrease in Bacteroidetes in the context of obesity; however, multiple inconsistencies between studies were recently reported, especially in pediatric populations, and there is a scarcity of studies performed in this age group.
Collapse
Affiliation(s)
- Lorena Elena Meliț
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| | - Cristina Oana Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
- Correspondence:
| | - Maria Oana Săsăran
- Department of Pediatrics III, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| |
Collapse
|
217
|
Jais A, Brüning JC. Arcuate Nucleus-Dependent Regulation of Metabolism-Pathways to Obesity and Diabetes Mellitus. Endocr Rev 2022; 43:314-328. [PMID: 34490882 PMCID: PMC8905335 DOI: 10.1210/endrev/bnab025] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 01/12/2023]
Abstract
The central nervous system (CNS) receives information from afferent neurons, circulating hormones, and absorbed nutrients and integrates this information to orchestrate the actions of the neuroendocrine and autonomic nervous systems in maintaining systemic metabolic homeostasis. Particularly the arcuate nucleus of the hypothalamus (ARC) is of pivotal importance for primary sensing of adiposity signals, such as leptin and insulin, and circulating nutrients, such as glucose. Importantly, energy state-sensing neurons in the ARC not only regulate feeding but at the same time control multiple physiological functions, such as glucose homeostasis, blood pressure, and innate immune responses. These findings have defined them as master regulators, which adapt integrative physiology to the energy state of the organism. The disruption of this fine-tuned control leads to an imbalance between energy intake and expenditure as well as deregulation of peripheral metabolism. Improving our understanding of the cellular, molecular, and functional basis of this regulatory principle in the CNS could set the stage for developing novel therapeutic strategies for the treatment of obesity and metabolic syndrome. In this review, we summarize novel insights with a particular emphasis on ARC neurocircuitries regulating food intake and glucose homeostasis and sensing factors that inform the brain of the organismal energy status.
Collapse
Affiliation(s)
- Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,National Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
218
|
Alnafjan AA, Alkhuriji AF, Alobaid HM, Babay ZA, Khalil MI. Association of FTO gene variants rs9939609 and rs1421085 with polycystic ovary syndrome. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00263-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) is among the most common complex genetic endocrinopathy, and its etiology and pathophysiology remain controversial. FTO is a large highly polymorphic gene and was coined as the first locus associated with adiposity. The association of the intronic variant FTO rs9939609 or FTO rs1421085 with PCOS has been controversial and unclear, mainly due to ethnic differences among populations. The present study aims to investigate the association of FTO rs9939609 or FTO rs1421085 polymorphisms with PCOS in Saudi Arabian women.
Results
A total of 98 PCOS patients and 99 healthy females were included in this study. PCR and genotyping (TaqMan®SNP Genotyping Assay) were employed. For FTO rs9939609, the genotype TA and the recessive model (TA + AA) in PCOS patients were significantly different compared with control subjects (p = 0.008 and p = 0.007, respectively). The allele frequency of the FTO rs9939609 gene variant was associated significantly (p = 0.027) with PCOS, suggesting that the A allele is a risk factor for PCOS susceptibility. However, for the FTO rs1421085 variant, the genotype and allele distributions did not differ significantly between PCOS patients and controls (p > 0.05).
Conclusions
This is the first report to study the association of FTO rs9939609 and FTO rs1421085 with PCOS in Saudi women. Results suggest that the FTO rs9939609 gene variant could be a genetic predisposing factor for PCOS Saudi women.
Collapse
|
219
|
Cataloging the potential SNPs (single nucleotide polymorphisms) associated with quantitative traits, viz. BMI (body mass index), IQ (intelligence quotient) and BP (blood pressure): an updated review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Single nucleotide polymorphism (SNP) variants are abundant, persistent and widely distributed across the genome and are frequently linked to the development of genetic diseases. Identifying SNPs that underpin complex diseases can aid scientists in the discovery of disease-related genes by allowing for early detection, effective medication and eventually disease prevention.
Main body
Various SNP or polymorphism-based studies were used to categorize different SNPs potentially related to three quantitative traits: body mass index (BMI), intelligence quotient (IQ) and blood pressure, and then uncovered common SNPs for these three traits. We employed SNPedia, RefSNP Report, GWAS Catalog, Gene Cards (Data Bases), PubMed and Google Scholar search engines to find relevant material on SNPs associated with three quantitative traits. As a result, we detected three common SNPs for all three quantitative traits in global populations: SNP rs6265 of the BDNF gene on chromosome 11p14.1, SNP rs131070325 of the SL39A8 gene on chromosome 4p24 and SNP rs4680 of the COMT gene on chromosome 22q11.21.
Conclusion
In our review, we focused on the prevalent SNPs and gene expression activities that influence these three quantitative traits. These SNPs have been used to detect and map complex, common illnesses in communities for homogeneity testing and pharmacogenetic studies. High blood pressure, diabetes and heart disease, as well as BMI, schizophrenia and IQ, can all be predicted using common SNPs. Finally, the results of our work can be used to find common SNPs and genes that regulate these three quantitative features across the genome.
Collapse
|
220
|
Marcos-Pasero H, Aguilar-Aguilar E, de la Iglesia R, Espinosa-Salinas I, Molina S, Colmenarejo G, Martínez JA, Ramírez de Molina A, Reglero G, Loria-Kohen V. "GENYAL" Study to Childhood Obesity Prevention: Methodology and Preliminary Results. Front Nutr 2022; 9:777384. [PMID: 35350411 PMCID: PMC8957940 DOI: 10.3389/fnut.2022.777384] [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: 09/15/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Objective This article describes the methodology and summarizes some preliminary results of the GENYAL study aiming to design and validate a predictive model, considering both environmental and genetic factors, that identifies children who would benefit most from actions aimed at reducing the risk of obesity and its complications. Design The study is a cluster randomized clinical trial with 5-year follow-up. The initial evaluation was carried out in 2017. The schools were randomly split into intervention (nutritional education) and control schools. Anthropometric measurements, social and health as well as dietary and physical activity data of schoolchildren and their families are annually collected. A total of 26 single nucleotide polymorphisms (SNPs) were assessed. Machine Learning models are being designed to predict obesity phenotypes after the 5-year follow-up. Settings Six schools in Madrid. Participants A total of 221 schoolchildren (6-8 years old). Results Collected results show that the prevalence of excess weight was 19.0, 25.4, and 32.2% (according to World Health Organization, International Obesity Task Force and Orbegozo Foundation criteria, respectively). Associations between the nutritional state of children with mother BMI [β = 0.21 (0.13-0.3), p (adjusted) <0.001], geographical location of the school [OR = 2.74 (1.24-6.22), p (adjusted) = 0.06], dairy servings per day [OR = 0.48 (0.29-0.75), p (adjusted) = 0.05] and 8 SNPs [rs1260326, rs780094, rs10913469, rs328, rs7647305, rs3101336, rs2568958, rs925946; p (not adjusted) <0.05] were found. Conclusions These baseline data support the evidence that environmental and genetic factors play a role in the development of childhood obesity. After 5-year follow-up, the GENYAL study pretends to validate the predictive model as a new strategy to fight against obesity. Clinical Trial Registration This study has been registered in ClinicalTrials.gov with the identifier NCT03419520, https://clinicaltrials.gov/ct2/show/NCT03419520.
Collapse
Affiliation(s)
- Helena Marcos-Pasero
- Nutrition and Clinical Trials Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
- Faculty of Health Sciences, Valencian International University (VIU), Valencia, Spain
| | - Elena Aguilar-Aguilar
- Nutrition and Clinical Trials Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Rocío de la Iglesia
- Departamento de Ciencias Farmaceúticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Isabel Espinosa-Salinas
- Nutritional Genomics and Health Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Susana Molina
- GenyalLab, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Gonzalo Colmenarejo
- Biostatistics and Bioinformatics Unit, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - J. Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
- IdisNA, Navarra Institute for Health Research, Pamplona, Spain
- Center of Biomedical Research in Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology and Nutritional Genomics of Cancer, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Guillermo Reglero
- Production and Development of Foods for Health, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), CEI UAM+CSIC, Madrid, Spain
| | - Viviana Loria-Kohen
- Nutrition and Clinical Trials Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Farmacia, Universidad Complutense de Madrid, Grupo de Investigación VALORNUT-UCM, Madrid, Spain
| |
Collapse
|
221
|
Ni A, Ernst C. Evidence That Substantia Nigra Pars Compacta Dopaminergic Neurons Are Selectively Vulnerable to Oxidative Stress Because They Are Highly Metabolically Active. Front Cell Neurosci 2022; 16:826193. [PMID: 35308118 PMCID: PMC8931026 DOI: 10.3389/fncel.2022.826193] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/28/2022] [Indexed: 12/21/2022] Open
Abstract
There are 400–500 thousand dopaminergic cells within each side of the human substantia nigra pars compacta (SNpc) making them a minuscule portion of total brain mass. These tiny clusters of cells have an outsized impact on motor output and behavior as seen in disorders such as Parkinson’s disease (PD). SNpc dopaminergic neurons are more vulnerable to oxidative stress compared to other brain cell types, but the reasons for this are not precisely known. Here we provide evidence to support the hypothesis that this selective vulnerability is because SNpc neurons sustain high metabolic rates compared to other neurons. A higher baseline requirement for ATP production may lead to a selective vulnerability to impairments in oxidative phosphorylation (OXPHOS) or genetic insults that impair Complex I of the electron transport chain. We suggest that the energy demands of the unique morphological and electrophysiological properties of SNpc neurons may be one reason these cells produce more ATP than other cells. We further provide evidence to support the hypothesis that transcription factors (TFs) required to drive induction, differentiation, and maintenance of midbrain dopaminergic neural progenitor cells which give rise to terminally differentiated SNpc neurons are uniquely involved in both developmental patterning and metabolism, a dual function unlike other TFs that program neurons in other brain regions. The use of these TFs during induction and differentiation may program ventral midbrain progenitor cells metabolically to higher ATP levels, allowing for the development of those specialized cell processes seen in terminally differentiated cells. This paper provides a cellular and developmental framework for understanding the selective vulnerability of SNpc dopaminergic cells to oxidative stress.
