101
|
John M, Lencz T. Potential application of elastic nets for shared polygenicity detection with adapted threshold selection. Int J Biostat 2023; 19:417-438. [PMID: 36327464 PMCID: PMC10154439 DOI: 10.1515/ijb-2020-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Current research suggests that hundreds to thousands of single nucleotide polymorphisms (SNPs) with small to modest effect sizes contribute to the genetic basis of many disorders, a phenomenon labeled as polygenicity. Additionally, many such disorders demonstrate polygenic overlap, in which risk alleles are shared at associated genetic loci. A simple strategy to detect polygenic overlap between two phenotypes is based on rank-ordering the univariate p-values from two genome-wide association studies (GWASs). Although high-dimensional variable selection strategies such as Lasso and elastic nets have been utilized in other GWAS analysis settings, they are yet to be utilized for detecting shared polygenicity. In this paper, we illustrate how elastic nets, with polygenic scores as the dependent variable and with appropriate adaptation in selecting the penalty parameter, may be utilized for detecting a subset of SNPs involved in shared polygenicity. We provide theory to better understand our approaches, and illustrate their utility using synthetic datasets. Results from extensive simulations are presented comparing the elastic net approaches with the rank ordering approach, in various scenarios. Results from simulations studies exhibit one of the elastic net approaches to be superior when the correlations among the SNPs are high. Finally, we apply the methods on two real datasets to illustrate further the capabilities, limitations and differences among the methods.
Collapse
Affiliation(s)
- Majnu John
- Institute of Behavioral Science, Feinstein Institutes of Medical Research, Manhasset, NY
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health System, Glen Oaks, NY
- Departments of Psychiatry and of Mathematics, Hofstra University, Hempstead, NY
| | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes of Medical Research, Manhasset, NY
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health System, Glen Oaks, NY
- Departments of Psychiatry and of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| |
Collapse
|
102
|
Hammad MM, Abu-Farha M, Hebbar P, Anoop E, Chandy B, Melhem M, Channanath A, Al-Mulla F, Thanaraj TA, Abubaker J. The miR-668 binding site variant rs1046322 on WFS1 is associated with obesity in Southeast Asians. Front Endocrinol (Lausanne) 2023; 14:1185956. [PMID: 37859980 PMCID: PMC10583568 DOI: 10.3389/fendo.2023.1185956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023] Open
Abstract
The Wolfram syndrome 1 gene (WFS1) is the main causative locus for Wolfram syndrome, an inherited condition characterized by childhood-onset diabetes mellitus, optic atrophy, and deafness. Global genome-wide association studies have listed at least 19 WFS1 variants that are associated with type 2 diabetes (T2D) and metabolic traits. It has been suggested that miRNA binding sites on WFS1 play a critical role in the regulation of the wolframin protein, and loss of WFS1 function may lead to the pathogenesis of diabetes. In the Hungarian population, it was observed that a 3' UTR variant from WFS1, namely rs1046322, influenced the affinity of miR-668 to WFS1 mRNA, and showed a strong association with T2D. In this study, we genotyped a large cohort of 2067 individuals of different ethnicities residing in Kuwait for the WFS1 rs1046322 polymorphism. The cohort included 362 Southeast Asians (SEA), 1045 Arabs, and 660 South Asians (SA). Upon performing genetic association tests, we observed significant associations between the rs1046322 SNP and obesity traits in the SEA population, but not in the Arab or SA populations. The associated traits in SEA cohort were body mass index, BMI (β=1.562, P-value=0.0035, Pemp=0.0072), waist circumference, WC (β=3.163, P-value=0.0197, Pemp=0.0388) and triglyceride, TGL (β=0.224, P-value=0.0340). The association with BMI remained statistically significant even after multiple testing correction. Among the SEA individuals, carriers of the effect allele at the SNP had significantly higher BMI [mean of 27.63 (3.6) Kg/m2], WC [mean of 89.9 (8.1) cm], and TGL levels [mean of 1.672 (0.8) mmol/l] than non-carriers of the effect allele. Our findings suggest a role for WFS1 in obesity, which is a risk factor for diabetes. The study also emphasizes the significant role the ethnic background may play in determining the effect of genetic variants on susceptibility to metabolic diseases.
Collapse
Affiliation(s)
- Maha M. Hammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait, Kuwait
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait, Kuwait
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Prashantha Hebbar
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Emil Anoop
- Special Service Facility Department, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Betty Chandy
- Special Service Facility Department, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Motasem Melhem
- Special Service Facility Department, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Arshad Channanath
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait, Kuwait
| | | | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait, Kuwait
| |
Collapse
|
103
|
Wang X, Dervishi L, Li W, Ayday E, Jiang X, Vaidya J. Privacy-preserving federated genome-wide association studies via dynamic sampling. Bioinformatics 2023; 39:btad639. [PMID: 37856329 PMCID: PMC10612407 DOI: 10.1093/bioinformatics/btad639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/15/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023] Open
Abstract
MOTIVATION Genome-wide association studies (GWAS) benefit from the increasing availability of genomic data and cross-institution collaborations. However, sharing data across institutional boundaries jeopardizes medical data confidentiality and patient privacy. While modern cryptographic techniques provide formal secure guarantees, the substantial communication and computational overheads hinder the practical application of large-scale collaborative GWAS. RESULTS This work introduces an efficient framework for conducting collaborative GWAS on distributed datasets, maintaining data privacy without compromising the accuracy of the results. We propose a novel two-step strategy aimed at reducing communication and computational overheads, and we employ iterative and sampling techniques to ensure accurate results. We instantiate our approach using logistic regression, a commonly used statistical method for identifying associations between genetic markers and the phenotype of interest. We evaluate our proposed methods using two real genomic datasets and demonstrate their robustness in the presence of between-study heterogeneity and skewed phenotype distributions using a variety of experimental settings. The empirical results show the efficiency and applicability of the proposed method and the promise for its application for large-scale collaborative GWAS. AVAILABILITY AND IMPLEMENTATION The source code and data are available at https://github.com/amioamo/TDS.
Collapse
Affiliation(s)
- Xinyue Wang
- Management Science and Information Systems Department, Rutgers University, New Brunswick, NJ 07102, United States
| | - Leonard Dervishi
- Department of Computer and Data Sciences, Cleveland, OH 44106, United States
| | - Wentao Li
- Department of Health Data Science and Artificial Intelligence, Houston, TX 77030, United States
| | - Erman Ayday
- Department of Computer and Data Sciences, Cleveland, OH 44106, United States
| | - Xiaoqian Jiang
- Department of Health Data Science and Artificial Intelligence, Houston, TX 77030, United States
| | - Jaideep Vaidya
- Management Science and Information Systems Department, Rutgers University, New Brunswick, NJ 07102, United States
| |
Collapse
|
104
|
Abstract
Obesity is a common complex trait that elevates the risk for various diseases, including type 2 diabetes and cardiovascular disease. A combination of environmental and genetic factors influences the pathogenesis of obesity. Advances in genomic technologies have driven the identification of multiple genetic loci associated with this disease, ranging from studying severe onset cases to investigating common multifactorial polygenic forms. Additionally, findings from epigenetic analyses of modifications to the genome that do not involve changes to the underlying DNA sequence have emerged as key signatures in the development of obesity. Such modifications can mediate the effects of environmental factors, including diet and lifestyle, on gene expression and clinical presentation. This review outlines what is known about the genetic and epigenetic contributors to obesity susceptibility, along with the albeit limited therapeutic options currently available. Furthermore, we delineate the potential mechanisms of actions through which epigenetic changes can mediate environmental influences and the related opportunities they present for future interventions in the management of obesity.
Collapse
Affiliation(s)
- Khanh Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Diabetes and Endocrinology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
105
|
Mocci E, Ward K, Perry JA, Starkweather A, Stone LS, Schabrun SM, Renn C, Dorsey SG, Ament SA. Genome wide association joint analysis reveals 99 risk loci for pain susceptibility and pleiotropic relationships with psychiatric, metabolic, and immunological traits. PLoS Genet 2023; 19:e1010977. [PMID: 37844115 PMCID: PMC10602383 DOI: 10.1371/journal.pgen.1010977] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 10/26/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
Chronic pain is at epidemic proportions in the United States, represents a significant burden on our public health system, and is coincident with a growing opioid crisis. While numerous genome-wide association studies have been reported for specific pain-related traits, many of these studies were underpowered, and the genetic relationship among these traits remains poorly understood. Here, we conducted a joint analysis of genome-wide association study summary statistics from seventeen pain susceptibility traits in the UK Biobank. This analysis revealed 99 genome-wide significant risk loci, 65 of which overlap loci identified in earlier studies. The remaining 34 loci are novel. We applied leave-one-trait-out meta-analyses to evaluate the influence of each trait on the joint analysis, which suggested that loci fall into four categories: loci associated with nearly all pain-related traits; loci primarily associated with a single trait; loci associated with multiple forms of skeletomuscular pain; and loci associated with headache-related pain. Overall, 664 genes were mapped to the 99 loci by genomic proximity, eQTLs, and chromatin interaction and ~15% of these genes showed differential expression in individuals with acute or chronic pain compared to healthy controls. Risk loci were enriched for genes involved in neurological and inflammatory pathways. Genetic correlation and two-sample Mendelian randomization indicated that psychiatric, metabolic, and immunological traits mediate some of these effects.
Collapse
Affiliation(s)
- Evelina Mocci
- Department of Pain & Translational Symptom Science, University of Maryland School of Nursing, Baltimore, Maryland, United States of America
- Center to Advance Chronic Pain Research (CACPR), University of Maryland Baltimore, Baltimore, Maryland, United States of America
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Kathryn Ward
- Department of Pain & Translational Symptom Science, University of Maryland School of Nursing, Baltimore, Maryland, United States of America
| | - James A. Perry
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Angela Starkweather
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, Florida, United States of America
| | - Laura S. Stone
- Department of Anesthesiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Siobhan M. Schabrun
- School of Physical Therapy, University of Western Ontario, London, Ontario, Canada
| | - Cynthia Renn
- Department of Pain & Translational Symptom Science, University of Maryland School of Nursing, Baltimore, Maryland, United States of America
- Center to Advance Chronic Pain Research (CACPR), University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Susan G. Dorsey
- Department of Pain & Translational Symptom Science, University of Maryland School of Nursing, Baltimore, Maryland, United States of America
- Center to Advance Chronic Pain Research (CACPR), University of Maryland Baltimore, Baltimore, Maryland, United States of America
| | - Seth A. Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
106
|
Jullian Fabres P, Lee SH. Phenotypic variance partitioning by transcriptomic gene expression levels and environmental variables for anthropometric traits using GTEx data. Genet Epidemiol 2023; 47:465-474. [PMID: 37318147 DOI: 10.1002/gepi.22531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/03/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
Phenotypic variation in human is the results of genetic variation and environmental influences. Understanding the contribution of genetic and environmental components to phenotypic variation is of great interest. The variance explained by genome-wide single nucleotide polymorphisms (SNPs) typically represents a small proportion of the phenotypic variance for complex traits, which may be because the genome is only a part of the whole biological process to shape the phenotypes. In this study, we propose to partition the phenotypic variance of three anthropometric traits, using gene expression levels and environmental variables from GTEx data. We use the gene expression of four tissues that are deemed relevant for the anthropometric traits (two adipose tissues, skeletal muscle tissue and blood tissue). Additionally, we estimate the transcriptome-environment correlation that partly underlies the phenotypes of the anthropometric traits. We found that genetic factors play a significant role in determining body mass index (BMI), with the proportion of phenotypic variance explained by gene expression levels of visceral adipose tissue being 0.68 (SE = 0.06). However, we also observed that environmental factors such as age, sex, ancestry, smoking status, and drinking alcohol status have a small but significant impact (0.005, SE = 0.001). Interestingly, we found a significant negative correlation between the transcriptomic and environmental effects on BMI (transcriptome-environment correlation = -0.54, SE = 0.14), suggesting an antagonistic relationship. This implies that individuals with lower genetic profiles may be more susceptible to the effects of environmental factors on BMI, while those with higher genetic profiles may be less susceptible. We also show that the estimated transcriptomic variance varies across tissues, e.g., the gene expression levels of whole blood tissue and environmental variables explain a lower proportion of BMI phenotypic variance (0.16, SE = 0.05 and 0.04, SE = 0.004 respectively). We observed a significant positive correlation between transcriptomic and environmental effects (1.21, SE = 0.23) for this tissue. In conclusion, phenotypic variance partitioning can be done using gene expression and environmental data even with a small sample size (n = 838 from GTEx data), which can provide insights into how the transcriptomic and environmental effects contribute to the phenotypes of the anthropometric traits.
