51
|
Briggs K, Tomar V, Ollberding N, Haberman Y, Bourgonje AR, Hu S, Chaaban L, Sunuwar L, Weersma RK, Denson LA, Melia JMP. Crohn's Disease-Associated Pathogenic Mutation in the Manganese Transporter ZIP8 Shifts the Ileal and Rectal Mucosal Microbiota Implicating Aberrant Bile Acid Metabolism. Inflamm Bowel Dis 2024; 30:1379-1388. [PMID: 38289995 PMCID: PMC11291615 DOI: 10.1093/ibd/izae003] [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: 09/27/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND A pathogenic mutation in the manganese transporter ZIP8 (A391T; rs13107325) increases the risk of Crohn's disease. ZIP8 regulates manganese homeostasis and given the shared need for metals between the host and resident microbes, there has been significant interest in alterations of the microbiome in carriers of ZIP8 A391T. Prior studies have not examined the ileal microbiome despite associations between ileal disease and ZIP8 A391T. METHODS Here, we used the Pediatric Risk Stratification Study (RISK) cohort to perform a secondary analysis of 16S ribosomal RNA gene sequencing data obtained from ileal and rectal mucosa to study associations between ZIP8 A391T carrier status and microbiota composition. RESULTS We found sequence variants mapping to Veillonella were decreased in the ileal mucosa of ZIP8 A391T carriers. Prior human studies have demonstrated the sensitivity of Veillonella to bile acid abundance. We therefore hypothesized that bile acid homeostasis is differentially regulated in carriers of ZIP8 A391T. Using a mouse model of ZIP8 A391T, we demonstrate an increase in total bile acids in the liver and stool and decreased fibroblast growth factor 15 (Fgf15) signaling, consistent with our hypothesis. We confirmed dysregulation of FGF19 in the 1000IBD cohort, finding that plasma FGF19 levels are lower in ZIP8 A391T carriers with ileocolonic Crohn's disease. CONCLUSIONS In the search for genotype-specific therapeutic paradigms for patients with Crohn's disease, these data suggest targeting the FGF19 pathway in ZIP8 A391T carriers. Aberrant bile acid metabolism may precede development of Crohn's disease and prioritize study of the interactions between manganese homeostasis, bile acid metabolism and signaling, and complicated ileal Crohn's disease.
Collapse
Affiliation(s)
- Kristi Briggs
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vartika Tomar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas Ollberding
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yael Haberman
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Sheba Medical Center, Tel-Hashomer, affiliated with Tel Aviv University, Tel Aviv, Israel
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lara Chaaban
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laxmi Sunuwar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lee A Denson
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joanna M P Melia
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
52
|
Kusminski CM, Perez-Tilve D, Müller TD, DiMarchi RD, Tschöp MH, Scherer PE. Transforming obesity: The advancement of multi-receptor drugs. Cell 2024; 187:3829-3853. [PMID: 39059360 PMCID: PMC11286204 DOI: 10.1016/j.cell.2024.06.003] [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: 11/09/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
For more than a century, physicians have searched for ways to pharmacologically reduce excess body fat. The tide has finally turned with recent advances in biochemically engineered agonists for the receptor of glucagon-like peptide-1 (GLP-1) and their use in GLP-1-based polyagonists. These polyagonists reduce body weight through complementary pharmacology by incorporating the receptors for glucagon and/or the glucose-dependent insulinotropic polypeptide (GIP). In their most advanced forms, gut-hormone polyagonists achieve an unprecedented weight reduction of up to ∼20%-30%, offering a pharmacological alternative to bariatric surgery. Along with favorable effects on glycemia, fatty liver, and kidney disease, they also offer beneficial effects on the cardiovascular system and adipose tissue. These new interventions, therefore, hold great promise for the future of anti-obesity medications.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Munich, Germany; German Center for Diabetes Research (DZD) and Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | | | - Matthias H Tschöp
- Helmholtz Munich, Munich, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
53
|
Vo N, Zhang Q, Sung HK. From fasting to fat reshaping: exploring the molecular pathways of intermittent fasting-induced adipose tissue remodeling. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13062. [PMID: 39104461 PMCID: PMC11298356 DOI: 10.3389/jpps.2024.13062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024]
Abstract
Obesity, characterised by excessive fat accumulation, is a complex chronic condition that results from dysfunctional adipose tissue expansion due to prolonged calorie surplus. This leads to rapid adipocyte enlargement that exceeds the support capacity of the surrounding neurovascular network, resulting in increased hypoxia, inflammation, and insulin resistance. Intermittent fasting (IF), a dietary regimen that cycles between periods of fasting and eating, has emerged as an effective strategy to combat obesity and improve metabolic homeostasis by promoting healthy adipose tissue remodeling. However, the precise molecular and cellular mechanisms behind the metabolic improvements and remodeling of white adipose tissue (WAT) driven by IF remain elusive. This review aims to summarise and discuss the relationship between IF and adipose tissue remodeling and explore the potential mechanisms through which IF induces alterations in WAT. This includes several key structural changes, including angiogenesis and sympathetic innervation of WAT. We will also discuss the involvement of key signalling pathways, such as PI3K, SIRT, mTOR, and AMPK, which potentially play a crucial role in IF-mediated metabolic adaptations.