Collapse
|
222
|
Balkhiyarova Z, Luciano R, Kaakinen M, Ulrich A, Shmeliov A, Bianchi M, Chioma L, Dallapiccola B, Prokopenko I, Manco M. Relationship between glucose homeostasis and obesity in early life-a study of Italian children and adolescents. Hum Mol Genet 2022; 31:816-826. [PMID: 34590674 PMCID: PMC8895752 DOI: 10.1093/hmg/ddab287] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 01/09/2023] Open
Abstract
Epidemic obesity is the most important risk factor for prediabetes and type 2 diabetes (T2D) in youth as it is in adults. Obesity shares pathophysiological mechanisms with T2D and is likely to share part of the genetic background. We aimed to test if weighted genetic risk scores (GRSs) for T2D, fasting glucose (FG) and fasting insulin (FI) predict glycaemic traits and if there is a causal relationship between obesity and impaired glucose metabolism in children and adolescents. Genotyping of 42 SNPs established by genome-wide association studies for T2D, FG and FI was performed in 1660 Italian youths aged between 2 and 19 years. We defined GRS for T2D, FG and FI and tested their effects on glycaemic traits, including FG, FI, indices of insulin resistance/beta cell function and body mass index (BMI). We evaluated causal relationships between obesity and FG/FI using one-sample Mendelian randomization analyses in both directions. GRS-FG was associated with FG (beta = 0.075 mmol/l, SE = 0.011, P = 1.58 × 10-11) and beta cell function (beta = -0.041, SE = 0.0090 P = 5.13 × 10-6). GRS-T2D also demonstrated an association with beta cell function (beta = -0.020, SE = 0.021 P = 0.030). We detected a causal effect of increased BMI on levels of FI in Italian youths (beta = 0.31 ln (pmol/l), 95%CI [0.078, 0.54], P = 0.0085), while there was no effect of FG/FI levels on BMI. Our results demonstrate that the glycaemic and T2D risk genetic variants contribute to higher FG and FI levels and decreased beta cell function in children and adolescents. The causal effects of adiposity on increased insulin resistance are detectable from childhood age.
Collapse
Affiliation(s)
- Zhanna Balkhiyarova
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre Russian Academy of Sciences, Ufa 450008, Russian Federation
- Department of Endocrinology, Bashkir State Medical University, Ufa 450054, Russian Federation
| | - Rosa Luciano
- Research Area for Multifactorial Disease, Bambino Gesù Children’s Hospital, IRCCS, Rome 00146, Italy
- Department of Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, Rome 00146, Italy
| | - Marika Kaakinen
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Anna Ulrich
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Aleksey Shmeliov
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Marzia Bianchi
- Research Area for Multifactorial Disease, Bambino Gesù Children’s Hospital, IRCCS, Rome 00146, Italy
| | - Laura Chioma
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome 00146, Italy
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome 00146, Italy
| | - Inga Prokopenko
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre Russian Academy of Sciences, Ufa 450008, Russian Federation
- UMR 8199—EGID, Institut Pasteur de Lille, CNRS, University of Lille, Lille 59000, France
| | - Melania Manco
- Research Area for Multifactorial Disease, Bambino Gesù Children’s Hospital, IRCCS, Rome 00146, Italy
| |
Collapse
|
223
|
Szalanczy AM, Key CCC, Woods LCS. Genetic variation in satiety signaling and hypothalamic inflammation: merging fields for the study of obesity. J Nutr Biochem 2022; 101:108928. [PMID: 34936921 PMCID: PMC8959400 DOI: 10.1016/j.jnutbio.2021.108928] [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: 06/29/2021] [Revised: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
Although obesity has been a longstanding health crisis, the genetic architecture of the disease remains poorly understood. Genome-wide association studies have identified many genomic loci associated with obesity, with genes being enriched in the brain, particularly in the hypothalamus. This points to the role of the central nervous system (CNS) in predisposition to obesity, and we emphasize here several key genes along the satiety signaling pathway involved in genetic susceptibility. Interest has also risen regarding the chronic, low-grade obesity-associated inflammation, with a growing concern toward inflammation in the hypothalamus as a precursor to obesity. Recent studies have found that genetic variation in inflammatory genes play a role in obesity susceptibility, and we highlight here several key genes. Despite the interest in the genetic variants of these pathways individually, there is a lack of research that investigates the relationship between the two. Understanding the interplay between genetic variation in obesity genes enriched in the CNS and inflammation genes will advance our understanding of obesity etiology and heterogeneity, improve genetic risk prediction analyses, and highlight new drug targets for the treatment of obesity. Additionally, this increased knowledge will assist in physician's ability to develop personalized nutrition and medication strategies for combating the obesity epidemic. Though it often seems to present universally, obesity is a highly individual disease, and there remains a need in the field to develop methods to treat at the individual level.
Collapse
|
224
|
Li D, Zhang B, Cheng J, Chen D, Wu Y, Luo Q, Zhou L. Obesity-related genetic polymorphisms are associated with the risk of early puberty in Han Chinese girls. Clin Endocrinol (Oxf) 2022; 96:319-327. [PMID: 34761429 DOI: 10.1111/cen.14631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/31/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To explore the association between obesity and precocious puberty from the perspective of genetic polymorphism. DESIGN Two hundred and ninety-eight pairs of girls in early puberty and age-matched controls (±3 months) were recruited. The genotypes of four obesity-related single-nucleotide polymorphism (SNP) loci (rs10968576, rs12935153, rs4674340 and rs7635103) were determined and the effect of variation on early puberty in Chinese Han girls was evaluated. The unstimulated luteinizing hormone (LH), follicle-stimulating hormone and estradiol levels were also measured to determine the relationship with SNP polymorphisms. RESULTS The effect allele A of rs12935153 was associated with early puberty (odds ratio [OR] = 1.256, 95% confidence interval [CI]: 1.010-1.585), but the significance disappeared after multiple comparisons. After adjusting for body mass index, rs12935153 variation increased the risk of early puberty in additive (OR = 1.589, 95% CI: 1.222-2.066), dominant (OR = 1.788, 95% CI: 1.210-2.642) and recessive (OR = 1.915, 95% CI: 1.207-3.038) models of inheritance. Individuals harbouring AA genotype in rs12935153 had a risk of higher LH levels than that of wild type (OR = 1.668, 95% CI: 1.093-2.546). CONCLUSIONS The association between obesity and precocity can be explained from a genetic perspective. Our study suggests that variations in rs12935153 increase the risk of early puberty in Chinese girls. Further studies are needed to verify our findings.
Collapse
Affiliation(s)
- Di Li
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
- Department of Clinical Nutrition, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Boxian Zhang
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
- Medical department, Hospital of Stomatology, SunYat-sen University, Guangzhou, Guangdong, China
| | - Jinquan Cheng
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Dingyan Chen
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Yu Wu
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Qingshan Luo
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Li Zhou
- Department of School Hygiene, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| |
Collapse
|
225
|
Subjective cognitive decline and total energy intake: Talk too much? Eur J Epidemiol 2022; 37:129-131. [PMID: 35211870 DOI: 10.1007/s10654-022-00849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/03/2022]
Abstract
The increasing longevity of the population has resulted in dementia becoming a leading cause of both death and disability. Dementia is not a single disease. Studies of rare Mendelian disorders have documented that Alzheimer's disease, the most common cause of dementia, is associated with a long incubation period from amyloid deposition to neurodegeneration to mild cognitive impairment and dementia. There are three broad hypotheses related to the causes of Alzheimer's dementia: (1) an aging process; (2) brain vascular disease; and (3) metabolic abnormalities associated with either increased production of amyloid-β or decreased clearance from the brain. Therefore, research on the early stages of the dementia process are of high priority. This paper reports that higher energy intake in both the Nurses' Health Study and Health Professionals Follow-up Study is associated with very early symptoms that lead to mild cognitive impairment and dementia. The results are very interesting but hard to interpret because they also show that higher energy intake is not related to body mass index, a very unusual observation. A likely hypothesis is that there is an association between reporting of dietary intake and subjective symptoms, i.e. reporting bias, accounting for their results.