Collapse
Affiliation(s)
- Pastor Jullian Fabres
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), University of South Australia, Adelaide, South Australia, Australia
| | - S Hong Lee
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
107
|
Sharma P, Halder A, Jain M, Tripathi M. Whole Exome Sequencing Reveals Rare Variants in Genes Associated with Metabolic Disorders in Women with PCOS. J Hum Reprod Sci 2023; 16:307-316. [PMID: 38322634 PMCID: PMC10841935 DOI: 10.4103/jhrs.jhrs_13_23] [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: 02/03/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 02/08/2024] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a complex genetic trait, the pathogenesis of which is governed by an interplay of genetic and epigenetic factors. However, the aetiology of PCOS is not fully understood. Aims The objective of this study was to investigate the genetic causes of PCOS by identifying rare variants in genes implicated in its pathophysiology. Settings and Design This was a hospital-based observational study. Materials and Methods We used whole-exome sequencing for 52 PCOS women to identify the rare variants in genes related to PCOS pathogenesis. Subsequently, we analysed these variants using in silico prediction software to determine their functional effects. We then assessed the relationship between these variants and the clinical outcomes of the patients. Statistical Analysis Used Student's t-test and Fisher's exact test were used to compare clinical parameters and frequency differences amongst PCOS patients with and without variants. Results A total of four rare exonic variants in obesity- and hyperinsulinaemia-related genes including UCP1 (p.Thr227Ile), UCP2 (p.Arg88Cys), IRS1 (p.Ser892Gly) and GHRL (p.Leu72Met) were identified in eight patients. Significant differences were observed between the patients carrying variants and those without variants. PCOS patients with identified variants exhibited significantly higher average body mass index and fasting insulin levels of PCOS subjects with identified variants compared to those without variants (P < 0.05). Additionally, there were significant differences in the variant frequencies of four variants when compared to the population database (P < 0.05). Conclusion This study shows a prevalence of rare variants in obesity and hyperinsulinaemia-related genes in a cohort of PCOS women, thereby underscoring the impact of the identified rare variants on the development of obesity and associated metabolic derangements in PCOS women.
Collapse
Affiliation(s)
- Priyal Sharma
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashutosh Halder
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Jain
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Tripathi
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
108
|
Brüning JC, Fenselau H. Integrative neurocircuits that control metabolism and food intake. Science 2023; 381:eabl7398. [PMID: 37769095 DOI: 10.1126/science.abl7398] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023]
Abstract
Systemic metabolism has to be constantly adjusted to the variance of food intake and even be prepared for anticipated changes in nutrient availability. Therefore, the brain integrates multiple homeostatic signals with numerous cues that predict future deviations in energy supply. Recently, our understanding of the neural pathways underlying these regulatory principles-as well as their convergence in the hypothalamus as the key coordinator of food intake, energy expenditure, and glucose metabolism-have been revealed. These advances have changed our view of brain-dependent control of metabolic physiology. In this Review, we discuss new concepts about how alterations in these pathways contribute to the development of prevalent metabolic diseases such as obesity and type 2 diabetes mellitus and how this emerging knowledge may provide new targets for their treatment.
Collapse
Affiliation(s)
- Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Henning Fenselau
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Research Group Synaptic Transmission in Energy Homeostasis, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| |
Collapse
|
109
|
Sun N, Victor MB, Park YP, Xiong X, Scannail AN, Leary N, Prosper S, Viswanathan S, Luna X, Boix CA, James BT, Tanigawa Y, Galani K, Mathys H, Jiang X, Ng AP, Bennett DA, Tsai LH, Kellis M. Human microglial state dynamics in Alzheimer's disease progression. Cell 2023; 186:4386-4403.e29. [PMID: 37774678 PMCID: PMC10644954 DOI: 10.1016/j.cell.2023.08.037] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/21/2023] [Accepted: 08/29/2023] [Indexed: 10/01/2023]
Abstract
Altered microglial states affect neuroinflammation, neurodegeneration, and disease but remain poorly understood. Here, we report 194,000 single-nucleus microglial transcriptomes and epigenomes across 443 human subjects and diverse Alzheimer's disease (AD) pathological phenotypes. We annotate 12 microglial transcriptional states, including AD-dysregulated homeostatic, inflammatory, and lipid-processing states. We identify 1,542 AD-differentially-expressed genes, including both microglia-state-specific and disease-stage-specific alterations. By integrating epigenomic, transcriptomic, and motif information, we infer upstream regulators of microglial cell states, gene-regulatory networks, enhancer-gene links, and transcription-factor-driven microglial state transitions. We demonstrate that ectopic expression of our predicted homeostatic-state activators induces homeostatic features in human iPSC-derived microglia-like cells, while inhibiting activators of inflammation can block inflammatory progression. Lastly, we pinpoint the expression of AD-risk genes in microglial states and differential expression of AD-risk genes and their regulators during AD progression. Overall, we provide insights underlying microglial states, including state-specific and AD-stage-specific microglial alterations at unprecedented resolution.
Collapse
Affiliation(s)
- Na Sun
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matheus B Victor
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yongjin P Park
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Pathology and Laboratory Medicine, Department of Statistics, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Xushen Xiong
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aine Ni Scannail
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Noelle Leary
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shaniah Prosper
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Soujanya Viswanathan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xochitl Luna
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carles A Boix
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin T James
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yosuke Tanigawa
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kyriaki Galani
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hansruedi Mathys
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Xueqiao Jiang
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ayesha P Ng
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Li-Huei Tsai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Manolis Kellis
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
110
|
Antkowiak M, Szydlowski M. Uncovering structural variants associated with body weight and obesity risk in labrador retrievers: a genome-wide study. Front Genet 2023; 14:1235821. [PMID: 37799139 PMCID: PMC10548226 DOI: 10.3389/fgene.2023.1235821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
Although obesity in the domestic dog (Canis lupus familiaris) is known to decrease well-being and shorten lifespan, the genetic risk variants associated with canine obesity remain largely unknown. In our study, which focused on the obesity-prone Labrador Retriever breed, we conducted a genome-wide analysis to identify structural variants linked to body weight and obesity. Obesity status was based on a 5-point body condition score (BCS) and the obese dog group included all dogs with a BCS of 5, along with dogs with the highest body weight within the BCS 4 group. Data from whole-gene sequencing of fifty dogs, including 28 obese dogs, were bioinformatically analyzed to identify potential structural variants that varied in frequency between obese and healthy dogs. The seven most promising variants were further analyzed by droplet digital PCR in a group of 110 dogs, including 63 obese. Our statistical evidence suggests that common structural mutations in or near six genes, specifically ALPL, KCTD8, SGSM1, SLC12A6, RYR3, and VPS26C, may contribute to the variability observed in body weight and body condition scores among Labrador Retriever dogs. These findings emphasize the need for additional research to validate the associations and explore the specific functions of these genes in relation to canine obesity.
Collapse
Affiliation(s)
| | - Maciej Szydlowski
- Department of Genetics and Animal Breeding, Poznań University of Life Sciences, Poznań, Poland
| |
Collapse
|
111
|
Viljakainen H, Sorlí JV, Dahlström E, Agrawal N, Portolés O, Corella D. Interaction between genetic susceptibility to obesity and food intake on BMI in Finnish school-aged children. Sci Rep 2023; 13:15265. [PMID: 37709841 PMCID: PMC10502078 DOI: 10.1038/s41598-023-42430-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023] Open
Abstract
Diet modulates the genetic risk of obesity, but the modulation has been rarely studied using genetic risk scores (GRSs) in children. Our objectives were to identify single nucleotide polymorphisms (SNPs) that drive the interaction of specific foods with obesity and combine these into GRSs. Genetic and food frequency data from Finnish Health in Teens study was utilized. In total, 1142 11-year-old subjects were genotyped on the Metabochip array. BMI-GRS with 30 well-known SNPs was computed and the interaction of individual SNPs with food items and their summary dietary scores were examined in relation to age- and sex-specific BMI z-score (BMIz). The whole BMI-GRS interacted with several foods on BMIz. We identified 7-11 SNPs responsible for each interaction and these were combined into food-specific GRS. The most predominant interaction was witnessed for pizza (p < 0.001): the effect on BMIz was b - 0.130 (95% CI - 0.23; - 0.031) in those with low-risk, and 0.153 (95% CI 0.072; 0.234) in high-risk. Corresponding, but weaker interactions were verified for sweets and chocolate, sugary juice drink, and hamburger and hotdog. In total 5 SNPs close to genes NEGR1, SEC16B, TMEM18, GNPDA2, and FTO were shared between these interactions. Our results suggested that children genetically prone to obesity showed a stronger association of unhealthy foods with BMIz than those with lower genetic susceptibility. Shared SNPs of the interactions suggest common differences in metabolic gene-diet interactions, which warrants further investigation.
Collapse
Affiliation(s)
- Heli Viljakainen
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland.
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Jose V Sorlí
- Department of Preventive Medicine and Public Health, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Emma Dahlström
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
| | - Nitin Agrawal
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olga Portolés
- Department of Preventive Medicine and Public Health, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Dolores Corella
- Department of Preventive Medicine and Public Health, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| |
Collapse
|
112
|
Hochner H, Butterman R, Margaliot I, Friedlander Y, Linial M. Obesity Prediction in Young Adults from the Jerusalem Perinatal Study: Contribution of Polygenic Risk and Early Life Exposures. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.05.23295076. [PMID: 37732179 PMCID: PMC10508819 DOI: 10.1101/2023.09.05.23295076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
We assessed whether adding early life exposures to a model based on polygenic risk score (PRS) improves prediction of obesity risk. We used a birth cohort with data at birth and BMI and waist circumference (WC) measured at age 32. The PRS was composed of SNPs identified in GWAS for BMI. Linear and logistic models were used to explore associations with obesity-related phenotypes. Improvement in prediction was assessed using measures of model discrimination (AUC), and net reclassification improvement (NRI). One SD change in PRS was associated with a significant increase in BMI and WC. These associations were slightly attenuated (13.7%-14.2%) with the addition of early life exposures to the model. Also, higher maternal pre-pregnancy BMI was associated with increase in offspring BMI and WC (p<0.001). For prediction obesity (BMI ≥ 30), the addition of early life exposures to the PRS model significantly increase the AUC from 0.69 to 0.73. At an obesity risk threshold of 15%, the addition of early life exposures to the PRS model provided a significant improvement in reclassification of obesity (NRI, 0.147; 95% CI 0.068-0.225). We conclude that inclusion of early life exposures to a model based on PRS improves obesity risk prediction in an Israeli population-sample.
Collapse
Affiliation(s)
- Hagit Hochner
- Braun school of public health, The Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| | - Rachely Butterman
- Braun school of public health, The Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| | - Ido Margaliot
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Yechiel Friedlander
- Braun school of public health, The Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| | - Michal Linial
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
113
|
Zhao F, Chen Y, Xie Y, Kong S, Song L, Li H, Guo C, Yin Y, Zhang W, Zhu T. Identification of Zip8-correlated hub genes in pulmonary hypertension by informatic analysis. PeerJ 2023; 11:e15939. [PMID: 37663293 PMCID: PMC10470448 DOI: 10.7717/peerj.15939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Background Pulmonary hypertension (PH) is a syndrome characterized by marked remodeling of the pulmonary vasculature and increased pulmonary vascular resistance, ultimately leading to right heart failure and even death. The localization of Zrt/Irt-like Protein 8 (ZIP8, a metal ion transporter, encoded by SLC39A8) was abundantly in microvasculature endothelium and its pivotal role in the lung has been demonstrated. However, the role of Zip8 in PH remains unclear. Methods Bioinformatics analysis was employed to identify SLC39A8 expression patterns and differentially expressed genes (DEGs) between PH patients and normal controls (NC), based on four datasets (GSE24988, GSE113439, GSE117261, and GSE15197) from the Biotechnology Gene Expression Omnibus (NCBI GEO) database. Gene set enrichment analysis (GSEA) was performed to analyze signaling pathways enriched for DEGs. Hub genes were identified by cytoHubba analysis in Cytoscape. Reverse transcriptase-polymerase chain reaction was used to validate SLC39A8 and its correlated metabolic DEGs expression in PH (SU5416/Hypoxia) mice. Results SLC39A8 expression was downregulated in PH patients, and this expression pattern was validated in PH (SU5416/Hypoxia) mouse lung tissue. SLC39A8-correlated genes were mainly enriched in the metabolic pathways. Within these SLC39A8-correlated genes, 202 SLC39A8-correlated metabolic genes were screened out, and seven genes were identified as SLC39A8-correlated metabolic hub genes. The expression patterns of hub genes were analyzed between PH patients and controls and further validated in PH mice. Finally, four genes (Fasn, Nsdhl, Acat2, and Acly) were downregulated in PH mice. However, there were no significant differences in the expression of the other three hub genes between PH mice and controls. Of the four genes, Fasn and Acly are key enzymes in fatty acids synthesis, Nsdhl is involved in cholesterol synthesis, and Acat2 is implicated in cholesterol metabolic transformation. Taken together, these results provide novel insight into the role of Zip8 in PH.