Collapse
Affiliation(s)
- Nathaniel Vo
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Qiwei Zhang
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
54
|
Lim AMW, Lim EU, Chen PL, Fann CSJ. Unsupervised clustering identified clinically relevant metabolic syndrome endotypes in UK and Taiwan Biobanks. iScience 2024; 27:109815. [PMID: 39040048 PMCID: PMC11260869 DOI: 10.1016/j.isci.2024.109815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/02/2024] [Accepted: 04/23/2024] [Indexed: 07/24/2024] Open
Abstract
Metabolic syndrome (MetS) is a collection of cardiovascular risk factors; however, the high prevalence and heterogeneity impede effective clinical management. We conducted unsupervised clustering on individuals from UK Biobank to reveal endotypes. Five MetS subgroups were identified: Cluster 1 (C1): non-descriptive, Cluster 2 (C2): hypertensive, Cluster 3 (C3): obese, Cluster 4 (C4): lipodystrophy-like, and Cluster 5 (C5): hyperglycemic. For all of the endotypes, we identified the corresponding cardiometabolic traits and their associations with clinical outcomes. Genome-wide association studies (GWASs) were conducted to identify associated genotypic traits. We then determined endotype-specific genotypic traits and constructed polygenic risk score (PRS) models specific to each endotype. GWAS of each MetS clusters revealed different genotypic traits. C1 GWAS revealed novel findings of TRIM63, MYBPC3, MYLPF, and RAPSN. Intriguingly, C1, C3, and C4 were associated with genes highly expressed in brain tissues. MetS clusters with comparable phenotypic and genotypic traits were identified in Taiwan Biobank.
Collapse
Affiliation(s)
- Aylwin Ming Wee Lim
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 112304, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- ASUS Intelligent Cloud Services (AICS), Taipei 112, Taiwan
| | - Evan Unit Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- College of Computing, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Cathy Shen Jang Fann
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 112304, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
55
|
Bai Z, Hao J, Chen M, Yao K, Zheng L, Liu L, Hu J, Guo K, Lv Y, Li F. Integrating plasma proteomics with genome-wide association data to identify novel drug targets for inflammatory bowel disease. Sci Rep 2024; 14:16251. [PMID: 39009667 PMCID: PMC11250821 DOI: 10.1038/s41598-024-66780-w] [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: 02/27/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disease that includes Crohn's disease (CD) and ulcerative colitis (UC). Although genome-wide association studies (GWASs) have identified many relevant genetic risk loci, the impact of these loci on protein abundance and their potential utility as clinical therapeutic targets remain uncertain. Therefore, this study aimed to investigate the pathogenesis of IBD and identify effective therapeutic targets through a comprehensive and integrated analysis. We systematically integrated GWAS data related to IBD, UC and CD (N = 25,305) by the study of de Lange KM with the human blood proteome (N = 7213) by the Atherosclerosis Risk in Communities (ARIC) study. Proteome-wide association study (PWAS), mendelian randomisation (MR) and Bayesian colocalisation analysis were used to identify proteins contributing to the risk of IBD. Integrative analysis revealed that genetic variations in IBD, UC and CD affected the abundance of five (ERAP2, RIPK2, TALDO1, CADM2 and RHOC), three (VSIR, HGFAC and CADM2) and two (MST1 and FLRT3) cis-regulated plasma proteins, respectively (P < 0.05). Among the proteins identified via Bayesian colocalisation analysis, CADM2 was found to be an important common protein between IBD and UC. A drug and five druggable target genes were identified from DGIdb after Bayesian colocalisation analysis. Our study's findings from genetic and proteomic approaches have identified compelling proteins that may serve as important leads for future functional studies and potential drug targets for IBD (UC and CD).
Collapse
Affiliation(s)
- Zhongyuan Bai
- First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Jiawei Hao
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Miaoran Chen
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Kaixin Yao
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Leilei Zheng
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Liu Liu
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Jingxi Hu
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Kaiqing Guo
- Hepatobiliary Pancreatogastric Surgery, Shanxi Province Cancer Hospital, Taiyuan, China.
- Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China.
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Yongqiang Lv
- Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China.
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
- Department of Scientific Research, Shanxi Province Cancer Hospital, Taiyuan, China.
| | - Feng Li
- Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China.
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
- Central Laboratory, Shanxi Province Cancer Hospital, Taiyuan, China.
| |
Collapse
|
56
|
Styrkarsdottir U, Tragante V, Stefansdottir L, Thorleifsson G, Oddsson A, Sørensen E, Erikstrup C, Schwarz P, Jørgensen HL, Lauritzen JB, Brunak S, Knowlton KU, Nadauld LD, Ullum H, Pedersen OBV, Ostrowski SR, Holm H, Gudbjartsson DF, Sulem P, Stefansson K. Obesity Variants in the GIPR Gene Are not Associated With Risk of Fracture or Bone Mineral Density. J Clin Endocrinol Metab 2024; 109:e1608-e1615. [PMID: 38118020 PMCID: PMC11244190 DOI: 10.1210/clinem/dgad734] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/22/2023]
Abstract
CONTEXT It is not clear if antagonizing the GIP (glucose-dependent insulinotropic polypeptide) receptor (GIPR) for treatment of obesity is likely to increase the risk of fractures, or to lower bone mineral density (BMD) beyond what is expected with rapid weight loss. OBJECTIVE The objective of this study was to investigate the risk of fracture and BMD of sequence variants in GIPR that reduce the activity of the GIP receptor and have been associated with reduced body mass index (BMI). METHODS We analyzed the association of 3 missense variants in GIPR, a common variant, rs1800437 (p.Glu354Gln), and 2 rare variants, rs139215588 (p.Arg190Gln) and rs143430880 (p.Glu288Gly), as well as a burden of predicted loss-of-function (LoF) variants with risk of fracture and with BMD in a large meta-analysis of up to 1.2 million participants. We analyzed associations with fractures at different skeletal sites in the general population: any fractures, hip fractures, vertebral fractures and forearm fractures, and specifically nonvertebral and osteoporotic fractures in postmenopausal women. We also evaluated associations with BMD at the lumbar spine, femoral neck, and total body measured with dual-energy x-ray absorptiometry (DXA), and with BMD estimated from heel ultrasound (eBMD). RESULTS None of the 3 missense variants in GIPR was significantly associated with increased risk of fractures or with lower BMD. Burden of LoF variants in GIPR was not associated with fractures or with BMD measured with clinically validated DXA, but was associated with eBMD. CONCLUSION Missense variants in GIPR, or burden of LoF variants in the gene, are not associated with risk of fractures or with lower BMD.