Collapse
|
226
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 3174] [Impact Index Per Article: 1058.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
227
|
Antoine D, Guéant-Rodriguez RM, Chèvre JC, Hergalant S, Sharma T, Li Z, Rouyer P, Chery C, Halvick S, Bui C, Oussalah A, Ziegler O, Quilliot D, Brunaud L, Guéant JL, Meyre D. Low-frequency Coding Variants Associated With Body Mass Index Affect the Success of Bariatric Surgery. J Clin Endocrinol Metab 2022; 107:e1074-e1084. [PMID: 34718599 DOI: 10.1210/clinem/dgab774] [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: 05/02/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT A recent study identified 14 low-frequency coding variants associated with body mass index (BMI) in 718 734 individuals predominantly of European ancestry. OBJECTIVE We investigated the association of 2 genetic scores (GS) with i) the risk of severe/morbid obesity, ii) BMI variation before weight-loss intervention, iii) BMI change in response to an 18-month lifestyle/behavioral intervention program, and iv) BMI change up to 24 months after bariatric surgery. METHODS The 14 low-frequency coding variants were genotyped or sequenced in 342 French adults with severe/morbid obesity and 574 French adult controls from the general population. We built risk and protective GS based on 6 BMI-increasing and 5 BMI-decreasing low-frequency coding variants that were polymorphic in our study. RESULTS While the risk GS was not associated with severe/morbid obesity status, BMI-decreasing low-frequency coding variants were significantly less frequent in patients with severe/morbid obesity than in French adults from the general population. Neither the risk nor the protective GS was associated with BMI before intervention in patients with severe/morbid obesity, nor did they affect BMI change in response to a lifestyle/behavioral modification program. The protective GS was associated with a greater BMI decrease following bariatric surgery. The risk and protective GS were associated with a higher and lower risk of BMI regain after bariatric surgery. CONCLUSION Our data indicate that in populations of European descent, low-frequency coding variants associated with BMI in the general population also affect the outcomes of bariatric surgery in patients with severe/morbid obesity.
Collapse
Affiliation(s)
- Darlène Antoine
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Jean-Claude Chèvre
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Sébastien Hergalant
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Tanmay Sharma
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Zhen Li
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
- Specialized Obesity Center and Endocrinology, Diabetology, department of Nutrition, Brabois Hospital, CHRU of Nancy, 54500 Vandoeuvre-Les-Nancy, France
| | - Pierre Rouyer
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Céline Chery
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Sarah Halvick
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Catherine Bui
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Abderrahim Oussalah
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - Olivier Ziegler
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
- Specialized Obesity Center and Endocrinology, Diabetology, department of Nutrition, Brabois Hospital, CHRU of Nancy, 54500 Vandoeuvre-Les-Nancy, France
- Department of Surgery, Endocrine and metabolic surgery, Multidisciplinary unit for obesity surgery (CVMC), University Hospital Centre of Nancy, Brabois Hospital, 54500 Nancy, France
| | - Didier Quilliot
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
- Specialized Obesity Center and Endocrinology, Diabetology, department of Nutrition, Brabois Hospital, CHRU of Nancy, 54500 Vandoeuvre-Les-Nancy, France
- Department of Surgery, Endocrine and metabolic surgery, Multidisciplinary unit for obesity surgery (CVMC), University Hospital Centre of Nancy, Brabois Hospital, 54500 Nancy, France
| | - Laurent Brunaud
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
- Department of Surgery, Endocrine and metabolic surgery, Multidisciplinary unit for obesity surgery (CVMC), University Hospital Centre of Nancy, Brabois Hospital, 54500 Nancy, France
| | - Jean-Louis Guéant
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
| | - David Meyre
- Inserm UMR_S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
- FHU ARRIMAGE, department of Biochemistry-Molecular Biology-Nutrition, University Hospital Centre of Nancy, 54500 Nancy, France
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
228
|
Cai Z, Christensen OF, Lund MS, Ostersen T, Sahana G. Large-scale association study on daily weight gain in pigs reveals overlap of genetic factors for growth in humans. BMC Genomics 2022; 23:133. [PMID: 35168569 PMCID: PMC8845347 DOI: 10.1186/s12864-022-08373-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/08/2022] [Indexed: 01/10/2023] Open
Abstract
Background Imputation from genotyping array to whole-genome sequence variants using resequencing of representative reference populations enhances our ability to map genetic factors affecting complex phenotypes in livestock species. The accumulation of knowledge about gene function in human and laboratory animals can provide substantial advantage for genomic research in livestock species. Results In this study, 201,388 pigs from three commercial Danish breeds genotyped with low to medium (8.5k to 70k) SNP arrays were imputed to whole genome sequence variants using a two-step approach. Both imputation steps achieved high accuracies, and in total this yielded 26,447,434 markers on 18 autosomes. The average estimated imputation accuracy of markers with minor allele frequency ≥ 0.05 was 0.94. To overcome the memory consumption of running genome-wide association study (GWAS) for each breed, we performed within-breed subpopulation GWAS then within-breed meta-analysis for average daily weight gain (ADG), followed by a multi-breed meta-analysis of GWAS summary statistics. We identified 15 quantitative trait loci (QTL). Our post-GWAS analysis strategy to prioritize of candidate genes including information like gene ontology, mammalian phenotype database, differential expression gene analysis of high and low feed efficiency pig and human GWAS catalog for height, obesity, and body mass index, we proposed MRAP2, LEPROT, PMAIP1, ENSSSCG00000036234, BMP2, ELFN1, LIG4 and FAM155A as the candidate genes with biological support for ADG in pigs. Conclusion Our post-GWAS analysis strategy helped to identify candidate genes not just by distance to the lead SNP but also by multiple sources of biological evidence. Besides, the identified QTL overlap with genes which are known for their association with human growth-related traits. The GWAS with this large data set showed the power to map the genetic factors associated with ADG in pigs and have added to our understanding of the genetics of growth across mammalian species. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08373-3.
Collapse
Affiliation(s)
- Zexi Cai
- Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark.
| | | | - Mogens Sandø Lund
- Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark
| | - Tage Ostersen
- SEGES Danish Pig Research Centre, Agro Food Park 15, 8200, Aarhus N, Denmark
| | - Goutam Sahana
- Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark
| |
Collapse
|
229
|
Zhang Y, Li X, Hu Y, Yuan H, Wu X, Yang Y, Zhao T, Hu K, Wang Z, Wang G, Zhang K, Liu H. Evaluation of mild cognitive impairment genetic susceptibility risks in a Chinese population. BMC Psychiatry 2022; 22:93. [PMID: 35135506 PMCID: PMC8822756 DOI: 10.1186/s12888-022-03756-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/02/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Mild cognitive impairment (MCI) is a kind of non-functional cognitive decline between normal aging and dementia. With the increase of individual age, the quality of cognitive function has become a more and more important topic. The study of gene loci in patients with MCI is essential for the prevention of dementia. In this study, we evaluate the gene polymorphism in Chinese Han patients with MCI by propensity score matching (PSM) and comparing them to healthy control (HC) subjects. METHODS Four hundred seventeen patients with mild cognitive impairment and 508 healthy people were included. The two groups were matched by applying one-to-one PSM, and the matching tolerance was set to 0.002. The matching covariates included gender,age,occupation,marital status,living mode. Then, a case-control associated analysis was conducted to analyze the genotype and allele frequencies of single nucleotide polymorphisms (SNPs) in the MCI group and the control group. RESULTS Three hundred eleven cases were successfully matched in each group, and there was no statistical difference on all the matching variables, gender, age, occupation, marital status, living mode between two groups after the match (P > 0.05). The allele frequency of bridging integrator 1(BIN1) rs7561528 showed minimal association with MCI in the Han Chinese population (P = 0.01). Compared with the healthy control (HC) group, A allele frequency of MCI group patients was significantly decreased. The genotype frequency of BIN1 rs6733839 showed minimal association with MCI in the recessive model (P = 0.03). The genotype frequency of rs7561528 showed minimal association with MCI in the codominant, dominant, overdominant, and log-additive model (P < 0.05). The genotype frequencies of StAR-related lipid transfer domain 6 (STARD6) rs10164112 showed nominal association with MCI in the codominant, dominant, and log-additive model (P < 0.05). Unfortunately, the significant differences did not survive Benjamini-Hochberg false discovery rate correction (adjusted P > 0.05). The patients with SPI1 rs1057233 may be the protective factor of MCI (OR = 0.733, 95%CI 0.625-0.859, P < 0.001), and patients with APOE rs10164112 may be a risk factor for MCI (OR = 1.323, 95%CI 1.023-1.711, P = 0.033). CONCLUSIONS The polymorphisms of rs7561528, rs6733839 loci in the BIN1 gene, and rs1057233 loci in the SPI1 gene may be associated with the MCI in Chinese Han population. APOE gene was the risk factor of MCI, but further verification in a large sample population is still needed.
Collapse
Affiliation(s)
- Yelei Zhang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China
- The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325007, China
| | - Xiaoyue Li
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China
| | - Yu Hu
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China
| | - Hongwei Yuan
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151, China
| | - Xiaodong Wu
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China
| | - Yating Yang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China
| | - Tongtong Zhao
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China
| | - Ke Hu
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151, China
| | - Zhiqiang Wang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151, China
| | - Guoqiang Wang
- Department of Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, 214151, China
| | - Kai Zhang
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China.