Collapse
Affiliation(s)
- FanRong Zhao
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yujing Chen
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yuliang Xie
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Shuang Kong
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - LiaoFan Song
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Hanfei Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Chao Guo
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yanyan Yin
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Weifang Zhang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Departments of Pharmacy, The Second Affiliated Hospital, Nanchang, China
| | - Tiantian Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| |
Collapse
|
114
|
Sag SJM, Mueller S, Wallner S, Strack C, Hubauer U, Mohr M, Zeller J, Loew T, Rehli M, Wimmer J, Zimmermann ME, Maier LS, Fischer M, Baessler A. A multilocus genetic risk score for obesity: Association with BMI and metabolic alterations in a cohort with severe obesity. Medicine (Baltimore) 2023; 102:e34597. [PMID: 37565910 PMCID: PMC10419793 DOI: 10.1097/md.0000000000034597] [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: 03/08/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Genome wide association studies have identified numerous single nucleotide polymorphisms (SNPs) associated with obesity, yet effect sizes of individual SNPs are small. Therefore, the aim of our study was to investigate whether a genetic risk score (GRS) comprising risk alleles of SNPs identified in the GIANT consortium meta-analyses shows association with body mass index (BMI) and other BMI related metabolic alterations in a cohort with an extreme phenotype. Genotyping of 93 SNPs was performed in 314 obese individuals (mean BMI 40.5 ± 7.8 kg/m², aged 45 ± 12 years), participating in a standardized weight reduction program, and in 74 lean controls (mean BMI 24.6 ± 3.3 kg/m², aged 41.7 ± 13.4 years). Allele numbers of all 93 SNPs were added to a GRS. Anthropometric parameters, parameters of glucose/insulin and lipid metabolism were assessed standardized after a 12 hours fast. GRS was significantly different between controls and obese individuals (unweighted GRS: 86.6 vs 89.0, P = .002; weighted GRS: 84.9 vs 88.3, P = .005). Furthermore, linear regression analysis showed significant associations of GRS with BMI ( P < .0001), weight ( P = .0005), waist circumference ( P = .0039), fat mass ( P < .0001) and epicardial fat thickness ( P = .0032), yet with small effect sizes ( r ² < 0.06). In conclusion, in our study GRS could differentiate between extreme obese and lean individuals, and was associated with BMI and its related traits, yet with small effect sizes.
Collapse
Affiliation(s)
| | - Stephanie Mueller
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wallner
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Christina Strack
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Ute Hubauer
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Margareta Mohr
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Judith Zeller
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Loew
- Department of Psychosomatics, University Hospital Regensburg, Regensburg, Germany
| | - Michael Rehli
- Clinic for Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Julia Wimmer
- Clinic for Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
- Deutsches Patent- und Markenamt, München, Germany
| | | | - Lars Siegfried Maier
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Marcus Fischer
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| | - Andrea Baessler
- Clinic for Internal Medicine 2, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
115
|
Cruz LA, Cooke Bailey JN, Crawford DC. Importance of Diversity in Precision Medicine: Generalizability of Genetic Associations Across Ancestry Groups Toward Better Identification of Disease Susceptibility Variants. Annu Rev Biomed Data Sci 2023; 6:339-356. [PMID: 37196357 PMCID: PMC10720270 DOI: 10.1146/annurev-biodatasci-122220-113250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Genome-wide association studies (GWAS) revolutionized our understanding of common genetic variation and its impact on common human disease and traits. Developed and adopted in the mid-2000s, GWAS led to searchable genotype-phenotype catalogs and genome-wide datasets available for further data mining and analysis for the eventual development of translational applications. The GWAS revolution was swift and specific, including almost exclusively populations of European descent, to the neglect of the majority of the world's genetic diversity. In this narrative review, we recount the GWAS landscape of the early years that established a genotype-phenotype catalog that is now universally understood to be inadequate for a complete understanding of complex human genetics. We then describe approaches taken to augment the genotype-phenotype catalog, including the study populations, collaborative consortia, and study design approaches aimed to generalize and then ultimately discover genome-wide associations in non-European descent populations. The collaborations and data resources established in the efforts to diversify genomic findings undoubtedly provide the foundations of the next chapters of genetic association studies with the advent of budget-friendly whole-genome sequencing.
Collapse
Affiliation(s)
- Lauren A Cruz
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA;
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA;
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA;
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
116
|
Mulder TA, Campbell PJ, Taylor PN, Peeters RP, Wilson SG, Medici M, Dayan C, Jaddoe VVW, Walsh JP, Martin NG, Tiemeier H, Korevaar TIM. Genetic determinants of thyroid function in children. Eur J Endocrinol 2023; 189:164-174. [PMID: 37530217 PMCID: PMC10402705 DOI: 10.1093/ejendo/lvad086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/20/2023] [Accepted: 06/06/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE Genome-wide association studies in adults have identified 42 loci associated with thyroid stimulating hormone (TSH) and 21 loci associated with free thyroxine (FT4) concentrations. While biologically plausible, age-dependent effects have not been assessed. We aimed to study the association of previously identified genetic determinants of TSH and FT4 with TSH and FT4 concentrations in newborns and (pre)school children. METHODS We selected participants from three population-based prospective cohorts with data on genetic variants and thyroid function: Generation R (N = 2169 children, mean age 6 years; N = 2388 neonates, the Netherlands), the Avon Longitudinal Study of Parents and Children (ALSPAC; N = 3382, age 7.5 years, United Kingdom), and the Brisbane Longitudinal Twin Study (BLTS; N = 1680, age 12.1 years, Australia). The association of single nucleotide polymorphisms (SNPs) with TSH and FT4 concentrations was studied with multivariable linear regression models. Weighted polygenic risk scores (PRSs) were defined to combine SNP effects. RESULTS In childhood, 30/60 SNPs were associated with TSH and 11/31 SNPs with FT4 after multiple testing correction. The effect sizes for AADAT, GLIS3, TM4SF4, and VEGFA were notably larger than in adults. The TSH PRS explained 5.3%-8.4% of the variability in TSH concentrations; the FT4 PRS explained 1.5%-4.2% of the variability in FT4 concentrations. Five TSH SNPs and no FT4 SNPs were associated with thyroid function in neonates. CONCLUSIONS The effects of many known thyroid function SNPs are already apparent in childhood and some might be notably larger in children as compared to adults. These findings provide new knowledge about genetic regulation of thyroid function in early life.
Collapse
Affiliation(s)
- Tessa A Mulder
- Generation R Study Group, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
| | - Purdey J Campbell
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Peter N Taylor
- Thyroid Research Group, Cardiff University School of Medicine, Cardiff, CF14 4YS, United Kingdom
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
| | - Scott G Wilson
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Department of Twin Research & Genetic Epidemiology, King's College London, London, WC2R 2LS, United Kingdom
| | - Marco Medici
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
| | - Colin Dayan
- Center for Endocrine and Diabetes Science, Cardiff University School of Medicine, Cardiff, CF14 4YS, United Kingdom
| | - Vincent V W Jaddoe
- Generation R Study Group, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - John P Walsh
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
- Medical School, The University of Western Australia, Crawley, WA 6009, Australia
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
- Department of Social and Behavioral Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
| | - Tim I M Korevaar
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, CA 3000, The Netherlands
| |
Collapse
|
117
|
You J, Ouyang S, Xie Z, Zhi C, Yu J, Tan X, Li P, Lin X, Ma W, Liu Z, Hou Q, Xie N, Peng T, Chen X, Li L, Xie W. The suppression of hyperlipid diet-induced ferroptosis of vascular smooth muscle cells protests against atherosclerosis independent of p53/SCL7A11/GPX4 axis. J Cell Physiol 2023; 238:1891-1908. [PMID: 37269460 DOI: 10.1002/jcp.31045] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 04/16/2023] [Accepted: 05/11/2023] [Indexed: 06/05/2023]
Abstract
Ferroptosis as a novel programmed cell death that involves metabolic dysfunction due to iron-dependent excessive lipid peroxidation has been implicated in atherosclerosis (AS) development characterized by disrupted lipid metabolism, but the atherogenic role of ferroptosis in vascular smooth muscle cells (VSMCs), which are principal components of atherosclerotic plaque fibrous cap, remains unclear. The aim of this study was to determine the effects of ferroptosis on AS induced by lipid overload, and the effects of that on VSMCs ferroptosis. We found intraperitoneal injection of Fer-1, a ferroptosis inhibitor, ameliorated obviously high-fat diet-induced high plasma levels of triglycerides, total cholesterol, low-density lipoprotein, glucose and atherosclerotic lesions in ApoE-/- mice. Moreover, in vivo and in vitro, Fer-1 reduced the iron accumulation of atherosclerotic lesions through affecting the expression of TFR1, FTH, and FTL in VSMCs. Interestingly, Fer-1 did augment nuclear factor E2-related factor 2/ferroptosis suppressor protein 1 to enhance endogenous resistance to lipid peroxidation, but not classic p53/SCL7A11/GPX4. Those observations indicated inhibition of VSMCs ferroptosis can improve AS lesions independent of p53/SLC7A11/GPX4, which preliminarily revealed the potential mechanism of ferroptosis in aortic VSMCs on AS and provided new therapeutic strategies and targets for AS.
Collapse
Affiliation(s)
- Jia You
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Siyu Ouyang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhongcheng Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chenxi Zhi
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang Yu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaoqian Tan
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Pin Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaoyan Lin
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wentao Ma
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhiyang Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qin Hou
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Nan Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
118
|
Kember RL, Vickers-Smith R, Zhou H, Xu H, Jennings M, Dao C, Davis L, Sanchez-Roige S, Justice AC, Gelernter J, Vujkovic M, Kranzler HR. Genetic Underpinnings of the Transition From Alcohol Consumption to Alcohol Use Disorder: Shared and Unique Genetic Architectures in a Cross-Ancestry Sample. Am J Psychiatry 2023; 180:584-593. [PMID: 37282553 PMCID: PMC10731616 DOI: 10.1176/appi.ajp.21090892] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
OBJECTIVE Recent genome-wide association studies (GWASs) of alcohol-related phenotypes have uncovered key differences in the underlying genetic architectures of alcohol consumption and alcohol use disorder (AUD), with the two traits having opposite genetic correlations with psychiatric disorders. Understanding the genetic factors that underlie the transition from heavy drinking to AUD has important theoretical and clinical implications. METHODS The authors used longitudinal data from the cross-ancestry Million Veteran Program sample to identify 1) novel loci associated with AUD and alcohol consumption (measured by the score on the consumption subscale of the Alcohol Use Disorders Identification Test [AUDIT-C]), 2) the impact of phenotypic variation on genetic discovery, and 3) genetic variants with direct effects on AUD that are not mediated through alcohol consumption. RESULTS The authors identified 26 loci associated with AUD and 22 loci associated with AUDIT-C score, including ancestry-specific and novel loci. In secondary GWASs that excluded individuals who report abstinence, the authors identified seven additional loci for AUD and eight additional loci for AUDIT-C score. Although the heterogeneity of the abstinent group biases the GWAS findings, unique variance between alcohol consumption and disorder remained after the abstinent group was excluded. Finally, using mediation analysis, the authors identified a set of variants with effects on AUD that are not mediated through alcohol consumption. CONCLUSIONS Differences in genetic architecture between alcohol consumption and AUD are consistent with their having different biological contributions. Genetic variants with direct effects on AUD are potentially relevant to understanding the transition from heavy alcohol consumption to AUD and may be targets for translational prevention and treatment efforts.