Collapse
Affiliation(s)
| | - Vinicius Tragante
- Population Genomics, deCODE genetics/Amgen Inc, Reykjavik 102, Iceland
| | | | | | - Asmundur Oddsson
- Population Genomics, deCODE genetics/Amgen Inc, Reykjavik 102, Iceland
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Peter Schwarz
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Endocrinology, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Henrik Løvendahl Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Clinical Biochemistry, Amager Hvidovre Hospital, Copenhagen 2650, Denmark
| | - Jes Bruun Lauritzen
- Department of Orthopedic Surgery, Bispebjerg Hospital, University of Copenhagen, Copenhagen 2400, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kirk U Knowlton
- Intermountain Health, Heart Institute, Salt Lake City, UT 84143, USA
| | | | - Henrik Ullum
- Statens Serum Institut, Copenhagen 2300, Denmark
| | - Ole Birger Vesterager Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge 4600, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Hilma Holm
- Population Genomics, deCODE genetics/Amgen Inc, Reykjavik 102, Iceland
| | - Daniel F Gudbjartsson
- Population Genomics, deCODE genetics/Amgen Inc, Reykjavik 102, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik 102, Iceland
| | - Patrick Sulem
- Population Genomics, deCODE genetics/Amgen Inc, Reykjavik 102, Iceland
| | - Kari Stefansson
- Population Genomics, deCODE genetics/Amgen Inc, Reykjavik 102, Iceland
- Faculty of Medicine, School of Health Science, University of Iceland, Reykjavik 102, Iceland
| |
Collapse
|
57
|
Tomar V, Kang J, Lin R, Brant SR, Lazarev M, Tressler C, Glunde K, Zachara N, Melia J. Aberrant N-glycosylation is a therapeutic target in carriers of a common and highly pleiotropic mutation in the manganese transporter ZIP8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601207. [PMID: 39005453 PMCID: PMC11244875 DOI: 10.1101/2024.06.28.601207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The treatment of defective glycosylation in clinical practice has been limited to patients with rare and severe phenotypes associated with congenital disorders of glycosylation (CDG). Carried by approximately 5% of the human population, the discovery of the highly pleiotropic, missense mutation in a manganese transporter ZIP8 has exposed under-appreciated roles for Mn homeostasis and aberrant Mn-dependent glycosyltransferases activity leading to defective N-glycosylation in complex human diseases. Here, we test the hypothesis that aberrant N-glycosylation contributes to disease pathogenesis of ZIP8 A391T-associated Crohn's disease. Analysis of N-glycan branching in intestinal biopsies demonstrates perturbation in active Crohn's disease and a genotype-dependent effect characterized by increased truncated N-glycans. A mouse model of ZIP8 391-Thr recapitulates the intestinal glycophenotype of patients carrying mutations in ZIP8. Borrowing from therapeutic strategies employed in the treatment of patients with CDGs, oral monosaccharide therapy with N-acetylglucosamine ameliorates the epithelial N-glycan defect, bile acid dyshomeostasis, intestinal permeability, and susceptibility to chemical-induced colitis in a mouse model of ZIP8 391-Thr. Together, these data support ZIP8 391-Thr alters N-glycosylation to contribute to disease pathogenesis, challenging the clinical paradigm that CDGs are limited to patients with rare diseases. Critically, the defect in glycosylation can be targeted with monosaccharide supplementation, providing an opportunity for genotype-driven, personalized medicine.