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China.
| | - Huanzhong Liu
- Department of Psychiatry, Chaohu Hospital, Anhui Medical University, 64 North Chaohu Road, Hefei, 238000, China.
- Anhui Psychiatric Center, Anhui Medical University, Hefei, 238000, China.
| |
Collapse
|
230
|
Leyden GM, Shapland CY, Davey Smith G, Sanderson E, Greenwood MP, Murphy D, Richardson TG. Harnessing tissue-specific genetic variation to dissect putative causal pathways between body mass index and cardiometabolic phenotypes. Am J Hum Genet 2022; 109:240-252. [PMID: 35090585 PMCID: PMC8874216 DOI: 10.1016/j.ajhg.2021.12.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Body mass index (BMI) is a complex disease risk factor known to be influenced by genes acting via both metabolic pathways and appetite regulation. In this study, we aimed to gain insight into the phenotypic consequences of BMI-associated genetic variants, which may be mediated by their expression in different tissues. First, we harnessed meta-analyzed gene expression datasets derived from subcutaneous adipose (n = 1257) and brain (n = 1194) tissue to identify 86 and 140 loci, respectively, which provided evidence of genetic colocalization with BMI. These two sets of tissue-partitioned loci had differential effects with respect to waist-to-hip ratio, suggesting that the way they influence fat distribution might vary despite their having very similar average magnitudes of effect on BMI itself (adipose = 0.0148 and brain = 0.0149 standard deviation change in BMI per effect allele). For instance, BMI-associated variants colocalized with TBX15 expression in adipose tissue (posterior probability [PPA] = 0.97), but not when we used TBX15 expression data derived from brain tissue (PPA = 0.04) This gene putatively influences BMI via its role in skeletal development. Conversely, there were loci where BMI-associated variants provided evidence of colocalization with gene expression in brain tissue (e.g., NEGR1, PPA = 0.93), but not when we used data derived from adipose tissue, suggesting that these genes might be more likely to influence BMI via energy balance. Leveraging these tissue-partitioned variant sets through a multivariable Mendelian randomization framework provided strong evidence that the brain-tissue-derived variants are predominantly responsible for driving the genetically predicted effects of BMI on cardiovascular-disease endpoints (e.g., coronary artery disease: odds ratio = 1.05, 95% confidence interval = 1.04-1.07, p = 4.67 × 10-14). In contrast, our analyses suggested that the adipose tissue variants might predominantly be responsible for the underlying relationship between BMI and measures of cardiac function, such as left ventricular stroke volume (beta = 0.21, 95% confidence interval = 0.09-0.32, p = 6.43 × 10-4).
Collapse
Affiliation(s)
- Genevieve M Leyden
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom; Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, United Kingdom.
| | - Chin Yang Shapland
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Michael P Greenwood
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - David Murphy
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom; Novo Nordisk Research Centre, Headington, Oxford, OX3 7FZ, United Kingdom.
| |
Collapse
|
231
|
Kwon SJ, Hong KW, Choi S, Hong JS, Kim JW, Kim JW, Lee HJ, Jang HB, Yum KS. Association of 3-hydroxy-3-methylglutaryl-coenzyme A reductase gene polymorphism with obesity and lipid metabolism in children and adolescents with autism spectrum disorder. Metab Brain Dis 2022; 37:319-328. [PMID: 34806144 DOI: 10.1007/s11011-021-00877-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/14/2021] [Indexed: 11/25/2022]
Abstract
The prevalence of obesity among children and adolescents with autism spectrum disorder (ASD) is higher than that among typically developing children and adolescents. However, very few studies have explored the genetic factors associated with obesity in children and adolescents with ASD. Thus, the aim of this study was to examine the associations between 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) gene polymorphisms and obesity among children and adolescents with ASD. The study participants consisted of 33 children and adolescents with ASD and 271 age- and sex-matched typically developing controls. We compared the metabolic traits (body mass index, blood pressure, triglyceride, high-density lipoprotein, and fasting glucose levels) between the ASD and control group. Furthermore, we assessed the genotypes of rs12654264 in the HMGCR gene within the participants with ASD, and compared metabolic traits among the different allele subgroups. The mean body mass index (BMI) and triglyceride level of the ASD group were significantly higher than those of the control group. Within the ASD group, the triglyceride level of participants with rs12654264-T alleles was significantly higher than that of participants with A-alleles. A pattern of increasing values in the BMI and fasting glucose was also observed in participants with T allele. This is the first study to show that obesity in children and adolescents with ASD is associated with the cholesterol synthesis pathway. Future studies are needed to further clarify the molecular mechanisms by which the HMGCR gene influences metabolic traits.
Collapse
Affiliation(s)
- Si Jin Kwon
- Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | - Silvia Choi
- Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Su Hong
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine/Center for Autism and Related Disorders, Kennedy Kreger Institute, Baltimore, MD, USA
| | - Jung Won Kim
- Department of Psychiatry and Behavioral Sciences, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ju Whi Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye-Ja Lee
- Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Han Byul Jang
- Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Chungcheongbuk-do, Republic of Korea
| | - Keun-Sang Yum
- Department of Family Medicine, Uijeongbu St. Mary's Hospital, The Catholic University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
232
|
Bond TA, Richmond RC, Karhunen V, Cuellar-Partida G, Borges MC, Zuber V, Couto Alves A, Mason D, Yang TC, Gunter MJ, Dehghan A, Tzoulaki I, Sebert S, Evans DM, Lewin AM, O'Reilly PF, Lawlor DA, Järvelin MR. Exploring the causal effect of maternal pregnancy adiposity on offspring adiposity: Mendelian randomisation using polygenic risk scores. BMC Med 2022; 20:34. [PMID: 35101027 PMCID: PMC8805234 DOI: 10.1186/s12916-021-02216-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/13/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Greater maternal adiposity before or during pregnancy is associated with greater offspring adiposity throughout childhood, but the extent to which this is due to causal intrauterine or periconceptional mechanisms remains unclear. Here, we use Mendelian randomisation (MR) with polygenic risk scores (PRS) to investigate whether associations between maternal pre-/early pregnancy body mass index (BMI) and offspring adiposity from birth to adolescence are causal. METHODS We undertook confounder adjusted multivariable (MV) regression and MR using mother-offspring pairs from two UK cohorts: Avon Longitudinal Study of Parents and Children (ALSPAC) and Born in Bradford (BiB). In ALSPAC and BiB, the outcomes were birthweight (BW; N = 9339) and BMI at age 1 and 4 years (N = 8659 to 7575). In ALSPAC only we investigated BMI at 10 and 15 years (N = 4476 to 4112) and dual-energy X-ray absorptiometry (DXA) determined fat mass index (FMI) from age 10-18 years (N = 2659 to 3855). We compared MR results from several PRS, calculated from maternal non-transmitted alleles at between 29 and 80,939 single nucleotide polymorphisms (SNPs). RESULTS MV and MR consistently showed a positive association between maternal BMI and BW, supporting a moderate causal effect. For adiposity at most older ages, although MV estimates indicated a strong positive association, MR estimates did not support a causal effect. For the PRS with few SNPs, MR estimates were statistically consistent with the null, but had wide confidence intervals so were often also statistically consistent with the MV estimates. In contrast, the largest PRS yielded MR estimates with narrower confidence intervals, providing strong evidence that the true causal effect on adolescent adiposity is smaller than the MV estimates (Pdifference = 0.001 for 15-year BMI). This suggests that the MV estimates are affected by residual confounding, therefore do not provide an accurate indication of the causal effect size. CONCLUSIONS Our results suggest that higher maternal pre-/early-pregnancy BMI is not a key driver of higher adiposity in the next generation. Thus, they support interventions that target the whole population for reducing overweight and obesity, rather than a specific focus on women of reproductive age.
Collapse
Affiliation(s)
- Tom A Bond
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK.
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ville Karhunen
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Center for Life-course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Research Unit of Mathematical Sciences, University of Oulu, Oulu, Finland
| | - Gabriel Cuellar-Partida
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia
- 23andMe, Inc., Sunnyvale, CA, USA
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Verena Zuber
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Dan Mason
- Born in Bradford, Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Tiffany C Yang
- Born in Bradford, Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Marc J Gunter
- Section of Nutrition and Metabolism, IARC, Lyon, France
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Sylvain Sebert
- Center for Life-course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - David M Evans
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Alex M Lewin
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Paul F O'Reilly
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Marjo-Riitta Järvelin
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Center for Life-course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| |
Collapse
|
233
|
Werneck AO, Silva DRPD, Silva ECMD, Collings P, Ohara D, Fernandes RA, Barbosa DS, Ronque ERV, Sardinha LB, Cyrino ES. Association of parents’ physical activity and weight status with obesity and metabolic risk of their offspring. CIENCIA & SAUDE COLETIVA 2022; 27:783-792. [DOI: 10.1590/1413-81232022272.06182021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/29/2021] [Indexed: 11/21/2022] Open
Abstract
Abstract Our aim was to analyze the joint association of parental characteristics and offspring obesity indicators with metabolic risk in adolescents. A cross-sectional study was carried out with 972 adolescents and their parents. We observed that overweight adolescents who have a normal weight mother show lower metabolic risk in comparison with their counterparts with overweight mothers. In conclusion, mother’s weight status moderates the relationship between offspring’ obesity indicators and metabolic risk in adolescents.