Collapse
Affiliation(s)
- Rachel L Kember
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Rachel Vickers-Smith
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Hang Zhou
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Heng Xu
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Mariela Jennings
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Cecilia Dao
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Lea Davis
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Sandra Sanchez-Roige
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Amy C Justice
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Joel Gelernter
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Marijana Vujkovic
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| | - Henry R Kranzler
- Mental Illness Research, Education, and Clinical Center, Veterans Integrated Service Network 4, Crescenz Veterans Affairs Medical Center, Philadelphia (Kember, Vickers-Smith, Kranzler); Center for Studies of Addiction, Department of Psychiatry (Kember, Xu, Kranzler) and Department of Epidemiology, University of Kentucky College of Public Health, Lexington (Vickers-Smith); Center on Drug and Alcohol Research, Department of Behavioral Science, University of Kentucky College of Medicine, Lexington (Vickers-Smith); VA Connecticut Healthcare System, West Haven (Zhou, Dao, Justice, Gelernter); Department of Psychiatry (Zhou, Gelernter), Department of Genetics (Gelernter), Department of Neuroscience (Gelernter), and Department of Internal Medicine (Justice), Yale School of Medicine, New Haven, Conn.; School of Public Health, Yale University, New Haven, Conn. (Dao, Justice); Department of Psychiatry, University of California San Diego, San Diego (Jennings, Sanchez-Roige); Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn. (Davis, Sanchez-Roige)
| |
Collapse
|
119
|
Reshetnikova Y, Churnosova M, Stepanov V, Bocharova A, Serebrova V, Trifonova E, Ponomarenko I, Sorokina I, Efremova O, Orlova V, Batlutskaya I, Ponomarenko M, Churnosov V, Eliseeva N, Aristova I, Polonikov A, Reshetnikov E, Churnosov M. Maternal Age at Menarche Gene Polymorphisms Are Associated with Offspring Birth Weight. Life (Basel) 2023; 13:1525. [PMID: 37511900 PMCID: PMC10381708 DOI: 10.3390/life13071525] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
In this study, the association between maternal age at menarche (AAM)-related polymorphisms and offspring birth weight (BW) was studied. The work was performed on a sample of 716 pregnant women and their newborns. All pregnant women underwent genotyping of 50 SNPs of AAM candidate genes. Regression methods (linear and Model-Based Multifactor Dimensionality Reduction (MB-MDR)) with permutation procedures (the indicator pperm was calculated) were used to identify the correlation between SNPs and newborn weight (transformed BW values were analyzed) and in silico bioinformatic examination was applied to assess the intended functionality of BW-associated loci. Four AAM-related genetic variants were BW-associated including genes such as POMC (rs7589318) (βadditive = 0.202/pperm = 0.015), KDM3B (rs757647) (βrecessive = 0.323/pperm = 0.005), INHBA (rs1079866) (βadditive = 0.110/pperm = 0.014) and NKX2-1 (rs999460) (βrecessive = -0.176/pperm = 0.015). Ten BW-significant models of interSNPs interactions (pperm ≤ 0.001) were identified for 20 polymorphisms. SNPs rs7538038 KISS1, rs713586 RBJ, rs12324955 FTO and rs713586 RBJ-rs12324955 FTO two-locus interaction were included in the largest number of BW-associated models (30% models each). BW-associated AAM-linked 22 SNPs and 350 proxy loci were functionally related to 49 genes relevant to pathways such as the hormone biosynthesis/process and female/male gonad development. In conclusion, maternal AMM-related genes polymorphism is associated with the offspring BW.
Collapse
Affiliation(s)
- Yuliya Reshetnikova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Vadim Stepanov
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Anna Bocharova
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Victoria Serebrova
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Ekaterina Trifonova
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Olga Efremova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Valentina Orlova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Irina Batlutskaya
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Marina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Vladimir Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Natalya Eliseeva
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (Y.R.); (M.C.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (N.E.); (I.A.); (A.P.); (E.R.)
| |
Collapse
|
120
|
Abstract
Obesity research is advancing swiftly, but the increase in obesity prevalence is faster. Over the past three decades, researchers have found that biopsychosocial factors determine weight gain much more than personal choices and responsibility. Various genes have found to predispose people to obesity by interacting with our obesogenic environment. In this review, we discuss the impact of physical inactivity, excessive caloric intake, intrauterine environment, postnatal influences, insufficient sleep, drugs, medical conditions, socioeconomic status, ethnicity, psychosocial stress, endocrine disrupting chemicals and the gastrointestinal microbiome, on the occurrence of obesity.
Collapse
|
121
|
Krishnan M, Phipps-Green A, Russell EM, Major TJ, Cadzow M, Stamp LK, Dalbeth N, Hindmarsh JH, Qasim M, Watson H, Liu S, Carlson JC, Minster RL, Hawley NL, Naseri T, Reupena MS, Deka R, McGarvey ST, Merriman TR, Murphy R, Weeks DE. Association of rs9939609 in FTO with BMI among Polynesian peoples living in Aotearoa New Zealand and other Pacific nations. J Hum Genet 2023; 68:463-468. [PMID: 36864286 PMCID: PMC10313811 DOI: 10.1038/s10038-023-01141-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/30/2023] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
The fat mass and obesity associated (FTO) locus consistently associates with higher body mass index (BMI) across diverse ancestral groups. However, previous small studies of people of Polynesian ancestries have failed to replicate the association. In this study, we used Bayesian meta-analysis to test rs9939609, the most replicated FTO variant, for association with BMI with a large sample (n = 6095) of Aotearoa New Zealanders of Polynesian (Māori and Pacific) ancestry and of Samoan people living in the Independent State of Samoa and in American Samoa. We did not observe statistically significant association within each separate Polynesian subgroup. Bayesian meta-analysis of the Aotearoa New Zealand Polynesian and Samoan samples resulted in a posterior mean effect size estimate of +0.21 kg/m2, with a 95% credible interval [+0.03 kg/m2, +0.39 kg/m2]. While the Bayes Factor (BF) of 0.77 weakly favors the null, the BF = 1.4 Bayesian support interval is [+0.04, +0.20]. These results suggest that rs9939609 in FTO may have a similar effect on mean BMI in people of Polynesian ancestries as previously observed in other ancestral groups.
Collapse
Affiliation(s)
- Mohanraj Krishnan
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Emily M Russell
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Jennie Harré Hindmarsh
- Ngāti Porou Hauora Charitable Trust, Te Puia Springs, Tairāwhiti East Coast, New Zealand
| | - Muhammad Qasim
- Ngāti Porou Hauora Charitable Trust, Te Puia Springs, Tairāwhiti East Coast, New Zealand
| | - Huti Watson
- Ngāti Porou Hauora Charitable Trust, Te Puia Springs, Tairāwhiti East Coast, New Zealand
| | - Shuwei Liu
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna C Carlson
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan L Minster
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicola L Hawley
- Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, CT, USA
| | - Take Naseri
- Ministry of Health, Government of Samoa, Apia, Samoa
- International Health Institute, Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA
| | | | - Ranjan Deka
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Stephen T McGarvey
- International Health Institute, Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA
- Department of Anthropology, Brown University, Providence, RI, USA
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Daniel E Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
122
|
He M, Yang T, Zhou P, Bu P, Yang X, Zou Y, Zhong A. A Mendelian randomization study on causal effects of 25(OH) vitamin D levels on diabetic nephropathy. BMC Nephrol 2023; 24:192. [PMID: 37369991 DOI: 10.1186/s12882-023-03186-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 04/27/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Vitamin D supplementation is associated with a lower incidence of diabetic nephropathy (DN); however, whether this association is causative is uncertain. METHODS We used two-sample Mendelian randomization to examine the causal influence of vitamin D on diabetic nephropathy in 7,751 individuals with type I diabetes-related nephropathy (T1DN) and 9,933 individuals with type II diabetes-related nephropathy (T2DN). Meanwhile, we repeated some previous studies on the influence of KIM-1 (kidney injury molecule 1) and body mass index (BMI) on DN. Additionally, to test the validity of the instruments variable for vitamin D, we conducted two negative controls Mendelian randomization (MR) on breast and prostate cancer, and a positive control MR on multiple sclerosis. RESULTS Results of the MR analysis showed that there was no causal association between 25(OH)D with the early/later stage of T1DN (early: OR = 0.903, 95%CI: 0.229 to 3.555; later: OR = 1.213, 95%CI: 0.367 to 4.010) and T2DN (early: OR = 0.588, 95%CI: 0.182 to 1.904; later: OR = 0.904, 95%CI: 0.376 to 2.173), nor with the kidney function of patients with diabetes mellitus: eGFRcyea (creatinine-based estimated GFR) (Beta = 0.007, 95%CI: -0.355 to 0.369)) or UACR (urinary albumin creatinine ratio) (Beta = 0.186, 95%CI: -0.961 to 1.333)). CONCLUSIONS We found no evidence that Vitamin D was causally associated with DN or kidney function in diabetic patients.
Collapse
Affiliation(s)
- Mingjie He
- Jiangxi Provincial Key Laboratory of Nephrology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ting Yang
- Jiangxi Provincial Key Laboratory of Nephrology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ping Zhou
- Jiangxi Provincial Key Laboratory of Nephrology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Peiyan Bu
- Medical College of Nanchang University, Nanchang University, 330006, Nanchang City, China
| | - Xionghui Yang
- Medical College of Nanchang University, Nanchang University, 330006, Nanchang City, China
| | - Yan Zou
- Jiangxi Provincial Key Laboratory of Nephrology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Aimin Zhong
- Jiangxi Provincial Key Laboratory of Nephrology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
- Medical College of Nanchang University, Nanchang University, 330006, Nanchang City, China.
| |
Collapse
|
123
|
Kasahara K, Kerby RL, Zhang Q, Pradhan M, Mehrabian M, Lusis AJ, Bergström G, Bäckhed F, Rey FE. Gut bacterial metabolism contributes to host global purine homeostasis. Cell Host Microbe 2023; 31:1038-1053.e10. [PMID: 37279756 PMCID: PMC10311284 DOI: 10.1016/j.chom.2023.05.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/25/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
The microbes and microbial pathways that influence host inflammatory disease progression remain largely undefined. Here, we show that variation in atherosclerosis burden is partially driven by gut microbiota and is associated with circulating levels of uric acid (UA) in mice and humans. We identify gut bacterial taxa spanning multiple phyla, including Bacillota, Fusobacteriota, and Pseudomonadota, that use multiple purines, including UA as carbon and energy sources anaerobically. We identify a gene cluster that encodes key steps of anaerobic purine degradation and that is widely distributed among gut-dwelling bacteria. Furthermore, we show that colonization of gnotobiotic mice with purine-degrading bacteria modulates levels of UA and other purines in the gut and systemically. Thus, gut microbes are important drivers of host global purine homeostasis and serum UA levels, and gut bacterial catabolism of purines may represent a mechanism by which gut bacteria influence health.
Collapse
Affiliation(s)
- Kazuyuki Kasahara
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Robert L Kerby
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Qijun Zhang
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Meenakshi Pradhan
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Margarete Mehrabian
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
124
|
Kosugi S, Kamatani Y, Harada K, Tomizuka K, Momozawa Y, Morisaki T, The BioBank Japan Project, Terao C. Detection of trait-associated structural variations using short-read sequencing. CELL GENOMICS 2023; 3:100328. [PMID: 37388916 PMCID: PMC10300613 DOI: 10.1016/j.xgen.2023.100328] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 02/17/2023] [Accepted: 04/25/2023] [Indexed: 07/01/2023]
Abstract
Genomic structural variation (SV) affects genetic and phenotypic characteristics in diverse organisms, but the lack of reliable methods to detect SV has hindered genetic analysis. We developed a computational algorithm (MOPline) that includes missing call recovery combined with high-confidence SV call selection and genotyping using short-read whole-genome sequencing (WGS) data. Using 3,672 high-coverage WGS datasets, MOPline stably detected ∼16,000 SVs per individual, which is over ∼1.7-3.3-fold higher than previous large-scale projects while exhibiting a comparable level of statistical quality metrics. We imputed SVs from 181,622 Japanese individuals for 42 diseases and 60 quantitative traits. A genome-wide association study with the imputed SVs revealed 41 top-ranked or nearly top-ranked genome-wide significant SVs, including 8 exonic SVs with 5 novel associations and enriched mobile element insertions. This study demonstrates that short-read WGS data can be used to identify rare and common SVs associated with a variety of traits.