Collapse
|
58
|
Hochner H, Butterman R, Margaliot I, Friedlander Y, Linial M. Obesity risk in young adults from the Jerusalem Perinatal Study (JPS): the contribution of polygenic risk and early life exposure. Int J Obes (Lond) 2024; 48:954-963. [PMID: 38472354 PMCID: PMC11216986 DOI: 10.1038/s41366-024-01505-7] [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: 10/11/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND/OBJECTIVES The effects of early life exposures on offspring life-course health are well established. This study assessed whether adding early socio-demographic and perinatal variables to a model based on polygenic risk score (PRS) improves prediction of obesity risk. METHODS We used the Jerusalem Perinatal study (JPS) with data at birth and body mass index (BMI) and waist circumference (WC) measured at age 32. The PRS was constructed using over 2.1M common SNPs identified in genome-wide association study (GWAS) for BMI. Linear and logistic models were applied in a stepwise approach. We first examined the associations between genetic variables and obesity-related phenotypes (e.g., BMI and WC). Secondly, socio-demographic variables were added and finally perinatal exposures, such as maternal pre-pregnancy BMI (mppBMI) and gestational weight gain (GWG) were added to the model. Improvement in prediction of each step was assessed using measures of model discrimination (area under the curve, AUC), net reclassification improvement (NRI) and integrated discrimination improvement (IDI). RESULTS One standard deviation (SD) change in PRS was associated with a significant increase in BMI (β = 1.40) and WC (β = 2.45). These associations were slightly attenuated (13.7-14.2%) with the addition of early life exposures to the model. Also, higher mppBMI was associated with increased offspring BMI (β = 0.39) and WC (β = 0.79) (p < 0.001). For obesity (BMI ≥ 30) prediction, the addition of early socio-demographic and perinatal exposures to the PRS model significantly increased AUC from 0.69 to 0.73. At an obesity risk threshold of 15%, the addition of early socio-demographic and perinatal exposures to the PRS model provided a significant improvement in reclassification of obesity (NRI, 0.147; 95% CI 0.068-0.225). CONCLUSIONS Inclusion of early life exposures, such as mppBMI and maternal smoking, 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, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachely Butterman
- Braun School of Public Health, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ido Margaliot
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Yechiel Friedlander
- Braun School of Public Health, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Michal Linial
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| |
Collapse
|
59
|
Pedroza Matute S, Turvey K, Iyavoo S. Advancing human genotyping: The Infinium HTS iSelect Custom microarray panel (Rita) development study. Forensic Sci Int Genet 2024; 71:103049. [PMID: 38653142 DOI: 10.1016/j.fsigen.2024.103049] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Single Nucleotide Polymorphisms (SNPs), as the most prevalent type of variation in the human genome, play a pivotal role in influencing human traits. They are extensively utilized in diverse fields such as population genetics, forensic science, and genetic medicine. This study focuses on the 'Rita' BeadChip, a custom SNP microarray panel developed using Illumina Infinium HTS technology. Designed for high-throughput genotyping, the panel facilitates the analysis of over 4000 markers efficiently and cost-effectively. After careful clustering performed on a set of 1000 samples, an evaluation of the Rita panel was undertaken, assessing its sensitivity, repeatability, reproducibility, precision, accuracy, and resistance to contamination. The panel's performance was evaluated in various scenarios, including sex estimation and parental relationship assessment, using GenomeStudio data analysis software. Findings show that over 95 % of the custom BeadChip assay markers were successful, with better performance of transitions over other mutations, and a considerably lower success rate for Y chromosome loci. An exceptional call rate exceeding 99 % was demonstrated for control samples, even with DNA input as low as 0.781 ng. Call rates above 80 % were still obtained with DNA quantities under 0.1 ng, indicating high sensitivity and suitability for forensic applications where DNA quantity is often limited. Repeatability, reproducibility, and precision studies revealed consistency of the panel's performance across different batches and operators, with no significant deviations in call rates or genotyping results. Accuracy assessments, involving comparison with multiple available genetic databases, including the 1000 Genome Project and HapMap, denoted over 99 % concordance, establishing the Rita panel's reliability in genotyping. The contamination study revealed insights into background noise and allowed the definition of thresholds for sample quality evaluation. Multiple metrics for differentiating between negative controls and true samples were highlighted, increasing the reliability of the obtained results. The sex estimation tool in GenomeStudio proved highly effective, correctly assigning sex in all samples with autosomal loci call rates above 97 %. The parental relationship assessment of family trios highlighted the utility of GenomeStudio in identifying genotyping errors or potential Mendelian inconsistencies, promoting the application of arrays such as Rita in kinship testing. Overall, this evaluation confirms the Rita microarray as a robust, high-throughput genotyping tool, underscoring its potential in genetic research and forensic applications. With its custom content and adaptable design, it not only meets current genotyping demands but also opens avenues for further research and application expansion in the field of genetic analysis.
Collapse
Affiliation(s)
| | - Kiera Turvey
- IDna Genetics Limited, Scottow Enterprise Park, Norwich, Norfolk NR10 5FB, United Kingdom
| | - Sasitaran Iyavoo
- IDna Genetics Limited, Scottow Enterprise Park, Norwich, Norfolk NR10 5FB, United Kingdom; School of Chemistry, College of Health and Science, University of Lincoln, Lincoln, Lincolnshire LN6 7TS, United Kingdom.
| |
Collapse
|
60
|
Huang T, Zhang X, Li Q, Li X, Yao J, Song J, Chen Y, Ye L, Li C, Xiran P, Wen Y. The Association between Obesity Susceptibility and Polymorphisms of MC4R, SH2B1, and NEGR1 in Tibetans. Genet Test Mol Biomarkers 2024; 28:267-274. [PMID: 39034913 DOI: 10.1089/gtmb.2023.0546] [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] [Indexed: 07/23/2024] Open
Abstract
Background: A high-altitude environment has inhibitory effects on obesity. Tibetans are not a high-risk population for obesity, but there are still obese individuals within that population. Obesity has become a worldwide health problem, and previous studies have found that obesity is closely associated with hereditary factors. Few studies have investigated obesity in Tibetans, and the association between gene polymorphisms and obesity in Tibetans remains unclear. Methods: Our study investigated the fat mass of 140 native Tibetan individuals (70 men and 70 women) from Lhasa and analyzed the associations between polymorphisms of melanocortin 4 receptor (MC4R), Src homology 2B adapter protein 1 (SH2B1), and neuronal growth regulator 1 (NEGR1) and obesity. Result: Among Tibetan individuals, there were differences in genotype and allele frequencies between those in the obesity group and those in the healthy group at MC4R (rs17782313) and SH2B1 (rs7359397). The polymorphisms of MC4R (rs17782313) were associated with fat mass and obesity in Tibetan men and women, and there was an association between SH2B1 (rs7359397) polymorphisms and fat mass and obesity in Tibetan men. However, polymorphisms of NEGR1 (rs3101336) were not associated with fat mass or obesity in Tibetan individuals. Conclusion: Among Tibetan individuals, polymorphisms of MC4R (rs17782313) and SH2B1 (rs7359397) were associated with obesity, but NEGR1 (rs3101336) polymorphisms were not associated with obesity.