Collapse
|
234
|
Anguita-Ruiz A, Zarza-Rebollo JA, Pérez-Gutiérrez AM, Molina E, Gutiérrez B, Bellón JÁ, Moreno-Peral P, Conejo-Cerón S, Aiarzagüena JM, Ballesta-Rodríguez MI, Fernández A, Fernández-Alonso C, Martín-Pérez C, Montón-Franco C, Rodríguez-Bayón A, Torres-Martos Á, López-Isac E, Cervilla J, Rivera M. Body mass index interacts with a genetic-risk score for depression increasing the risk of the disease in high-susceptibility individuals. Transl Psychiatry 2022; 12:30. [PMID: 35075110 PMCID: PMC8786870 DOI: 10.1038/s41398-022-01783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 11/24/2021] [Accepted: 01/04/2022] [Indexed: 11/22/2022] Open
Abstract
Depression is strongly associated with obesity among other chronic physical diseases. The latest mega- and meta-analysis of genome-wide association studies have identified multiple risk loci robustly associated with depression. In this study, we aimed to investigate whether a genetic-risk score (GRS) combining multiple depression risk single nucleotide polymorphisms (SNPs) might have utility in the prediction of this disorder in individuals with obesity. A total of 30 depression-associated SNPs were included in a GRS to predict the risk of depression in a large case-control sample from the Spanish PredictD-CCRT study, a national multicentre, randomized controlled trial, which included 104 cases of depression and 1546 controls. An unweighted GRS was calculated as a summation of the number of risk alleles for depression and incorporated into several logistic regression models with depression status as the main outcome. Constructed models were trained and evaluated in the whole recruited sample. Non-genetic-risk factors were combined with the GRS in several ways across the five predictive models in order to improve predictive ability. An enrichment functional analysis was finally conducted with the aim of providing a general understanding of the biological pathways mapped by analyzed SNPs. We found that an unweighted GRS based on 30 risk loci was significantly associated with a higher risk of depression. Although the GRS itself explained a small amount of variance of depression, we found a significant improvement in the prediction of depression after including some non-genetic-risk factors into the models. The highest predictive ability for depression was achieved when the model included an interaction term between the GRS and the body mass index (BMI), apart from the inclusion of classical demographic information as marginal terms (AUC = 0.71, 95% CI = [0.65, 0.76]). Functional analyses on the 30 SNPs composing the GRS revealed an over-representation of the mapped genes in signaling pathways involved in processes such as extracellular remodeling, proinflammatory regulatory mechanisms, and circadian rhythm alterations. Although the GRS on its own explained a small amount of variance of depression, a significant novel feature of this study is that including non-genetic-risk factors such as BMI together with a GRS came close to the conventional threshold for clinical utility used in ROC analysis and improves the prediction of depression. In this study, the highest predictive ability was achieved by the model combining the GRS and the BMI under an interaction term. Particularly, BMI was identified as a trigger-like risk factor for depression acting in a concerted way with the GRS component. This is an interesting finding since it suggests the existence of a risk overlap between both diseases, and the need for individual depression genetics-risk evaluation in subjects with obesity. This research has therefore potential clinical implications and set the basis for future research directions in exploring the link between depression and obesity-associated disorders. While it is likely that future genome-wide studies with large samples will detect novel genetic variants associated with depression, it seems clear that a combination of genetics and non-genetic information (such is the case of obesity status and other depression comorbidities) will still be needed for the optimization prediction of depression in high-susceptibility individuals.
Collapse
Affiliation(s)
- Augusto Anguita-Ruiz
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.4489.10000000121678994Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.413448.e0000 0000 9314 1427CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Juan Antonio Zarza-Rebollo
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain. .,Institute of Neurosciences 'Federico Olóriz', Biomedical Research Center (CIBM), University of Granada, Granada, Spain.
| | - Ana M Pérez-Gutiérrez
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Esther Molina
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.4489.10000000121678994Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
| | - Blanca Gutiérrez
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.4489.10000000121678994Department of Psychiatry, Faculty of Medicine, University of Granada, Granada, Spain
| | - Juan Ángel Bellón
- grid.452525.1Primary Care District of Málaga-Guadalhorce, Biomedical Research Institute of Málaga (IBIMA), Primary Care Prevention and Health Promotion Network (redIAPP), Málaga, Spain ,grid.10215.370000 0001 2298 7828Department of Public Health and Psychiatry, Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Patricia Moreno-Peral
- grid.452525.1Primary Care District of Málaga-Guadalhorce, Biomedical Research Institute of Málaga (IBIMA), Primary Care Prevention and Health Promotion Network (redIAPP), Málaga, Spain
| | - Sonia Conejo-Cerón
- grid.452525.1Primary Care District of Málaga-Guadalhorce, Biomedical Research Institute of Málaga (IBIMA), Primary Care Prevention and Health Promotion Network (redIAPP), Málaga, Spain
| | | | | | - Anna Fernández
- grid.428876.7Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona, Spain ,grid.466571.70000 0004 1756 6246CIBERESP, Centro de Investigacion Biomedica en Red de Epidemiologia y Salud Publica, Madrid, Spain
| | | | - Carlos Martín-Pérez
- grid.418355.eMarquesado Health Centre, Servicio Andaluz de Salud, Granada, Spain
| | - Carmen Montón-Franco
- grid.488737.70000000463436020Casablanca Health Centre, Aragonese Institute of Health Sciences, IIS Aragón, Zaragoza, Spain ,grid.11205.370000 0001 2152 8769Department of Medicine and Psychiatry, University of Zaragoza, Zaragoza, Spain
| | | | - Álvaro Torres-Martos
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Elena López-Isac
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Jorge Cervilla
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.4489.10000000121678994Department of Psychiatry, Faculty of Medicine, University of Granada, Granada, Spain
| | - Margarita Rivera
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
235
|
Wong HSC, Tsai SY, Chu HW, Lin MR, Lin GH, Tai YT, Shen CY, Chang WC. Genome-wide association study identifies genetic risk loci for adiposity in a Taiwanese population. PLoS Genet 2022; 18:e1009952. [PMID: 35051171 PMCID: PMC8853642 DOI: 10.1371/journal.pgen.1009952] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 02/17/2022] [Accepted: 11/16/2021] [Indexed: 01/22/2023] Open
Abstract
Overweight and obese are risk factors for various diseases. In Taiwan, the combined prevalence of overweight and obesity has increased dramatically. Here, we conducted a genome-wide association study (GWAS) on four adiposity traits, including body-mass index (BMI), body fat percentage (BF%), waist circumference (WC), and waist-hip ratio (WHR), using the data for more than 21,000 subjects in Taiwan Biobank. Associations were evaluated between 6,546,460 single-nucleotide polymorphisms (SNPs) and adiposity traits, yielding 13 genome-wide significant (GWS) adiposity-associated trait-loci pairs. A known gene, FTO, as well as two BF%-associated loci (GNPDA2-GABRG1 [4p12] and RNU6-2-PIAS1 [15q23]) were identified as pleiotropic effects. Moreover, RALGAPA1 was found as a specific genetic predisposing factor to high BMI in a Taiwanese population. Compared to other populations, a slightly lower heritability of the four adiposity traits was found in our cohort. Surprisingly, we uncovered the importance of neural pathways that might influence BF%, WC and WHR in the Taiwanese (East Asian) population. Additionally, a moderate genetic correlation between the WHR and BMI (γg = 0.52; p = 2.37×10−9) was detected, suggesting different genetic determinants exist for abdominal adiposity and overall adiposity. In conclusion, the obesity-related genetic loci identified here provide new insights into the genetic underpinnings of adiposity in the Taiwanese population.
Collapse
Affiliation(s)
- Henry Sung-Ching Wong
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Szu-Yi Tsai
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hou-Wei Chu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Gan-Hong Lin
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ting Tai
- Department of Anesthesiology, Taipei Municipal Wanfang Hospital, Taipei, Taiwan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- College of Public Health, China Medical University, Taichung, Taiwan
- * E-mail: (C-YS); (W-CC)
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- * E-mail: (C-YS); (W-CC)
| |
Collapse
|
236
|
Isgin-Atici K, Alathari BE, Turan-Demirci B, Sendur SN, Lay I, Ellahi B, Alikasifoglu M, Erbas T, Buyuktuncer Z, Vimaleswaran KS. Interaction between Dietary Fat Intake and Metabolic Genetic Risk Score on 25-Hydroxyvitamin D Concentrations in a Turkish Adult Population. Nutrients 2022; 14:382. [PMID: 35057563 PMCID: PMC8778439 DOI: 10.3390/nu14020382] [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: 10/10/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/19/2022] Open
Abstract
Previous studies have pointed out a link between vitamin D status and metabolic traits, however, consistent evidence has not been provided yet. This cross-sectional study has used a nutrigenetic approach to investigate the interaction between metabolic-genetic risk score (GRS) and dietary intake on serum 25-hydroxyvitamin D [25(OH)D] concentrations in 396 unrelated Turkish adults, aged 24-50 years. Serum 25(OH)D concentration was significantly lower in those with a metabolic-GRS ≥ 1 risk allele than those with a metabolic-GRS < 1 risk allele (p = 0.020). A significant interaction between metabolic-GRS and dietary fat intake (energy%) on serum 25(OH)D levels was identified (Pinteraction = 0.040). Participants carrying a metabolic-GRS ≥ 1 risk allele and consuming a high fat diet (≥38% of energy = 122.3 ± 52.51 g/day) had significantly lower serum 25(OH)D concentration (p = 0.006) in comparison to those consuming a low-fat diet (<38% of energy = 82.5 ± 37.36 g/d). In conclusion, our study suggests a novel interaction between metabolic-GRS and dietary fat intake on serum 25(OH)D level, which emphasises that following the current dietary fat intake recommendation (<35% total fat) could be important in reducing the prevalence of vitamin D deficiency in this Turkish population. Nevertheless, further larger studies are needed to verify this interaction, before implementing personalized dietary recommendations for the maintenance of optimal vitamin D status.