Collapse
Affiliation(s)
- Shunichi Kosugi
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
| | - Yoichiro Kamatani
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Japan
| | - Katsutoshi Harada
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Takayuki Morisaki
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokane-dai, Minato-ku, Tokyo 108-8639, Japan
| | | | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
125
|
Murray SB, Rokicki J, Sartorius AM, Winterton A, Andreassen OA, Westlye LT, Nagata JM, Quintana DS. Brain-based gene expression of putative risk genes for anorexia nervosa. Mol Psychiatry 2023; 28:2612-2619. [PMID: 37221367 DOI: 10.1038/s41380-023-02110-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/25/2023]
Abstract
The etiology of anorexia nervosa (AN) remains elusive. Recent genome-wide association studies identified the first genes liked to AN which reached genome-wide significance, although our understanding of how these genes confer risk remains preliminary. Here, we leverage the Allen Human Brain Atlas to characterize the spatially distributed gene expression patterns of genes linked to AN in the non-disordered human brain, developing whole-brain maps of AN gene expression. We found that genes associated with AN are most expressed in the brain, relative to all other body tissue types, and demonstrate gene-specific expression patterns which extend to cerebellar, temporal and basal ganglia structures in particular. fMRI meta-analyses reveal that AN gene expression maps correspond with functional brain activity involved in processing and anticipating appetitive and aversive cues. Findings offer novel insights around putative mechanisms through which genes associated with AN may confer risk.
Collapse
Affiliation(s)
- Stuart B Murray
- Department of Psychiatry & Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jaroslav Rokicki
- Norwegian Centre for Mental Disorders Research (NORMENT), Division for Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Alina M Sartorius
- Norwegian Centre for Mental Disorders Research (NORMENT), Division for Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Adriano Winterton
- Norwegian Centre for Mental Disorders Research (NORMENT), Division for Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division for Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Division for Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Jason M Nagata
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel S Quintana
- Norwegian Centre for Mental Disorders Research (NORMENT), Division for Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway.
- Department of Psychology, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway.
- NevSom, Department of Rare Disorders and Disabilities, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
126
|
Purnell JQ. What is Obesity?: Definition as a Disease, with Implications for Care. Gastroenterol Clin North Am 2023; 52:261-275. [PMID: 37197872 DOI: 10.1016/j.gtc.2023.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Advances in the understanding of weight regulation provide the framework for the recognition of obesity as a chronic disease. Lifestyle approaches are foundational in the prevention of obesity and should be continued while weight management interventions, including antiobesity medications and metabolic-bariatric procedures, are offered to eligible patients. Clinical challenges remain, however, including overcoming obesity stigma and bias within the medical community toward medical and surgical approaches, ensuring insurance coverage for obesity management (including medications and surgery), and promoting policies that reverse the upward worldwide trend in obesity and adiposity complications in populations.
Collapse
Affiliation(s)
- Jonathan Q Purnell
- Knight Cardiovascular Institute and Division of Endocrinology, Diabetes, and Clinical Nutrition, Oregon Health & Science University, Mailcode: HRC5N, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
127
|
Chu P, Guo W, You H, Lu B. Regulation of Satiety by Bdnf-e2-Expressing Neurons through TrkB Activation in Ventromedial Hypothalamus. Biomolecules 2023; 13:biom13050822. [PMID: 37238691 DOI: 10.3390/biom13050822] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The transcripts for Bdnf (brain-derived neurotrophic factor), driven by different promoters, are expressed in different brain regions to control different body functions. Specific promoter(s) that regulates energy balance remain unclear. We show that disruption of Bdnf promoters I and II but not IV and VI in mice (Bdnf-e1-/-, Bdnf-e2-/-) results in obesity. Whereas Bdnf-e1-/- exhibited impaired thermogenesis, Bdnf-e2-/- showed hyperphagia and reduced satiety before the onset of obesity. The Bdnf-e2 transcripts were primarily expressed in ventromedial hypothalamus (VMH), a nucleus known to regulate satiety. Re-expressing Bdnf-e2 transcript in VMH or chemogenetic activation of VMH neurons rescued the hyperphagia and obesity of Bdnf-e2-/- mice. Deletion of BDNF receptor TrkB in VMH neurons in wildtype mice resulted in hyperphagia and obesity, and infusion of TrkB agonistic antibody into VMH of Bdnf-e2-/- mice alleviated these phenotypes. Thus, Bdnf-e2-transcripts in VMH neurons play a key role in regulating energy intake and satiety through TrkB pathway.
Collapse
Affiliation(s)
- Pengcheng Chu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - He You
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Centre, 10 Marais Street, Stellenbosch 7600, South Africa
| |
Collapse
|
128
|
Edwin Thanarajah S, Hanssen R, Melzer C, Tittgemeyer M. Increased meso-striatal connectivity mediates trait impulsivity in FTO variant carriers. Front Endocrinol (Lausanne) 2023; 14:1130203. [PMID: 37223038 PMCID: PMC10200952 DOI: 10.3389/fendo.2023.1130203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/31/2023] [Indexed: 05/25/2023] Open
Abstract
Objective While variations in the first intron of the fat mass and obesity-associated gene (FTO, rs9939609 T/A variant) have long been identified as a major contributor to polygenic obesity, the mechanisms underlying weight gain in risk allele carriers still remain elusive. On a behavioral level, FTO variants have been robustly linked to trait impulsivity. The regulation of dopaminergic signaling in the meso-striatal neurocircuitry by these FTO variants might represent one mechanism for this behavioral alteration. Notably, recent evidence indicates that variants of FTO also modulate several genes involved in cell proliferation and neuronal development. Hence, FTO polymorphisms might establish a predisposition to heightened trait impulsivity during neurodevelopment by altering structural meso-striatal connectivity. We here explored whether the greater impulsivity of FTO variant carriers was mediated by structural differences in the connectivity between the dopaminergic midbrain and the ventral striatum. Methods Eighty-seven healthy normal-weight volunteers participated in the study; 42 FTO risk allele carriers (rs9939609 T/A variant, FTO + group: AT, AA) and 39 non-carriers (FTO - group: TT) were matched for age, sex and body mass index (BMI). Trait impulsivity was assessed via the Barratt Impulsiveness Scale (BIS-11) and structural connectivity between the ventral tegmental area/substantia nigra (VTA/SN) and the nucleus accumbens (NAc) was measured via diffusion weighted MRI and probabilistic tractography. Results We found that FTO risk allele carriers compared to non-carriers, demonstrated greater motor impulsivity (p = 0.04) and increased structural connectivity between VTA/SN and the NAc (p< 0.05). Increased connectivity partially mediated the effect of FTO genetic status on motor impulsivity. Conclusion We report altered structural connectivity as one mechanism by which FTO variants contribute to increased impulsivity, indicating that FTO variants may exert their effect on obesity-promoting behavioral traits at least partially through neuroplastic alterations in humans.
Collapse
Affiliation(s)
- Sharmili Edwin Thanarajah
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Ruth Hanssen
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEPD), University of Cologne, Cologne, Germany
| | - Corina Melzer
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
129
|
Zabad S, Gravel S, Li Y. Fast and accurate Bayesian polygenic risk modeling with variational inference. Am J Hum Genet 2023; 110:741-761. [PMID: 37030289 PMCID: PMC10183379 DOI: 10.1016/j.ajhg.2023.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/13/2023] [Indexed: 04/10/2023] Open
Abstract
The advent of large-scale genome-wide association studies (GWASs) has motivated the development of statistical methods for phenotype prediction with single-nucleotide polymorphism (SNP) array data. These polygenic risk score (PRS) methods use a multiple linear regression framework to infer joint effect sizes of all genetic variants on the trait. Among the subset of PRS methods that operate on GWAS summary statistics, sparse Bayesian methods have shown competitive predictive ability. However, most existing Bayesian approaches employ Markov chain Monte Carlo (MCMC) algorithms, which are computationally inefficient and do not scale favorably to higher dimensions, for posterior inference. Here, we introduce variational inference of polygenic risk scores (VIPRS), a Bayesian summary statistics-based PRS method that utilizes variational inference techniques to approximate the posterior distribution for the effect sizes. Our experiments with 36 simulation configurations and 12 real phenotypes from the UK Biobank dataset demonstrated that VIPRS is consistently competitive with the state-of-the-art in prediction accuracy while being more than twice as fast as popular MCMC-based approaches. This performance advantage is robust across a variety of genetic architectures, SNP heritabilities, and independent GWAS cohorts. In addition to its competitive accuracy on the "White British" samples, VIPRS showed improved transferability when applied to other ethnic groups, with up to 1.7-fold increase in R2 among individuals of Nigerian ancestry for low-density lipoprotein (LDL) cholesterol. To illustrate its scalability, we applied VIPRS to a dataset of 9.6 million genetic markers, which conferred further improvements in prediction accuracy for highly polygenic traits, such as height.
Collapse
Affiliation(s)
- Shadi Zabad
- School of Computer Science, McGill University, Montreal, QC, Canada
| | - Simon Gravel
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| | - Yue Li
- School of Computer Science, McGill University, Montreal, QC, Canada.
| |
Collapse
|
130
|
Lv HY, Shi G, Li C, Ye YF, Chen YH, Chen LH, Tung TH, Zhang M. Association of SULT1A2 rs1059491 with obesity and dyslipidaemia in southern Chinese adults. Sci Rep 2023; 13:7256. [PMID: 37142702 PMCID: PMC10160091 DOI: 10.1038/s41598-023-34296-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/27/2023] [Indexed: 05/06/2023] Open
Abstract
In the sulfotransferase (SULT) superfamily, members of the SULT1 family mainly catalyse the sulfonation reaction of phenolic compounds, which is involved in the phase II metabolic detoxification process and plays a key role in endocrine homeostasis. A coding variant rs1059491 in the SULT1A2 gene has been reported to be associated with childhood obesity. This study aimed to investigate the association of rs1059491 with the risk of obesity and cardiometabolic abnormalities in adults. This case‒control study included 226 normal weight, 168 overweight and 72 obese adults who underwent a health examination in Taizhou, China. Genotyping of rs1059491 was performed by Sanger sequencing in exon 7 of the SULT1A2 coding region. Chi-squared tests, one-way ANOVA, and logistic regression models were applied. The minor allele frequencies of rs1059491 in the overweight combined with obesity and control groups were 0.0292 and 0.0686, respectively. No differences in weight and body mass index were detected between the TT genotype and GT + GG genotype under the dominant model, but the levels of serum triglycerides were significantly lower in G-allele carriers than in non-G-allele carriers (1.02 (0.74-1.32) vs. 1.35 (0.83-2.13) mmol/L, P = 0.011). The GT + GG genotype of rs1059491 versus the TT genotype reduced the risk of overweight and obesity by 54% (OR 0.46, 95% CI 0.22-0.96, P = 0.037) after adjusting for sex and age. Similar results were observed for hypertriglyceridaemia (OR 0.25, 95% CI 0.08-0.74, P = 0.013) and dyslipidaemia (OR 0.37, 95% CI 0.17-0.83, P = 0.015). However, these associations disappeared after correction for multiple tests. This study revealed that the coding variant rs1059491 is nominally associated with a decreased risk of obesity and dyslipidaemia in southern Chinese adults. The findings will be validated in larger studies including more detailed information on genetic background, lifestyle and weight change with age.