Collapse
Affiliation(s)
- Ting Huang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Xianpeng Zhang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Qiang Li
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Wuwei Occupational College, Wuwei, China
| | - Xin Li
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Jie Yao
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Jia Song
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Ying Chen
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Liping Ye
- Department of Radiology, Nagqu District People's Hospital, Nagqu, China
| | - Chunshan Li
- Department of Radiology, Nagqu District People's Hospital, Nagqu, China
| | - Pingcuo Xiran
- Maternal and Children Health Care Hospital of Ali District, Ali, China
| | - Youfeng Wen
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
61
|
Eghbali M, Mottaghi A, Taghizadeh S, Cheraghi S. Genetic Variants in the Fat Mass and Obesity-Associated Gene and Risk of Obesity/Overweight in Children and Adolescents: A Systematic Review and Meta-Analysis. Endocrinol Diabetes Metab 2024; 7:e00510. [PMID: 38973101 PMCID: PMC11227992 DOI: 10.1002/edm2.510] [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: 05/07/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 07/09/2024] Open
Abstract
OBJECTIVE The variations in the single-nucleotide polymorphisms (SNPs) of the fat mass and obesity (FTO)-associated gene have been linked to being overweight or obese in children. In this research a thorough examination was performed to elucidate the connection between various FTO gene SNPs and overweight or obesity in children and adolescents. METHOD We searched PubMed, Google scholar, Web of Science and Scopus until January 2024 to find studies that investigate the association between different SNPs of FTO gene and the risk of overweight/obesity in children and adolescents. After filtering the relevant studies, meta-analysis was used to quantify the association of FTO gene SNPs within different genetic inheritance models. RESULTS We have identified 32 eligible studies with 14,930 obese/overweight cases and 24,765 healthy controls. Our recessive model showed a significant association with rs9939609 (OR: 1.56, 95% CI: 1.20; 2.02, p < 0.01) and rs1421085 (OR: 1.77, 95% CI: 1.14; 2.75, p < 0.01). Besides, in the homozygote model, rs1421085 showed the highest association (OR: 2.32, 95% CI: 1.38; 3.89, p < 0.01) with the risk of obesity in a population of children and adolescents. Moreover, there are other SNPs of FTO genes, such as rs9921255, rs9928094 and rs9930333, which showed a positive association with obesity and overweight. However, their effects were evaluated in very few numbers of studies. CONCLUSION In this study, we have found that the FTO rs9939609 and rs1421085 are associated to an increased risk of obesity among children and adolescents. Besides, the findings of this study further reaffirmed the established link between rs9939609 and obesity in children and adolescents.
Collapse
Affiliation(s)
- Maryam Eghbali
- Endocrine Research Center, Institute of Endocrinology and MetabolismIran University of Medical SciencesTehranIran
| | - Azadeh Mottaghi
- Research Center for Prevention of Cardiovascular Diseases, Endocrinology & Metabolism, Institute of Endocrinology MetabolismIran University of Medical SciencesTehranIran
| | - Sara Taghizadeh
- Translational Ophthalmology Research CenterTehran University of Medical SciencesTehranIran
| | - Sara Cheraghi
- Endocrine Research Center, Institute of Endocrinology and MetabolismIran University of Medical SciencesTehranIran
| |
Collapse
|
62
|
Xu H, Gupta S, Dinsmore I, Kollu A, Cawley AM, Anwar MY, Chen HH, Petty LE, Seshadri S, Graff M, Below P, Brody JA, Chittoor G, Fisher-Hoch SP, Heard-Costa NL, Levy D, Lin H, Loos RJF, Mccormick JB, Rotter JI, Mirshahi T, Still CD, Destefano A, Cupples LA, Mohlke KL, North KE, Justice AE, Liu CT. Integrating Genetic and Transcriptomic Data to Identify Genes Underlying Obesity Risk Loci. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.11.24308730. [PMID: 38903089 PMCID: PMC11188121 DOI: 10.1101/2024.06.11.24308730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Genome-wide association studies (GWAS) have identified numerous body mass index (BMI) loci. However, most underlying mechanisms from risk locus to BMI remain unknown. Leveraging omics data through integrative analyses could provide more comprehensive views of biological pathways on BMI. We analyzed genotype and blood gene expression data in up to 5,619 samples from the Framingham Heart Study (FHS). Using 3,992 single nucleotide polymorphisms (SNPs) at 97 BMI loci and 20,692 transcripts within 1 Mb, we performed separate association analyses of transcript with BMI and SNP with transcript (PBMI and PSNP, respectively) and then a correlated meta-analysis between the full summary data sets (PMETA). We identified transcripts that met Bonferroni-corrected significance for each omic, were more significant in the correlated meta-analysis than each omic, and were at least nominally associated with BMI in FHS data. Among 308 significant SNP-transcript-BMI associations, we identified seven genes (NT5C2, GSTM3, SNAPC3, SPNS1, TMEM245, YPEL3, and ZNF646) in five association regions. Using an independent sample of blood gene expression data, we validated results for SNAPC3 and YPEL3. We tested for generalization of these associations in hypothalamus, nucleus accumbens, and liver and observed significant (PMETA<0.05 & PMETA
Collapse
Affiliation(s)
- Hanfei Xu
- Department of Biostatistics, School of Public Health, Boston University, 801 Massachusettes Ave, Boston, MA, 02118, USA
| | - Shreyash Gupta
- Department of Population Health Sciences, Geisinger, 100 N. Academy Ave., Danville, PA, 17822, USA