Collapse
Affiliation(s)
- Kubra Isgin-Atici
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06230, Turkey; (K.I.-A.); (B.T.-D.)
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Amasya University, Amasya 05000, Turkey
| | - Buthaina E. Alathari
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK;
- Department of Food Science and Nutrition, Faculty of Health Sciences, The Public Authority for Applied Education and Training, AlFaiha 72853, Kuwait
| | - Busra Turan-Demirci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06230, Turkey; (K.I.-A.); (B.T.-D.)
| | - Suleyman Nahit Sendur
- Department of Endocrinology and Metabolism, School of Medicine, Hacettepe University, Ankara 06230, Turkey; (S.N.S.); (T.E.)
| | - Incilay Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey;
- Clinical Pathology Laboratory, Hacettepe University Hospitals, Ankara 06230, Turkey
| | - Basma Ellahi
- Faculty of Health and Social Care, University of Chester, Chester CH1 4DS, UK;
| | - Mehmet Alikasifoglu
- Department of Medical Genetics, School of Medicine, Hacettepe University, Ankara 06230, Turkey;
- Genetics Diagnostic Centre, DAMAGEN, Ankara 06230, Turkey
| | - Tomris Erbas
- Department of Endocrinology and Metabolism, School of Medicine, Hacettepe University, Ankara 06230, Turkey; (S.N.S.); (T.E.)
| | - Zehra Buyuktuncer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06230, Turkey; (K.I.-A.); (B.T.-D.)
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK;
- Institute for Food, Nutrition, and Health, University of Reading, Reading RG6 6AH, UK
| |
Collapse
|
237
|
Ding R, Zhuang Z, Qiu Y, Ruan D, Wu J, Ye J, Cao L, Zhou S, Zheng E, Huang W, Wu Z, Yang J. Identify known and novel candidate genes associated with backfat thickness in Duroc pigs by large-scale genome-wide association analysis. J Anim Sci 2022; 100:6509022. [PMID: 35034121 PMCID: PMC8867564 DOI: 10.1093/jas/skac012] [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: 06/28/2021] [Accepted: 01/14/2022] [Indexed: 01/18/2023] Open
Abstract
Backfat thickness (BFT) is complex and economically important traits in the pig industry, since it reflects fat deposition and can be used to measure the carcass lean meat percentage in pigs. In this study, all 6,550 pigs were genotyped using the Geneseek Porcine 50K SNP Chip to identify SNPs related to BFT and to search for candidate genes through genome-wide association analysis in two Duroc populations. In total, 80 SNPs, including 39 significant and 41 suggestive SNPs, and 6 QTLs were identified significantly associated with the BFT. In addition, 9 candidate genes, including a proven major gene MC4R, 3 important candidate genes (RYR1, HMGA1, and NUDT3) which were previously described as related to BFT, and 5 novel candidate genes (SIRT2, NKAIN2, AMH, SORCS1, and SORCS3) were found based on their potential functional roles in BFT. The functions of candidate genes and gene set enrichment analysis indicate that most important pathways are related to energy homeostasis and adipogenesis. Finally, our data suggest that most of the candidate genes can be directly used for genetic improvement through molecular markers, except that the MC4R gene has an antagonistic effect on growth rate and carcass lean meat percentage in breeding. Our results will advance our understanding of the complex genetic architecture of BFT traits and laid the foundation for additional genetic studies to increase carcass lean meat percentage of pig through marker-assisted selection and/or genomic selection.
Collapse
Affiliation(s)
- Rongrong Ding
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China,Guangdong Wens Breeding Swine Technology Co., Ltd., Guangdong, 527400, P. R. China
| | - Zhanwei Zhuang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China
| | - Yibin Qiu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China
| | - Donglin Ruan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China
| | - Jie Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China
| | - Jian Ye
- Guangdong Wens Breeding Swine Technology Co., Ltd., Guangdong, 527400, P. R. China
| | - Lu Cao
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China
| | - Shenping Zhou
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China
| | - Enqin Zheng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, P. R. China
| | - Wen Huang
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Zhenfang Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China,Guangdong Wens Breeding Swine Technology Co., Ltd., Guangdong, 527400, P. R. China,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, P. R. China
| | - Jie Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, P. R. China,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, P. R. China,Corresponding authors:
| |
Collapse
|
238
|
Ershova AI, Ivanova AA, Kiseleva AV, Sotnikova EA, Meshkov AN, Drapkina OM. From biobanking to personalized prevention of obesity, diabetes and metabolic syndrome. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022. [DOI: 10.15829/1728-8800-2021-3123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The growing prevalence of metabolic disorders creates an increasing demand for novel approaches to their prevention and therapy. Novel genetic diagnostic technologies are developed every year, which makes it possible to identify people who are at the highest genetic risk of diabetes, non-alcoholic fatty liver disease, and metabolic syndrome. Early intervention strategies can be used to prevent metabolic disorders in this group of people. Genetic risk scores (GRSs) are a powerful tool to identify people with a high genetic risk. Millions of genetic variants are analyzed in genome-wide association studies in order to combine them into GRSs. It has become possible to store and process such huge amounts of data with the help of biobanks, where biological samples are stored according to international standards. Genetic studies include more and more people every year that increases the predictive power of GRSs. It has already been demonstrated that the use of GRSs makes future preventive measures more effective. In the near future, GRSs are likely to become part of clinical guidelines so that they can be widely used to identify people at high risk for metabolic syndrome and its components.
Collapse
Affiliation(s)
- A. I. Ershova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. A. Ivanova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. V. Kiseleva
- National Medical Research Center for Therapy and Preventive Medicine
| | - E. A. Sotnikova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. N. Meshkov
- National Medical Research Center for Therapy and Preventive Medicine; Pirogov Russian National Research Medical University
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
239
|
Arends D, Kärst S, Heise S, Korkuc P, Hesse D, Brockmann GA. Transmission distortion and genetic incompatibilities between alleles in a multigenerational mouse advanced intercross line. Genetics 2022; 220:iyab192. [PMID: 34791189 PMCID: PMC8733443 DOI: 10.1093/genetics/iyab192] [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: 09/10/2021] [Accepted: 10/15/2021] [Indexed: 11/23/2022] Open
Abstract
While direct additive and dominance effects on complex traits have been mapped repeatedly, additional genetic factors contributing to the heterogeneity of complex traits have been scarcely investigated. To assess genetic background effects, we investigated transmission ratio distortions (TRDs) of alleles from parent to offspring using an advanced intercross line (AIL) of an initial cross between the mouse inbred strains C57BL/6NCrl (B6N) and BFMI860-12 [Berlin Fat Mouse Inbred (BFMI)]. A total of 341 males of generation 28 and their respective 61 parents and 66 grandparents were genotyped using Mega Mouse Universal Genotyping Arrays. TRDs were investigated using allele transmission asymmetry tests, and pathway overrepresentation analysis was performed. Sequencing data were used to test for overrepresentation of nonsynonymous SNPs (nsSNPs) in TRD regions. Genetic incompatibilities were tested using the Bateson-Dobzhansky-Muller two-locus model. A total of 62 TRD regions were detected, many in close proximity to the telocentric centromere. TRD regions contained 44.5% more nsSNPs than randomly selected regions (182 vs 125.9 ± 17.0, P < 1 × 10-4). Testing for genetic incompatibilities between TRD regions identified 29 genome-wide significant incompatibilities between TRD regions [P(BF) < 0.05]. Pathway overrepresentation analysis of genes in TRD regions showed that DNA methylation, epigenetic regulation of RNA, and meiotic/meiosis regulation pathways were affected independent of the parental origin of the TRD. Paternal BFMI TRD regions showed overrepresentation in the small interfering RNA biogenesis and in the metabolism of lipids and lipoproteins. Maternal B6N TRD regions harbored genes involved in meiotic recombination, cell death, and apoptosis pathways. The analysis of genes in TRD regions suggests the potential distortion of protein-protein interactions influencing obesity and diabetic retinopathy as a result of disadvantageous combinations of allelic variants in Aass, Pgx6, and Nme8. Using an AIL significantly improves the resolution at which we can investigate TRD. Our analysis implicates distortion of protein-protein interactions as well as meiotic drive as the underlying mechanisms leading to the observed TRD in our AIL. Furthermore, genes with large amounts of nsSNPs located in TRD regions are more likely to be involved in pathways that are related to the phenotypic differences between the parental strains. Genes in these TRD regions provide new targets for investigating genetic adaptation, protein-protein interactions, and determinants of complex traits such as obesity.