Collapse
Affiliation(s)
- Hai-Yan Lv
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Guifeng Shi
- Department of Preventive Health Care, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Cai Li
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Ya-Fei Ye
- Health Management Centre, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Ya-Hong Chen
- Health Management Centre, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Li-Hua Chen
- Public Scientific Research Platform, Taizhou Hospital of Zhejiang Province Affiliated To Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Tao-Hsin Tung
- Evidence-Based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, 317000, Zhejiang, China
| | - Meixian Zhang
- Evidence-Based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, 317000, Zhejiang, China.
| |
Collapse
|
131
|
Powers M, Minchella D, Gonzalez-Acevedo M, Escutia-Plaza D, Wu J, Heger C, Milne G, Aschner M, Liu Z. Loss of hepatic manganese transporter ZIP8 disrupts serum transferrin glycosylation and the glutamate-glutamine cycle. J Trace Elem Med Biol 2023; 78:127184. [PMID: 37163821 DOI: 10.1016/j.jtemb.2023.127184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/07/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND ZIP8, encoded by SLC39A8, is a membrane transporter that facilitates the cellular uptake of divalent biometals including zinc (Zn), manganese (Mn), and iron (Fe). The hepatic system has long been accepted as the central modulator for whole-body biometal distribution. Earlier investigations suggest the propensity of ZIP8 to prioritize Mn influx, as opposed to Fe or Zn, in hepatocytes. Hepatic ZIP8 Mn transport is crucial for maintaining homeostasis of various Mn-dependent metalloenzymes and their associated pathways. Herein, we hypothesize that a drastic decrease in systemic Mn, via the loss of hepatic ZIP8, disrupts two unique cellular pathways, post-translational glycosylation and the glutamate-glutamine cycle. METHODS ZIP8 liver-specific knockout (LSKO) mice were chosen in an attempt to substantially decrease whole-body Mn levels. To further elucidate the role of Mn in serum glycosylation, a Mn-deficient diet was adopted in conjunction with the LSKO mice to model a near-complete loss of systemic Mn. After the treatment course, transferrin sialylation profiles were determined using imaged capillary isoelectric focusing (icIEF). We also investigated the role of Mn in the glutamate-glutamine cycle; the conversion of glutamate to glutamine in F/F and LSKO mice was assessed by the glutamine/glutamate ratio in cerebrospinal fluid (CSF) via HPLC-MS. An open-field study was ultimately conducted to check if these mice displayed atypical behavior. RESULTS Two major biological pathways were found to be significantly altered due to the loss of hepatic ZIP8. We identified a disparity between F/F and LSKO transferrin sialylation profiles that were exacerbated under a Mn-deficient diet. Additionally, we discovered a neurotransmitter imbalance between the levels of glutamine and glutamate, exclusive to LSKO mice. This was characterized by the decreased glutamine/glutamate ratio in CSF. Secondary to the neurotransmitter alteration, LSKO mice exhibited an increase in locomotor activity in an open-field. CONCLUSION Our model successfully established a connection between the loss of hepatic ZIP8 and two Mn-dependent cellular pathways, namely, protein glycosylation and the glutamate-glutamine cycle.
Collapse
Affiliation(s)
- Michael Powers
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Dean Minchella
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | | | | | - Jiaqi Wu
- ProteinSimple, A Bio-Techne Brand, San Jose, CA, USA
| | - Chris Heger
- ProteinSimple, A Bio-Techne Brand, San Jose, CA, USA
| | - Ginger Milne
- Neurochemistry Core, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Michael Aschner
- Department of Cellular Biology and Pharmacology, Albert Einstein Medical College, New York, USA
| | - Zijuan Liu
- Department of Biological Sciences, Oakland University, Rochester, MI, USA.
| |
Collapse
|
132
|
Nigro P, Vamvini M, Yang J, Caputo T, Ho LL, Carbone NP, Papadopoulos D, Conlin R, He J, Hirshman MF, White JD, Robidoux J, Hickner RC, Nielsen S, Pedersen BK, Kellis M, Middelbeek RJW, Goodyear LJ. Exercise training remodels inguinal white adipose tissue through adaptations in innervation, vascularization, and the extracellular matrix. Cell Rep 2023; 42:112392. [PMID: 37058410 PMCID: PMC10374102 DOI: 10.1016/j.celrep.2023.112392] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/13/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023] Open
Abstract
Inguinal white adipose tissue (iWAT) is essential for the beneficial effects of exercise training on metabolic health. The underlying mechanisms for these effects are not fully understood, and here, we test the hypothesis that exercise training results in a more favorable iWAT structural phenotype. Using biochemical, imaging, and multi-omics analyses, we find that 11 days of wheel running in male mice causes profound iWAT remodeling including decreased extracellular matrix (ECM) deposition and increased vascularization and innervation. We identify adipose stem cells as one of the main contributors to training-induced ECM remodeling, show that the PRDM16 transcriptional complex is necessary for iWAT remodeling and beiging, and discover neuronal growth regulator 1 (NEGR1) as a link between PRDM16 and neuritogenesis. Moreover, we find that training causes a shift from hypertrophic to insulin-sensitive adipocyte subpopulations. Exercise training leads to remarkable adaptations to iWAT structure and cell-type composition that can confer beneficial changes in tissue metabolism.
Collapse
Affiliation(s)
- Pasquale Nigro
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Maria Vamvini
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jiekun Yang
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tiziana Caputo
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Lun Ho
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas P Carbone
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Danae Papadopoulos
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Royce Conlin
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jie He
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Joseph D White
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, USA
| | - Jacques Robidoux
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, USA
| | - Robert C Hickner
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, USA; Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Søren Nielsen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bente K Pedersen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Manolis Kellis
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
133
|
Keijer J, Escoté X, Galmés S, Palou-March A, Serra F, Aldubayan MA, Pigsborg K, Magkos F, Baker EJ, Calder PC, Góralska J, Razny U, Malczewska-Malec M, Suñol D, Galofré M, Rodríguez MA, Canela N, Malcic RG, Bosch M, Favari C, Mena P, Del Rio D, Caimari A, Gutierrez B, Del Bas JM. Omics biomarkers and an approach for their practical implementation to delineate health status for personalized nutrition strategies. Crit Rev Food Sci Nutr 2023; 64:8279-8307. [PMID: 37077157 DOI: 10.1080/10408398.2023.2198605] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Personalized nutrition (PN) has gained much attention as a tool for empowerment of consumers to promote changes in dietary behavior, optimizing health status and preventing diet related diseases. Generalized implementation of PN faces different obstacles, one of the most relevant being metabolic characterization of the individual. Although omics technologies allow for assessment the dynamics of metabolism with unprecedented detail, its translatability as affordable and simple PN protocols is still difficult due to the complexity of metabolic regulation and to different technical and economical constrains. In this work, we propose a conceptual framework that considers the dysregulation of a few overarching processes, namely Carbohydrate metabolism, lipid metabolism, inflammation, oxidative stress and microbiota-derived metabolites, as the basis of the onset of several non-communicable diseases. These processes can be assessed and characterized by specific sets of proteomic, metabolomic and genetic markers that minimize operational constrains and maximize the information obtained at the individual level. Current machine learning and data analysis methodologies allow the development of algorithms to integrate omics and genetic markers. Reduction of dimensionality of variables facilitates the implementation of omics and genetic information in digital tools. This framework is exemplified by presenting the EU-Funded project PREVENTOMICS as a use case.
Collapse
Affiliation(s)
- Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Xavier Escoté
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, Reus, Spain
| | - Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Andreu Palou-March
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Francisca Serra
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Mona Adnan Aldubayan
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Nutrition, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Kristina Pigsborg
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ella J Baker
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Joanna Góralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Urszula Razny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | | | - David Suñol
- Digital Health, Eurecat, Centre Tecnològic de Catalunya, Barcelona, Spain
| | - Mar Galofré
- Digital Health, Eurecat, Centre Tecnològic de Catalunya, Barcelona, Spain
| | - Miguel A Rodríguez
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, Reus, Spain
| | - Núria Canela
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, Reus, Spain
| | - Radu G Malcic
- Health and Biomedicine, LEITAT Technological Centre, Barcelona, Spain
| | - Montserrat Bosch
- Applied Microbiology and Biotechnologies, LEITAT Technological Centre, Terrassa, Spain
| | - Claudia Favari
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology area, Reus, Spain
| | | | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology area, Reus, Spain
| |
Collapse
|
134
|
Deng WQ, Belisario K, Gray JC, Levitt EE, Mohammadi-Shemirani P, Singh D, Pare G, MacKillop J. Leveraging related health phenotypes for polygenic prediction of impulsive choice, impulsive action, and impulsive personality traits in 1534 European ancestry community adults. GENES, BRAIN, AND BEHAVIOR 2023:e12848. [PMID: 37060189 DOI: 10.1111/gbb.12848] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/17/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023]
Abstract
Impulsivity refers to a number of conceptually related phenotypes reflecting self-regulatory capacity that are considered promising endophenotypes for mental and physical health. Measures of impulsivity can be broadly grouped into three domains, namely, impulsive choice, impulsive action, and impulsive personality traits. In a community-based sample of ancestral Europeans (n = 1534), we conducted genome-wide association studies (GWASs) of impulsive choice (delay discounting), impulsive action (behavioral inhibition), and impulsive personality traits (UPPS-P), and evaluated 11 polygenic risk scores (PRSs) of phenotypes previously linked to self-regulation. Although there were no individual genome-wide significant hits, the neuroticism PRS was positively associated with negative urgency (adjusted R2 = 1.61%, p = 3.6 × 10-7 ) and the educational attainment PRS was inversely associated with delay discounting (adjusted R2 = 1.68%, p = 2.2 × 10-7 ). There was also evidence implicating PRSs of attention-deficit/hyperactivity disorder, externalizing, risk-taking, smoking cessation, smoking initiation, and body mass index with one or more impulsivity phenotypes (adjusted R2 s: 0.35%-1.07%; FDR adjusted ps = 0.05-0.0006). These significant associations between PRSs and impulsivity phenotypes are consistent with established genetic correlations. The combined PRS explained 0.91%-2.46% of the phenotypic variance for individual impulsivity measures, corresponding to 8.7%-32.5% of their reported single-nucleotide polymorphism (SNP)-based heritability, suggesting a non-negligible portion of the SNP-based heritability can be recovered by PRSs. These results support the predictive validity and utility of PRSs, even derived from related phenotypes, to inform the genetics of impulsivity phenotypes.
Collapse
Affiliation(s)
- Wei Q Deng
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Kyla Belisario
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Joshua C Gray
- Department of Medical and Clinical Psychology, Uniformed Services University, Bethesda, Maryland, USA
| | - Emily E Levitt
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | | | - Desmond Singh
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Biology, University of Waterloo, Wterloo, Canada
| | - Guillaume Pare
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - James MacKillop
- Peter Boris Centre for Addictions Research, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| |
Collapse
|
135
|
Radford-Smith DE, Anthony DC. Prebiotic and Probiotic Modulation of the Microbiota-Gut-Brain Axis in Depression. Nutrients 2023; 15:nu15081880. [PMID: 37111100 PMCID: PMC10146605 DOI: 10.3390/nu15081880] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Emerging evidence demonstrates that alterations to the gut microbiota can affect mood, suggesting that the microbiota-gut-brain (MGB) axis contributes to the pathogenesis of depression. Many of these pathways overlap with the way in which the gut microbiota are thought to contribute to metabolic disease progression and obesity. In rodents, prebiotics and probiotics have been shown to modulate the composition and function of the gut microbiota. Together with germ-free rodent models, probiotics have provided compelling evidence for a causal relationship between microbes, microbial metabolites, and altered neurochemical signalling and inflammatory pathways in the brain. In humans, probiotic supplementation has demonstrated modest antidepressant effects in individuals with depressive symptoms, though more studies in clinically relevant populations are needed. This review critically discusses the role of the MGB axis in depression pathophysiology, integrating preclinical and clinical evidence, as well as the putative routes of communication between the microbiota-gut interface and the brain. A critical overview of the current approaches to investigating microbiome changes in depression is provided. To effectively translate preclinical breakthroughs in MGB axis research into novel therapies, rigorous placebo-controlled trials alongside a mechanistic and biochemical understanding of prebiotic and probiotic action are required from future research.
Collapse
Affiliation(s)
- Daniel E Radford-Smith
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX3 7JX, UK
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
136
|
Austin GO, Tomas A. Variation in responses to incretin therapy: Modifiable and non-modifiable factors. Front Mol Biosci 2023; 10:1170181. [PMID: 37091864 PMCID: PMC10119428 DOI: 10.3389/fmolb.2023.1170181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Type 2 diabetes (T2D) and obesity have reached epidemic proportions. Incretin therapy is the second line of treatment for T2D, improving both blood glucose regulation and weight loss. Glucagon-like peptide-1 (GLP-1) and glucose-stimulated insulinotropic polypeptide (GIP) are the incretin hormones that provide the foundations for these drugs. While these therapies have been highly effective for some, the results are variable. Incretin therapies target the class B G protein-coupled receptors GLP-1R and GIPR, expressed mainly in the pancreas and the hypothalamus, while some therapeutical approaches include additional targeting of the related glucagon receptor (GCGR) in the liver. The proper functioning of these receptors is crucial for incretin therapy success and here we review several mechanisms at the cellular and molecular level that influence an individual's response to incretin therapy.