| | - Ian Dinsmore
- Department of Genomic Health, Geisinger, 100 N. Academy Ave., Danville, PA, 17822, USA
| | - Abbey Kollu
- Department of Psychology and Neuroscience, University of North Carolina, 235 E. Cameron Avenue, Chapel Hill, NC, 27599, USA
| | - Anne Marie Cawley
- Marsico Lung Institute, University of North Carolina, 125 Mason Farm Rd, Chapel Hill, NC, 27599, USA
| | - Mohammad Y. Anwar
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Hung-Hsin Chen
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Section 2, Academia Rd., Taipei, Nangang District, 115201, Taiwan
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, 1161 21st Ave S, Nashville, TN, 37232, USA
| | - Lauren E. Petty
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, 1161 21st Ave S, Nashville, TN, 37232, USA
| | - Sudha Seshadri
- Department of Biostatistics, School of Public Health, Boston University, 801 Massachusettes Ave, Boston, MA, 02118, USA
- Department of Neurology, School of Medicine, Boston University, 85 East Concord Street, Boston, MA, 02118, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, UT Health San Antonio, 8300 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Misa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Piper Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, 1161 21st Ave S, Nashville, TN, 37232, USA
| | - Jennifer A. Brody
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, 1730 Minor Ave, Seattle, WA, 98101, USA
| | - Geetha Chittoor
- Department of Population Health Sciences, Geisinger, 100 N. Academy Ave., Danville, PA, 17822, USA
| | - Susan P. Fisher-Hoch
- Department of Epidemiology, School of Public Health, UT Health Houston, Regional Academic Health Center, One West University Blvd, Brownsville, TX, 78520, USA
| | - Nancy L. Heard-Costa
- Framingham Heart Study, 73 Mt Wayte Ave, Framingham, MA, 01702, USA
- Department of Neurology, Chobanian & Avedisian School of Medicine, Boston University, 72 E Concord St, Boston, MA, 02118, USA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute of the National Institutes of Health, 6701 Rockledge Drive, Bethesda, MD, 20892, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, 55 N Lake Ave, Worcester, MA, 01655, USA
| | - Ruth JF. Loos
- Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, 2200, Copenhagen, Denmark
| | - Joseph B. Mccormick
- Department of Epidemiology, School of Public Health, UT Health Houston, Regional Academic Health Center, One West University Blvd, Brownsville, TX, 78520, USA
| | - Jerome I. Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA, 90502, USA
| | - Tooraj Mirshahi
- Department of Genomic Health, Geisinger, 100 N. Academy Ave., Danville, PA, 17822, USA
| | - Christopher D. Still
- Center for Obesity and Metabolic Health, Geisinger, 100 N. Academy Ave., Danville, PA, 17822, USA
| | - Anita Destefano
- Department of Biostatistics, School of Public Health, Boston University, 801 Massachusettes Ave, Boston, MA, 02118, USA
- Department of Neurology, School of Medicine, Boston University, 85 East Concord Street, Boston, MA, 02118, USA
| | - L. Adrienne Cupples
- Department of Biostatistics, School of Public Health, Boston University, 801 Massachusettes Ave, Boston, MA, 02118, USA
| | - Karen L Mohlke
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kari E. North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, 135 Dauer Drive, Chapel Hill, NC, 27599, USA
| | - Anne E. Justice
- Department of Population Health Sciences, Geisinger, 100 N. Academy Ave., Danville, PA, 17822, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, 801 Massachusettes Ave, Boston, MA, 02118, USA
| |
Collapse
|
63
|
Zhang M, Ward J, Strawbridge RJ, Celis-Morales C, Pell JP, Lyall DM, Ho FK. How do lifestyle factors modify the association between genetic predisposition and obesity-related phenotypes? A 4-way decomposition analysis using UK Biobank. BMC Med 2024; 22:230. [PMID: 38853248 PMCID: PMC11163778 DOI: 10.1186/s12916-024-03436-6] [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: 12/14/2023] [Accepted: 05/22/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Obesity and central obesity are multifactorial conditions with genetic and non-genetic (lifestyle and environmental) contributions. There is incomplete understanding of whether lifestyle modifies the translation from respective genetic risks into phenotypic obesity and central obesity, and to what extent genetic predisposition to obesity and central obesity is mediated via lifestyle factors. METHODS This is a cross-sectional study of 201,466 (out of approximately 502,000) European participants from UK Biobank and tested for interactions and mediation role of lifestyle factors (diet quality; physical activity levels; total energy intake; sleep duration, and smoking and alcohol intake) between genetic risk for obesity and central obesity. BMI-PRS and WHR-PRS are exposures and obesity and central obesity are outcomes. RESULTS Overall, 42.8% of the association between genetic predisposition to obesity and phenotypic obesity was explained by lifestyle: 0.9% by mediation and 41.9% by effect modification. A significant difference between men and women was found in central obesity; the figures were 42.1% (association explained by lifestyle), 1.4% (by mediation), and 40.7% (by modification) in women and 69.6% (association explained by lifestyle), 3.0% (by mediation), and 66.6% (by modification) in men. CONCLUSIONS A substantial proportion of the association between genetic predisposition to obesity/central obesity and phenotypic obesity/central obesity was explained by lifestyles. Future studies with repeated measures of obesity and lifestyle would be needed to clarify causation.