Collapse
Affiliation(s)
- Danny Arends
- Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer Institute for Agricultural and Horticultural Sciences, Humboldt University Berlin, Berlin D-10115, Germany
| | - Stefan Kärst
- Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer Institute for Agricultural and Horticultural Sciences, Humboldt University Berlin, Berlin D-10115, Germany
| | - Sebastian Heise
- Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer Institute for Agricultural and Horticultural Sciences, Humboldt University Berlin, Berlin D-10115, Germany
| | - Paula Korkuc
- Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer Institute for Agricultural and Horticultural Sciences, Humboldt University Berlin, Berlin D-10115, Germany
| | - Deike Hesse
- Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer Institute for Agricultural and Horticultural Sciences, Humboldt University Berlin, Berlin D-10115, Germany
| | - Gudrun A Brockmann
- Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer Institute for Agricultural and Horticultural Sciences, Humboldt University Berlin, Berlin D-10115, Germany
| |
Collapse
|
240
|
Yang D, Wu X, Wang W, Zhou Y, Wang Z. Ciliary Type III Adenylyl Cyclase in the VMH Is Crucial for High-Fat Diet-Induced Obesity Mediated by Autophagy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102568. [PMID: 34783461 PMCID: PMC8787410 DOI: 10.1002/advs.202102568] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Neuronal primary cilia are crucial for body weight maintenance. Type III adenylyl cyclase (AC3) is abundantly enriched in neuronal cilia, and mice with global AC3 ablation are obese. However, whether AC3 regulates body weight through its ciliary expression and the mechanism underlying this potential regulation are not clear. In this study, humanized AC3 knock-in mice that are resistant to high-fat diet (HFD)-induced obesity are generated, and increases in the number and length of cilia in the ventromedial hypothalamus (VMH) are shown. It is demonstrated that mice with specifically knocked down ciliary AC3 expression in the VMH show pronounced HFD-induced obesity. In addition, in vitro and in vivo analyses of the VMH show that ciliary AC3 regulates autophagy by binding an autophagy-related gene, gamma-aminobutyric acid A receptor-associated protein (GABARAP). Mice with GABARAP knockdown in the VMH exhibit exacerbated HFD-induced obesity. Overall, the findings may reveal a potential mechanism by which ciliary AC3 expression regulates body weight in the mouse VMH.
Collapse
Affiliation(s)
- Dong Yang
- College of Life ScienceInstitute of Life Science and Green DevelopmentHebei UniversityBaodingHebei071002China
| | - Xiangbo Wu
- College of Life ScienceInstitute of Life Science and Green DevelopmentHebei UniversityBaodingHebei071002China
| | - Weina Wang
- College of Life ScienceInstitute of Life Science and Green DevelopmentHebei UniversityBaodingHebei071002China
| | - Yanfen Zhou
- College of Life ScienceInstitute of Life Science and Green DevelopmentHebei UniversityBaodingHebei071002China
| | - Zhenshan Wang
- College of Life ScienceInstitute of Life Science and Green DevelopmentHebei UniversityBaodingHebei071002China
| |
Collapse
|
241
|
Abstract
The prevalence of obesity has tripled over the past four decades, imposing an enormous burden on people's health. Polygenic (or common) obesity and rare, severe, early-onset monogenic obesity are often polarized as distinct diseases. However, gene discovery studies for both forms of obesity show that they have shared genetic and biological underpinnings, pointing to a key role for the brain in the control of body weight. Genome-wide association studies (GWAS) with increasing sample sizes and advances in sequencing technology are the main drivers behind a recent flurry of new discoveries. However, it is the post-GWAS, cross-disciplinary collaborations, which combine new omics technologies and analytical approaches, that have started to facilitate translation of genetic loci into meaningful biology and new avenues for treatment.
Collapse
Affiliation(s)
- Ruth J. F. Loos
- grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark ,grid.59734.3c0000 0001 0670 2351Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Giles S. H. Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| |
Collapse
|
242
|
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
|
243
|
Abstract
PURPOSE OF REVIEW At elevated levels, the essential element manganese (Mn) is neurotoxic and increasing evidence indicates that environmental Mn exposure early in life negatively affects neurodevelopment. In this review, we describe how underlying genetics may confer susceptibility to elevated Mn concentrations and how the epigenetic effects of Mn may explain the association between Mn exposure early in life and its toxic effects later in life. RECENT FINDINGS Common polymorphisms in the Mn transporter genes SLC30A10 and SLC39A8 seem to have a large impact on intracellular Mn levels and, in turn, neurotoxicity. Genetic variation in iron regulatory genes may to lesser extent also influence Mn levels and toxicity. Recent studies on Mn and epigenetic mechanisms indicate that Mn-related changes in DNA methylation occur early in life. One human and two animal studies found persistent changes from in utero exposure to Mn but whether these changes have functional effects remains unknown. Genetics seems to play a major role in susceptibility to Mn toxicity and should therefore be considered in risk assessment. Mn appears to interfere with epigenetic processes, potentially leading to persistent changes in developmental programming, which warrants further study.
Collapse
|
244
|
Michałowska J, Miller-Kasprzak E, Seraszek-Jaros A, Mostowska A, Bogdański P. Association of GLP1R variants rs2268641 and rs6923761 with obesity and other metabolic parameters in a Polish cohort. Front Endocrinol (Lausanne) 2022; 13:1000185. [PMID: 36339410 PMCID: PMC9626533 DOI: 10.3389/fendo.2022.1000185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Obesity is a complex disease associated with excessive fat accumulation and numerous metabolic complications. So far, many factors leading to the development of this disorder have been identified, including genetic susceptibility. Various studies linked GLP1R variants with anthropometric and metabolic parameters, suggesting the role of the variation in this gene in metabolic health. OBJECTIVE The aim of this study is to investigate the association of two single nucleotide variants of GLP1R gene, rs2268641 and rs6923761, with excessive weight, metabolic syndrome, anthropometric measurements and selected metabolic parameters. METHODS Normal-weight subjects (n= 340, control group) and subjects with excessive body mass (n = 600, study group) participated in this study. For all participants, anthropometric measurements and metabolic parameters were collected, and genotyping of the two single nucleotide variants of GLP1R gene, rs2268641 and rs6923761, was performed using the high-resolution melting curve analysis. RESULTS Significant differences in the genotype distribution of rs2268641 were found, where homozygous TT genotype was significantly less frequent in the study group with excessive body mass (OR=0.66; p=0.0298). For rs6923761, A allele and homozygous AA genotype were significantly more frequent in the study group with excessive weight than in the control group (OR=1.27; p=0.0239 and OR=1.69; p=0.0205, respectively). The association of studied variants with metabolic parameters was found for rs6923761. For this variant, AA carriers had higher body mass in comparison to GG carriers (p=0.0246), and AA carriers had higher glucose concentration in comparison to AG carriers (p=0.0498). We did not find an association of rs2268641 and rs6923761 with metabolic syndrome. CONCLUSION In our study, AA carriers of rs6923761 had higher risk of excessive body mass, whereas TT carriers of rs2268641 had lower risk of being overweight. Moreover, homozygous carriers of the minor allele of rs6923761 had higher glucose concentration in comparison to heterozygous subjects. None of the studied variants were associated with metabolic syndrome in the studied population.
Collapse
Affiliation(s)
- Joanna Michałowska
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
- *Correspondence: Joanna Michałowska,
| | - Ewa Miller-Kasprzak
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Agnieszka Seraszek-Jaros
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, Poznan, Poland
| | - Adrianna Mostowska
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
245
|
Ceja-Galicia Z, Calderón-DuPont D, Daniel A, Chiu LM, Díaz-Villaseñor A. Leptin and adiponectin synthesis and secretion in mature 3T3-L1 adipocytes are differentially down-regulated by arsenic and palmitic acid exposure throughout different stages of adipogenesis. Life Sci 2021; 291:120262. [PMID: 34968464 DOI: 10.1016/j.lfs.2021.120262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022]
Abstract
AIMS Arsenic is a risk factor for type 2 diabetes and cardiovascular disease. However, little is known about arsenic effects over adipocyte endocrine functionality, particularly for leptin and adiponectin, and about its interaction with dietary components, which are the main environmental regulators of adipose tissue functionality. The aim of this work was to evaluate leptin and adiponectin in mature 3T3-L1 adipocytes exposed to palmitate (simulating excess fat intake), arsenite, or both throughout two different stages of adipogenesis. MATERIAL AND METHODS 3T3-L1 adipocytes were exposed starting from the beginning of its differentiation process during 11 d or once adipocytes were mature for 72 h. Adipokines secretion was evaluated by ELISA, intracellular protein levels and secreted adiponectin multimers by Western blot and mRNA abundance by qPCR. KEY FINDINGS Leptin and adiponectin secretion decreased by arsenite alone or in combination with palmitate due to reduced gene and protein expression of both adipokines. However, leptin was impaired more at the transcriptional level, whereas affections to adiponectin were more relevant at the intracellular protein amount level with changes in the multimers proportion. The gene expression of several of their transcription factors was altered. Additionally, the magnitude of the effects depends on the adipocyte cell stage at which exposure began; adiponectin was more affected when exposure started from differentiation and leptin once adipocytes were mature. SIGNIFICANCE These results in an in vivo model could be translated into less satiety and reduced insulin sensitivity.