Collapse
Affiliation(s)
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
137
|
Fu M, Zhou X, Liu Z, Wang T, Liu B. Genome-wide association study reveals a genomic region on SSC7 simultaneously associated with backfat thickness, skin thickness and carcass length in a Large White × Tongcheng advanced generation intercross resource population. Anim Genet 2023; 54:216-219. [PMID: 36511585 DOI: 10.1111/age.13285] [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: 01/19/2022] [Revised: 11/17/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022]
Abstract
In order to identify important genetic markers associated with backfat thickness, skin thickness and carcass length, we first constructed Large White × Tongcheng (Chinese local breed), an advanced generation intercross population, then performed a genome-wide association study (GWAS) to reveal the key genomic region associated with these traits through whole genome sequencing. The GWAS results of backfat thickness, skin thickness and carcass length showed that all the most significant SNPs associated with these three traits were located on SSC7, and that 14.9, 27.0 and 21.1% of phenotypic variances were explained by these three SNPs, respectively. Through linkage disequilibrium analysis, we found that a 66.9 kb (30.23-30.31 Mb) genetic region was overlapped among these three traits, and that NUDT3 and HMGA1 were identified as major candidate genes for backfat thickness and carcass length, and GRM4 as a potential candidate gene for skin thickness. In addition, 13 highly linked SNPs significantly associated with the three traits were also identified in overlapped region, and three completely linked SNPs formed two haplotypes Q and q. The backfat thickness of individuals with the qq genotype was significantly lower than that of individuals with the QQ genotype, but their carcass length and skin thickness were significantly higher than those with the QQ genotype. Our detected candidate genes and SNPs will provide the foundation for genetic improvement of these three traits.
Collapse
Affiliation(s)
- Ming Fu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Xiang Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,Hubei Hongshan Laboratory, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,The Engineering Technology Research Center of Hubei Province Local Pig Breed Improvement, Wuhan, China
| | - Zuhong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Tengfei Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Bang Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,Hubei Hongshan Laboratory, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,The Engineering Technology Research Center of Hubei Province Local Pig Breed Improvement, Wuhan, China
| |
Collapse
|
138
|
Al-Sawaf O, Weiss J, Skrzypski M, Lam JM, Karasaki T, Zambrana F, Kidd AC, Frankell AM, Watkins TBK, Martínez-Ruiz C, Puttick C, Black JRM, Huebner A, Bakir MA, Sokač M, Collins S, Veeriah S, Magno N, Naceur-Lombardelli C, Prymas P, Toncheva A, Ward S, Jayanth N, Salgado R, Bridge CP, Christiani DC, Mak RH, Bay C, Rosenthal M, Sattar N, Welsh P, Liu Y, Perrimon N, Popuri K, Beg MF, McGranahan N, Hackshaw A, Breen DM, O'Rahilly S, Birkbak NJ, Aerts HJWL, Jamal-Hanjani M, Swanton C. Body composition and lung cancer-associated cachexia in TRACERx. Nat Med 2023; 29:846-858. [PMID: 37045997 PMCID: PMC7614477 DOI: 10.1038/s41591-023-02232-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/24/2023] [Indexed: 04/14/2023]
Abstract
Cancer-associated cachexia (CAC) is a major contributor to morbidity and mortality in individuals with non-small cell lung cancer. Key features of CAC include alterations in body composition and body weight. Here, we explore the association between body composition and body weight with survival and delineate potential biological processes and mediators that contribute to the development of CAC. Computed tomography-based body composition analysis of 651 individuals in the TRACERx (TRAcking non-small cell lung Cancer Evolution through therapy (Rx)) study suggested that individuals in the bottom 20th percentile of the distribution of skeletal muscle or adipose tissue area at the time of lung cancer diagnosis, had significantly shorter lung cancer-specific survival and overall survival. This finding was validated in 420 individuals in the independent Boston Lung Cancer Study. Individuals classified as having developed CAC according to one or more features at relapse encompassing loss of adipose or muscle tissue, or body mass index-adjusted weight loss were found to have distinct tumor genomic and transcriptomic profiles compared with individuals who did not develop such features. Primary non-small cell lung cancers from individuals who developed CAC were characterized by enrichment of inflammatory signaling and epithelial-mesenchymal transitional pathways, and differentially expressed genes upregulated in these tumors included cancer-testis antigen MAGEA6 and matrix metalloproteinases, such as ADAMTS3. In an exploratory proteomic analysis of circulating putative mediators of cachexia performed in a subset of 110 individuals from TRACERx, a significant association between circulating GDF15 and loss of body weight, skeletal muscle and adipose tissue was identified at relapse, supporting the potential therapeutic relevance of targeting GDF15 in the management of CAC.
Collapse
Affiliation(s)
- Othman Al-Sawaf
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Jakob Weiss
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Diagnostic and Interventional Radiology, University Freiburg, Freiburg, Germany
| | - Marcin Skrzypski
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Jie Min Lam
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Andrew C Kidd
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Thomas B K Watkins
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare Puttick
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Mateo Sokač
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Susie Collins
- Early Clinical Development, Pfizer UK Ltd, Cambridge, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Neil Magno
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Paulina Prymas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Antonia Toncheva
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Nick Jayanth
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - David C Christiani
- Department of Medicine, Massachusetts General Hospital/Harvard Medicine School, and Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Raymond H Mak
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Camden Bay
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | - Michael Rosenthal
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Burnaby, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, British Colombia, Canada
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Danna M Breen
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Nicolai J Birkbak
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, The Netherlands
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
139
|
Sánchez-Maldonado JM, Cabrera-Serrano AJ, Chattopadhyay S, Campa D, Garrido MDP, Macauda A, Ter Horst R, Jerez A, Netea MG, Li Y, Hemminki K, Canzian F, Försti A, Sainz J. GWAS-Identified Variants for Obesity Do Not Influence the Risk of Developing Multiple Myeloma: A Population-Based Study and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24076029. [PMID: 37047000 PMCID: PMC10094344 DOI: 10.3390/ijms24076029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Multiple myeloma (MM) is an incurable disease characterized by the presence of malignant plasma cells in the bone marrow that secrete specific monoclonal immunoglobulins into the blood. Obesity has been associated with the risk of developing solid and hematological cancers, but its role as a risk factor for MM needs to be further explored. Here, we evaluated whether 32 genome-wide association study (GWAS)-identified variants for obesity were associated with the risk of MM in 4189 German subjects from the German Multiple Myeloma Group (GMMG) cohort (2121 MM cases and 2068 controls) and 1293 Spanish subjects (206 MM cases and 1087 controls). Results were then validated through meta-analysis with data from the UKBiobank (554 MM cases and 402,714 controls) and FinnGen cohorts (914 MM cases and 248,695 controls). Finally, we evaluated the correlation of these single nucleotide polymorphisms (SNPs) with cQTL data, serum inflammatory proteins, steroid hormones, and absolute numbers of blood-derived cell populations (n = 520). The meta-analysis of the four European cohorts showed no effect of obesity-related variants on the risk of developing MM. We only found a very modest association of the POC5rs2112347G and ADCY3rs11676272G alleles with MM risk that did not remain significant after correction for multiple testing (per-allele OR = 1.08, p = 0.0083 and per-allele OR = 1.06, p = 0.046). No correlation between these SNPs and functional data was found, which confirms that obesity-related variants do not influence MM risk.
Collapse
Affiliation(s)
- José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
| | - Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
| | - Subhayan Chattopadhyay
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany
| | - Daniele Campa
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | | | - Angelica Macauda
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Andrés Jerez
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, 08035 Barcelona, Spain
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Germany Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Asta Försti
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| |
Collapse
|
140
|
Queen NJ, Huang W, Komatineni S, Mansour AG, Xiao R, Chrislip LA, Cao L. Social isolation exacerbates diet-induced obesity and peripheral inflammation in young male mice under thermoneutrality. iScience 2023; 26:106259. [PMID: 36915694 PMCID: PMC10006833 DOI: 10.1016/j.isci.2023.106259] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/10/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Social isolation (SI) is associated with an increased risk of mortality and various chronic diseases-including obesity-in humans. Murine studies probing SI metabolic outcomes remain inconsistent, due in part to a lack of consideration for housing temperature. Such experiments typically occur at room temperature, subjecting mice to chronic cold stress. Single housing prevents social thermoregulation, further exacerbating cold stress and obscuring psychosocial influences on metabolism at room temperature. In this study, C57BL/6 and BALB/c male mice were group- and single-housed under thermoneutral conditions to determine whether SI affects the development of high-fat diet-induced obesity. We report SI promotes weight gain, increases food intake, increases adiposity, worsens glycemic control, reduces insulin signaling, exacerbates systemic and adipose inflammatory responses, and induces a molecular signature within the hypothalamus. This study establishes a murine model that recapitulates the SI-induced propensity for obesity, which may further our understanding of SI's influence on health and disease.
Collapse
Affiliation(s)
- Nicholas J. Queen
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Wei Huang
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Suraj Komatineni
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Anthony G. Mansour
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope, National Medical Center and the Beckman Research Institute, Los Angeles, CA 91010, USA
| | - Run Xiao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Logan A. Chrislip
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
141
|
Vourdoumpa A, Paltoglou G, Charmandari E. The Genetic Basis of Childhood Obesity: A Systematic Review. Nutrients 2023; 15:1416. [PMID: 36986146 PMCID: PMC10058966 DOI: 10.3390/nu15061416] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Overweight and obesity in childhood and adolescence represents one of the most challenging public health problems of our century owing to its epidemic proportions and the associated significant morbidity, mortality, and increase in public health costs. The pathogenesis of polygenic obesity is multifactorial and is due to the interaction among genetic, epigenetic, and environmental factors. More than 1100 independent genetic loci associated with obesity traits have been currently identified, and there is great interest in the decoding of their biological functions and the gene-environment interaction. The present study aimed to systematically review the scientific evidence and to explore the relation of single-nucleotide polymorphisms (SNPs) and copy number variants (CNVs) with changes in body mass index (BMI) and other measures of body composition in children and adolescents with obesity, as well as their response to lifestyle interventions. Twenty-seven studies were included in the qualitative synthesis, which consisted of 7928 overweight/obese children and adolescents at different stages of pubertal development who underwent multidisciplinary management. The effect of polymorphisms in 92 different genes was assessed and revealed SNPs in 24 genetic loci significantly associated with BMI and/or body composition change, which contribute to the complex metabolic imbalance of obesity, including the regulation of appetite and energy balance, the homeostasis of glucose, lipid, and adipose tissue, as well as their interactions. The decoding of the genetic and molecular/cellular pathophysiology of obesity and the gene-environment interactions, alongside with the individual genotype, will enable us to design targeted and personalized preventive and management interventions for obesity early in life.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
142
|
Pahl MC, Grant SFA, Leibel RL, Stratigopoulos G. Technologies, strategies, and cautions when deconvoluting genome-wide association signals: FTO in focus. Obes Rev 2023; 24:e13558. [PMID: 36882962 DOI: 10.1111/obr.13558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/08/2022] [Accepted: 01/31/2023] [Indexed: 03/09/2023]
Abstract
Genome-wide association studies have revealed a plethora of genetic variants that correlate with polygenic conditions. However, causal molecular mechanisms have proven challenging to fully define. Without such information, the associations are not physiologically useful or clinically actionable. By reviewing studies of the FTO locus in the genetic etiology of obesity, we wish to highlight advances in the field fueled by the evolution of technical and analytic strategies in assessing the molecular bases for genetic associations. Particular attention is drawn to extrapolating experimental findings from animal models and cell types to humans, as well as technical aspects used to identify long-range DNA interactions and their biological relevance with regard to the associated trait. A unifying model is proposed by which independent obesogenic pathways regulated by multiple FTO variants and genes are integrated at the primary cilium, a cellular antenna where signaling molecules that control energy balance convene.