Collapse
Affiliation(s)
- Mengrong Zhang
- School of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byers Road, Glasgow, G12 8TB, UK
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byers Road, Glasgow, G12 8TB, UK
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byers Road, Glasgow, G12 8TB, UK
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Carlos Celis-Morales
- School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, UK
- Human Performance Lab, Education, Physical Activity, and Health Research Unit, Universidad Católica del Maule, Talca, Chile
- Centro de Investigación en Medicina de Altura (CEIMA), Universidad Arturo Prat, Iquique, Chile
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byers Road, Glasgow, G12 8TB, UK
| | - Donald M Lyall
- School of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byers Road, Glasgow, G12 8TB, UK
| | - Frederick K Ho
- School of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byers Road, Glasgow, G12 8TB, UK.
| |
Collapse
|
64
|
Morrow NM, Morissette A, Mulvihill EE. Immunomodulation and inflammation: Role of GLP-1R and GIPR expressing cells within the gut. Peptides 2024; 176:171200. [PMID: 38555054 DOI: 10.1016/j.peptides.2024.171200] [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: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are peptide hormones produced by enteroendocrine cells in the small intestine. Despite being produced in the gut, the leveraging of their role in potentiating glucose-stimulated insulin secretion, also known as the incretin effect, has distracted from discernment of direct intestinal signaling circuits. Both preclinical and clinical evidence have highlighted a role for the incretins in inflammation. In this review, we highlight the discoveries of GLP-1 receptor (GLP-1R)+ natural (TCRαβ and TCRγδ) and induced (TCRαβ+CD4+ cells and TCRαβ+CD8αβ+) intraepithelial lymphocytes. Both endogenous signaling and pharmacological activation of GLP-1R impact local and systemic inflammation, the gut microbiota, whole-body metabolism, as well as the control of GLP-1 bioavailability. While GIPR signaling has been documented to impact hematopoiesis, the impact of these bone marrow-derived cells in gut immunology is not well understood. We uncover gaps in the literature of the evaluation of the impact of sex in these GLP-1R and GIP receptor (GIPR) signaling circuits and provide speculations of the maintenance roles these hormones play within the gut in the fasting-refeeding cycles. GLP-1R agonists and GLP-1R/GIPR agonists are widely used as treatments for diabetes and weight loss, respectively; however, their impact on gut homeostasis has not been fully explored. Advancing our understanding of the roles of GLP-1R and GIPR signaling within the gut at homeostasis as well as metabolic and inflammatory diseases may provide targets to improve disease management.
Collapse
Affiliation(s)
- Nadya M Morrow
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario KIY 4W7, Canada; Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Faculty of Medicine, 451 Smyth Rd, Ottawa, Ontario K1H 8L1, Canada
| | - Arianne Morissette
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario KIY 4W7, Canada
| | - Erin E Mulvihill
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario KIY 4W7, Canada; Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Faculty of Medicine, 451 Smyth Rd, Ottawa, Ontario K1H 8L1, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada; Montreal Diabetes Research Group, Montreal, Quebec, Canada; Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada.
| |
Collapse
|
65
|
Liskiewicz A, Müller TD. Regulation of energy metabolism through central GIPR signaling. Peptides 2024; 176:171198. [PMID: 38527521 DOI: 10.1016/j.peptides.2024.171198] [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: 01/31/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
In recent years, significant progress has been made to pharmacologically combat the obesity pandemic, particularly with regard to biochemically tailored drugs that simultaneously target the receptors for glucagon-like peptide-1 (GLP-1) and the glucose-dependent insulinotropic polypeptide (GIP). But while the pharmacological benefits of GLP-1 receptor (GLP-1R) agonism are widely acknowledged, the role of the GIP system in regulating systems metabolism remains controversial. When given in adjunct to GLP-1R agonism, both agonism and antagonism of the GIP receptor (GIPR) improves metabolic outcome in preclinical and clinical studies, and despite persistent concerns about its potential obesogenic nature, there is accumulating evidence indicating that GIP has beneficial metabolic effects via central GIPR agonism. Nonetheless, despite growing recognition of the GIP system as a valuable pharmacological target, there remains great uncertainty as to where and how GIP acts in the brain to regulate metabolism, and how GIPR agonism may differ from GIPR antagonism in control of energy metabolism. In this review we highlight current knowledge on the central action of GIP, and discuss open questions related to its multifaceted biology in the brain and the periphery.
Collapse
Affiliation(s)
- Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.
| |
Collapse
|
66
|
Liu N, Pang B, Kang L, Li D, Jiang X, Zhou CM. TUFM in health and disease: exploring its multifaceted roles. Front Immunol 2024; 15:1424385. [PMID: 38868764 PMCID: PMC11167084 DOI: 10.3389/fimmu.2024.1424385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
The nuclear-encoded mitochondrial protein Tu translation elongation factor, mitochondrial (TUFM) is well-known for its role in mitochondrial protein translation. Originally discovered in yeast, TUFM demonstrates significant evolutionary conservation from prokaryotes to eukaryotes. Dysregulation of TUFM has been associated with mitochondrial disorders. Although early hypothesis suggests that TUFM is localized within mitochondria, recent studies identify its presence in the cytoplasm, with this subcellular distribution being linked to distinct functions of TUFM. Significantly, in addition to its established function in mitochondrial protein quality control, recent research indicates a broader involvement of TUFM in the regulation of programmed cell death processes (e.g., autophagy, apoptosis, necroptosis, and pyroptosis) and its diverse roles in viral infection, cancer, and other disease conditions. This review seeks to offer a current summary of TUFM's biological functions and its complex regulatory mechanisms in human health and disease. Insight into these intricate pathways controlled by TUFM may lead to the potential development of targeted therapies for a range of human diseases.