Collapse
Affiliation(s)
- Zeltzin Ceja-Galicia
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Maestría en Ciencias de la Producción y de la Salud Animal, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Diana Calderón-DuPont
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Alberto Daniel
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico; Maestría en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Luz María Chiu
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico
| | - Andrea Díaz-Villaseñor
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 045010, Mexico.
| |
Collapse
|
246
|
Raymond PW, Velie BD, Wade CM. Forensic DNA phenotyping: Canis familiaris breed classification and skeletal phenotype prediction using functionally significant skeletal SNPs and indels. Anim Genet 2021; 53:247-263. [PMID: 34963196 DOI: 10.1111/age.13165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2021] [Accepted: 12/12/2021] [Indexed: 11/29/2022]
Abstract
This review highlights a novel application of breed identification and prediction of skeletal traits in forensic investigations using canine DNA evidence. Currently, genotyping methods used for canine breed classification involve the application of highly polymorphic short tandem repeats in addition to larger commercially available SNP arrays. Both applications face technical challenges. An additional approach to breed identification could be through genotyping SNPs and indels that characterise the array of skeletal differences displayed across domestic dog populations. Research has shown that a small number of genetic variants of large effect drive differences in skeletal phenotypes among domestic dog breeds. This feature makes functionally significant canine skeletal variants a cost-effective target for forensic investigators to classify individuals according to their breed. Further analysis of these skeletal variants would enable the prediction of external appearance. To date, functionally significant genes with genetic variants associated with differences in size, bulk, skull shape, ear shape, limb length, digit type, and tail morphology have been uncovered. Recommendations of a cost-effective genotyping method that can be readily designed and applied by forensic investigators have been given. Further advances to improve the field of canine skeletal forensic DNA phenotyping include the refinement of phenotyping methods, further biological validation of the skeletal genetic variants and establishing a publicly available database for storage of allele frequencies of the skeletal genetic variants in the wider domestic dog population.
Collapse
Affiliation(s)
- Patrick W Raymond
- School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Brandon D Velie
- School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Claire M Wade
- School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| |
Collapse
|
247
|
Fujishiro H, Kambe T. Manganese transport in mammals by zinc transporter family proteins, ZNT and ZIP. J Pharmacol Sci 2021; 148:125-133. [PMID: 34924116 DOI: 10.1016/j.jphs.2021.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Manganese (Mn) is an essential trace element required for various biological processes. However, excess Mn causes serious side effects in humans, including parkinsonism. Thus, elucidation of Mn homeostasis at the systemic, cellular, and molecular levels is important. Many metal transporters and channels can be involved in the transport and homeostasis of Mn, and an increasing body of evidence shows that several zinc (Zn) transporters belonging to the ZIP and ZNT families, specifically, ZNT10, ZIP8, and ZIP14, play pivotal roles in Mn metabolism. Mutations in the genes encoding these transporter proteins are associated with congenital disorders related to dysregulated Mn homeostasis in humans. Moreover, single nucleotide polymorphisms of ZIP8 are associated with multiple clinical phenotypes. In this review, we discuss the recent literature on the structural and biochemical features of ZNT10, ZIP8, and ZIP14, including transport mechanisms, regulation of expression, and pathophysiological functions. Because a disturbance in Mn homeostasis is closely associated with a variety of phenotypes and risk of human diseases, these transporters constitute a significant target for drug development. An understanding of the roles of these key transporters in Mn metabolism should provide new insights into pharmacological applications of their inhibitors and enhancers in human diseases.
Collapse
Affiliation(s)
- Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan.
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan.
| |
Collapse
|
248
|
Larison B, Pinho GM, Haghani A, Zoller JA, Li CZ, Finno CJ, Farrell C, Kaelin CB, Barsh GS, Wooding B, Robeck TR, Maddox D, Pellegrini M, Horvath S. Epigenetic models developed for plains zebras predict age in domestic horses and endangered equids. Commun Biol 2021; 4:1412. [PMID: 34921240 PMCID: PMC8683477 DOI: 10.1038/s42003-021-02935-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/02/2021] [Indexed: 01/09/2023] Open
Abstract
Effective conservation and management of threatened wildlife populations require an accurate assessment of age structure to estimate demographic trends and population viability. Epigenetic aging models are promising developments because they estimate individual age with high accuracy, accurately predict age in related species, and do not require invasive sampling or intensive long-term studies. Using blood and biopsy samples from known age plains zebras (Equus quagga), we model epigenetic aging using two approaches: the epigenetic clock (EC) and the epigenetic pacemaker (EPM). The plains zebra EC has the potential for broad application within the genus Equus given that five of the seven extant wild species of the genus are threatened. We test the EC's ability to predict age in sister taxa, including two endangered species and the more distantly related domestic horse, demonstrating high accuracy in all cases. By comparing chronological and estimated age in plains zebras, we investigate age acceleration as a proxy of health status. An interaction between chronological age and inbreeding is associated with age acceleration estimated by the EPM, suggesting a cumulative effect of inbreeding on biological aging throughout life.
Collapse
Affiliation(s)
- Brenda Larison
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, 90095, USA.
- Center for Tropical Research, Institute of the Environment and Sustainability, University of California, Los Angeles, CA, 90095, USA.
| | - Gabriela M Pinho
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, 90095, USA
| | - Amin Haghani
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Joseph A Zoller
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Caesar Z Li
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Carrie J Finno
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Colin Farrell
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Christopher B Kaelin
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Gregory S Barsh
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Bernard Wooding
- Quagga Project, Elandsberg Farms, Hermon, 7308, South Africa
| | - Todd R Robeck
- Zoological Operations, SeaWorld Parks and Entertainment, 7007 SeaWorld Drive, Orlando, FL, USA
| | - Dewey Maddox
- White Oak Conservation, 581705 White Oak Road, Yulee, FL, 32097, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Steve Horvath
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA.
- Altos Labs, San Diego, CA, USA.
| |
Collapse
|
249
|
Ahamed SK, Barek MA, Roy UK, Kouser M, Reza MS, Mannan AB, Alam MA, Uddin SMN. A review on association and correlation of genetic variants with eating disorders and obesity. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Now, eating disorders and obesity and their correlations are danger signal in worldwide which is caused by multifactor and associated with significant mortality and morbidity.
Main body
Every aspect of a patient’s life is influenced by eating disorders and obesity and their correlations. Due to frequent seeing of obese patients, eating disorders have been included in the review as they can sometimes be associated with obesity. However, it should be noted that most patients having eating disorder are at risk to be obese or overweight. This research explores the risk factors for the two disorders, as well as the assessment of medical complications and treatment recommendations for the disorders. In these two disorders, there is also a correlation. The essential consideration is that eating disorders are impulse-control disorders which are similar to addictive behaviors in some aspects. So it is a crying need to treat a patient with obesity and eating disorders simultaneously to ensure success. Genome-wide association studies (GWASs) have increased our knowledge of the pathophysiology of eating disorders (EDs) and obesity and their correlation.
Conclusion
This review enlightens on the summary of eating disorder, obesity, genotypic traits, molecular relations, interaction, correlation, and effect of eating disorder and obesity which outline potential future directions and clinical implications for patients with EDs and obesity.
Collapse
|
250
|
Payer LM, Steranka JP, Kryatova MS, Grillo G, Lupien M, Rocha PP, Burns KH. Alu insertion variants alter gene transcript levels. Genome Res 2021; 31:2236-2248. [PMID: 34799402 PMCID: PMC8647820 DOI: 10.1101/gr.261305.120] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/23/2021] [Indexed: 12/23/2022]
Abstract
Alu are high copy number interspersed repeats that have accumulated near genes during primate and human evolution. They are a pervasive source of structural variation in modern humans. Impacts that Alu insertions may have on gene expression are not well understood, although some have been associated with expression quantitative trait loci (eQTLs). Here, we directly test regulatory effects of polymorphic Alu insertions in isolation of other variants on the same haplotype. To screen insertion variants for those with such effects, we used ectopic luciferase reporter assays and evaluated 110 Alu insertion variants, including more than 40 with a potential role in disease risk. We observed a continuum of effects with significant outliers that up- or down-regulate luciferase activity. Using a series of reporter constructs, which included genomic context surrounding the Alu, we can distinguish between instances in which the Alu disrupts another regulator and those in which the Alu introduces new regulatory sequence. We next focused on three polymorphic Alu loci associated with breast cancer that display significant effects in the reporter assay. We used CRISPR to modify the endogenous sequences, establishing cell lines varying in the Alu genotype. Our findings indicate that Alu genotype can alter expression of genes implicated in cancer risk, including PTHLH, RANBP9, and MYC These data show that commonly occurring polymorphic Alu elements can alter transcript levels and potentially contribute to disease risk.
Collapse
Affiliation(s)
- Lindsay M Payer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jared P Steranka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Maria S Kryatova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Giacomo Grillo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Pedro P Rocha
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland 20892-4340, USA
- National Cancer Institute, NIH, Bethesda, Maryland 20892, USA
| | - Kathleen H Burns
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- McKusick-Nathans Institute of Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|