Collapse
Affiliation(s)
- Matthew C Pahl
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Diabetes and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rudolph L Leibel
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, New York, USA
| | - George Stratigopoulos
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
143
|
Wei Y, Han S, Wen J, Liao J, Liang J, Yu J, Chen X, Xiang S, Huang Z, Zhang B. E26 transformation-specific transcription variant 5 in development and cancer: modification, regulation and function. J Biomed Sci 2023; 30:17. [PMID: 36872348 PMCID: PMC9987099 DOI: 10.1186/s12929-023-00909-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
E26 transformation-specific (ETS) transcription variant 5 (ETV5), also known as ETS-related molecule (ERM), exerts versatile functions in normal physiological processes, including branching morphogenesis, neural system development, fertility, embryonic development, immune regulation, and cell metabolism. In addition, ETV5 is repeatedly found to be overexpressed in multiple malignant tumors, where it is involved in cancer progression as an oncogenic transcription factor. Its roles in cancer metastasis, proliferation, oxidative stress response and drug resistance indicate that it is a potential prognostic biomarker, as well as a therapeutic target for cancer treatment. Post-translational modifications, gene fusion events, sophisticated cellular signaling crosstalk and non-coding RNAs contribute to the dysregulation and abnormal activities of ETV5. However, few studies to date systematically summarized the role and molecular mechanisms of ETV5 in benign diseases and in oncogenic progression. In this review, we specify the molecular structure and post-translational modifications of ETV5. In addition, its critical roles in benign and malignant diseases are summarized to draw a panorama for specialists and clinicians. The updated molecular mechanisms of ETV5 in cancer biology and tumor progression are delineated. Finally, we prospect the further direction of ETV5 research in oncology and its potential translational applications in the clinic.
Collapse
Affiliation(s)
- Yi Wei
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenqi Han
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyuan Wen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shuai Xiang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
144
|
Trinh S, Keller L, Herpertz-Dahlmann B, Seitz J. The role of the brain-derived neurotrophic factor (BDNF) in anorexia nervosa. Psychoneuroendocrinology 2023; 151:106069. [PMID: 36878115 DOI: 10.1016/j.psyneuen.2023.106069] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/28/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023]
Abstract
The brain-derived neurotrophic factor (BDNF) is a growth factor belonging to the neurotrophin family which plays a pivotal role in the differentiation, survival, and plasticity of neurons in the central nervous system. Evidence suggests that BDNF is an important signal molecule in the regulation of energy balance and thus implicated in body weight control. The discovery of BDNF-expressing neurons in the paraventricular hypothalamus which is important in the regulation of energy intake, physical activity, and thermogenesis gives more evidence to the suggested participation of BDNF in eating behavior. Until now it remains questionable whether BDNF can be used as a reliable biomarker for eating disorders such as anorexia nervosa (AN) as available findings on BDNF levels in patients with AN are ambiguous. AN is an eating disorder characterized by a pathological low body weight in combination with a body image disturbance typically developing during adolescence. A severe drive for thinness leads to restrictive eating behavior often accompanied by physical hyperactivity. During therapeutic weight restoration an increase of BDNF expression levels seems desirable as it might improve neuronal plasticity and survival which is essential for learning processes and thereby essential for the success of the psychotherapeutic treatment of patients. On the contrary, the well-known anorexigenic effect of BDNF might favor relapse in patients as soon as the BDNF levels significantly increase during weight rehabilitation. The present review summarizes the association between BDNF and general eating behavior and especially focuses on the eating disorder AN. In this regard findings from preclinical AN studies (activity-based anorexia model) are outlined as well.
Collapse
Affiliation(s)
- Stefanie Trinh
- Institute for Neuroanatomy, University Hospital, RWTH University Aachen, Wendlingweg 2, Aachen D-52074, Germany.
| | - Lara Keller
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH University Aachen, Neuenhofer Weg 21, Aachen D-52074, Germany.
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH University Aachen, Neuenhofer Weg 21, Aachen D-52074, Germany.
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH University Aachen, Neuenhofer Weg 21, Aachen D-52074, Germany.
| |
Collapse
|
145
|
Arrona-Cardoza P, Labonté K, Cisneros-Franco JM, Nielsen DE. The Effects of Food Advertisements on Food Intake and Neural Activity: A Systematic Review and Meta-Analysis of Recent Experimental Studies. Adv Nutr 2023; 14:339-351. [PMID: 36914293 DOI: 10.1016/j.advnut.2022.12.003] [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: 08/03/2022] [Revised: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 01/01/2023] Open
Abstract
Food advertisements are ubiquitous in our daily environment. However, the relationships between exposure to food advertising and outcomes related to ingestive behavior require further investigation. The objective was to conduct a systematic review and meta-analysis of behavioral and neural responses to food advertising in experimental studies. PubMed, Web of Science, and Scopus were searched for articles published from January 2014 to November 2021 using a search strategy following PRISMA guidelines. Experimental studies conducted with human participants were included. A random-effects inverse-variance meta-analysis was performed on standardized mean differences (SMD) of food intake (behavioral outcome) between the food advertisement and nonfood advertisement conditions of each study. Subgroup analyses were performed by age, BMI group, study design, and advertising media type. A seed-based d mapping meta-analysis of neuroimaging studies was performed to evaluate neural activity between experimental conditions. Nineteen articles were eligible for inclusion, 13 for food intake (n = 1303) and 6 for neural activity (n = 303). The pooled analysis of food intake revealed small, but statistically significant, effects of increased intake after viewing food advertising compared with the control condition among adults and children (adult SMD: 0.16; 95% CI: 0.03, 0.28; P = 0.01; I2 = 0; 95% CI: 0, 95.0%; Children SMD: 0.25; 95% CI: 0.14, 0.37; P < 0.0001; I2 = 60.4%; 95% CI: 25.6%, 79.0%). The neuroimaging studies involved children only, and the pooled analysis corrected for multiple comparisons identified one significant cluster, the middle occipital gyrus, with increased activity after food advertising exposure compared with the control condition (peak coordinates: 30, -86, 12; z-value: 6.301, size: 226 voxels; P < 0.001). These findings suggest that acute exposure to food advertising increases food intake among children and adults and that the middle occipital gyrus is an implicated brain region among children. (PROSPERO registration: CRD42022311357).
Collapse
Affiliation(s)
| | - Katherine Labonté
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, Canada
| | - José Miguel Cisneros-Franco
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, Canada; Desautels Faculty of Management, McGill University, Montreal, Canada
| | - Daiva E Nielsen
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, Canada.
| |
Collapse
|
146
|
Kuckuck S, van der Valk ES, Scheurink AJW, van der Voorn B, Iyer AM, Visser JA, Delhanty PJD, van den Berg SAA, van Rossum EFC. Glucocorticoids, stress and eating: The mediating role of appetite-regulating hormones. Obes Rev 2023; 24:e13539. [PMID: 36480471 PMCID: PMC10077914 DOI: 10.1111/obr.13539] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
Disrupted hormonal appetite signaling plays a crucial role in obesity as it may lead to uncontrolled reward-related eating. Such disturbances can be induced not only by weight gain itself but also by glucocorticoid overexposure, for example, due to chronic stress, disease, or medication use. However, the exact pathways are just starting to be understood. Here, we present a conceptual framework of how glucocorticoid excess may impair hormonal appetite signaling and, consequently, eating control in the context of obesity. The evidence we present suggests that counteracting glucocorticoid excess can lead to improvements in appetite signaling and may therefore pose a crucial target for obesity prevention and treatment. In turn, targeting hormonal appetite signals may not only improve weight management and eating behavior but may also decrease detrimental effects of glucocorticoid excess on cardio-metabolic outcomes and mood. We conclude that gaining a better understanding of the relationship between glucocorticoid excess and circulating appetite signals will contribute greatly to improvements in personalized obesity prevention and treatment.
Collapse
Affiliation(s)
- Susanne Kuckuck
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Eline S van der Valk
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Anton J W Scheurink
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Bibian van der Voorn
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Anand M Iyer
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| | - Patric J D Delhanty
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| | - Sjoerd A A van den Berg
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Department of Clinical Chemistry, Erasmus MC, Rotterdam, Netherlands
| | - Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| |
Collapse
|
147
|
Hou J, Chan SF, Wang X, Cai T. Risk prediction with imperfect survival outcome information from electronic health records. Biometrics 2023; 79:190-202. [PMID: 34747010 PMCID: PMC9741856 DOI: 10.1111/biom.13599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Readily available proxies for the time of disease onset such as the time of the first diagnostic code can lead to substantial risk prediction error if performing analyses based on poor proxies. Due to the lack of detailed documentation and labor intensiveness of manual annotation, it is often only feasible to ascertain for a small subset the current status of the disease by a follow-up time rather than the exact time. In this paper, we aim to develop risk prediction models for the onset time efficiently leveraging both a small number of labels on the current status and a large number of unlabeled observations on imperfect proxies. Under a semiparametric transformation model for onset and a highly flexible measurement error model for proxy onset time, we propose the semisupervised risk prediction method by combining information from proxies and limited labels efficiently. From an initially estimator solely based on the labeled subset, we perform a one-step correction with the full data augmenting against a mean zero rank correlation score derived from the proxies. We establish the consistency and asymptotic normality of the proposed semisupervised estimator and provide a resampling procedure for interval estimation. Simulation studies demonstrate that the proposed estimator performs well in a finite sample. We illustrate the proposed estimator by developing a genetic risk prediction model for obesity using data from Mass General Brigham Healthcare Biobank.
Collapse
Affiliation(s)
- Jue Hou
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Stephanie F. Chan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Xuan Wang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Tianxi Cai
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
148
|
Wu T, Xu S. Understanding the contemporary high obesity rate from an evolutionary genetic perspective. Hereditas 2023; 160:5. [PMID: 36750916 PMCID: PMC9903520 DOI: 10.1186/s41065-023-00268-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
The topic of obesity is gaining increasing popularity globally. From an evolutionary genetic perspective, it is believed that the main cause of the high obesity rate is the mismatch between environment and genes after people have shifted toward a modern high-calorie diet. However, it has been debated for over 60 years about how obesity-related genes become prevalent all over the world. Here, we review the three most influential hypotheses or viewpoints, i.e., the thrifty gene hypothesis, the drifty gene hypothesis, and the maladaptation viewpoint. In particular, genome-wide association studies in the recent 10 years have provided rich findings and evidence to be considered for a better understanding of the evolutionary genetic mechanisms of obesity. We anticipate this brief review to direct further studies and inspire the future application of precision medicine in obesity treatment.
Collapse
Affiliation(s)
- Tong Wu
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438, China. .,Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and Ministry of Education Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, 201203, China. .,Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
149
|
Genetic risk score for common obesity and anthropometry in Spanish schoolchildren. ENDOCRINOL DIAB NUTR 2023; 70:107-114. [PMID: 36868927 DOI: 10.1016/j.endien.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/22/2022] [Indexed: 03/05/2023]
Abstract
IntroductionCommon or non-syndromic obesity is a complex polygenic trait conditioned by biallelic or single-base polymorphisms called SNPs (Single-Nucleotide Polymorphisms) that present an additive effect and act synergistically. Most genotype-obese phenotype association studies include body mass index (BMI) or waist-to-height ratio (WtHR), and very few introduce a broad anthropometric profile. ObjectiveTo verify whether a genetic risk score (GRS) developed from 10 SNPs is associated with the obesity phenotype assessed from anthropometric measures indicative of excess weight, adiposity and fat distribution. Material and methodsA series of 438 Spanish schoolchildren (6-16 years old) were evaluated anthropometrically (weight, height, waist circumference, skinfold thickness, BMI, WtHR, body fat percentage [%BF]). Ten SNPs were genotyped from saliva samples, generating a GRS for obesity, establishing genotype-phenotype association. ResultsSchoolchildren categorised as obese by BMI, ICT and %BF had higher GRS than their non-obese peers. The prevalence of overweight and adiposity was higher in subjects with a GRS above the median. Similarly, between 11 and 16 years of age, all anthropometric variables presented higher averages. ConclusionsGRS estimated from the 10 SNPs can be a diagnostic tool for the potential risk of obesity in Spanish schoolchildren and could be useful from the preventive perspective.
Collapse
|
150
|
Hampl SE, Hassink SG, Skinner AC, Armstrong SC, Barlow SE, Bolling CF, Avila Edwards KC, Eneli I, Hamre R, Joseph MM, Lunsford D, Mendonca E, Michalsky MP, Mirza N, Ochoa ER, Sharifi M, Staiano AE, Weedn AE, Flinn SK, Lindros J, Okechukwu K. Clinical Practice Guideline for the Evaluation and Treatment of Children and Adolescents With Obesity. Pediatrics 2023; 151:e2022060640. [PMID: 36622135 DOI: 10.1542/peds.2022-060640] [Citation(s) in RCA: 438] [Impact Index Per Article: 219.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 01/10/2023] Open
|