Collapse
Affiliation(s)
- Ning Liu
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bo Pang
- The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Longfei Kang
- The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dongyun Li
- The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xia Jiang
- The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuan-min Zhou
- The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
67
|
Novikoff A, Müller TD. Pharmacological Advances in Incretin-Based Polyagonism: What We Know and What We Don't. Physiology (Bethesda) 2024; 39:142-156. [PMID: 38353610 PMCID: PMC11368522 DOI: 10.1152/physiol.00032.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/21/2024] Open
Abstract
The prevalence of obesity continues to rise in both adolescents and adults, in parallel obesity is strongly associated with the increased incidence of type 2 diabetes, heart failure, certain types of cancer, and all-cause mortality. In relation to obesity, many pharmacological approaches of the past have tried and failed to combat the rising obesity epidemic, particularly due to insufficient efficacy or unacceptable side effects. However, while the history of antiobesity medication is plagued by failures and disappointments, we have witnessed over the last 10 years substantial progress, particularly in regard to biochemically optimized agonists at the receptor for glucagon-like peptide-1 (GLP-1R) and unimolecular coagonists at the receptors for GLP-1 and the glucose-dependent insulinotropic polypeptide (GIP). Although the GIP receptor:GLP-1R coagonists are being heralded as premier pharmacological tools for the treatment of obesity and diabetes, uncertainty remains as to why these drugs testify superiority over best-in-class GLP-1R monoagonists. Particularly with regard to GIP, there remains great uncertainty if and how GIP acts on systems metabolism and if the GIP system should be activated or inhibited to improve metabolic outcome in adjunct to GLP-1R agonism. In this review, we summarize recent advances in GLP-1- and GIP-based pharmacology and discuss recent findings and open questions related to how the GIP system affects systemic energy and glucose metabolism.
Collapse
Affiliation(s)
- Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
68
|
Chermon D, Birk R. Brain-derived neurotrophic factor gene rs925946 associates with Israeli females' obesity predisposition: An interaction between genetics, eating habits, and physical inactivity. Nutr Res 2024; 125:61-68. [PMID: 38503023 DOI: 10.1016/j.nutres.2024.02.005] [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: 07/02/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 03/21/2024]
Abstract
The global obesity pandemic presents a pressing health challenge, with an increasing prevalence shaped by an intricate interplay of genetics and environment. Brain-derived neurotrophic factor (BDNF) plays a pivotal role in regulating feeding behavior and energy expenditure. BDNF single nucleotide polymorphisms have been linked to obesity risk. We hypothesized that BDNF rs925946 is positively associated with obesity susceptibility in the Israeli population. We aimed to study BDNF rs925946 association with obesity susceptibility and its interaction with environmental factors, including eating habits, sugar-sweetened beverages, and physical activity. A data cohort of 4668 Israeli adults (≥18 years, Jewish) was analyzed. Participants' genotypic data for the BDNF rs925946 and lifestyle and eating behavior questionnaire data were analyzed for the association between obesity predisposition and gene-environment interactions. Female (n = 3259) BDNF rs925946 T-allele carriers had an elevated obesity odd (odds ratio [OR] = 1.2; 95% confidence interval [CI], 1.03-1.4, P = .02). BDNF rs925946 genotype interacted significantly with physical inactivity, sugar-sweetened beverage consumption, and eating habits score to enhance obesity odds (OR = 1.4; 95% CI, 1.14-1.7; OR = 1.54, 95% CI, 1.1-2.15; and OR = 1.4; 95% CI, 1.2-2.11, respectively). Our data demonstrated a significant association between BDNF rs925946 T-allele female carriers and a higher obesity predisposition, affected by modifiable lifestyle factors.
Collapse
Affiliation(s)
- Danyel Chermon
- Nutrition Department, Health Sciences Faculty, Ariel University, 407000, Israel
| | - Ruth Birk
- Nutrition Department, Health Sciences Faculty, Ariel University, 407000, Israel.
| |
Collapse
|
69
|
Chermon D, Birk R. Deciphering the Interplay between Genetic Risk Scores and Lifestyle Factors on Individual Obesity Predisposition. Nutrients 2024; 16:1296. [PMID: 38732542 PMCID: PMC11085817 DOI: 10.3390/nu16091296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Obesity's variability is significantly influenced by the interplay between genetic and environmental factors. We aimed to integrate the combined impact of genetic risk score (GRSBMI) with physical activity (PA), sugar-sweetened beverages (SSB), wine intake, and eating habits score (EHS) on obesity predisposition risk. Adults' (n = 5824) data were analyzed for common obesity-related single nucleotide polymorphisms and lifestyle habits. The weighted GRSBMI was constructed and categorized into quartiles (Qs), and the adjusted multivariate logistic regression models examined the association of GRSBMI with obesity (BMI ≥ 30) and lifestyle factors. GRSBMI was significantly associated with obesity risk. Each GRSBMI unit was associated with an increase of 3.06 BMI units (p ≤ 0.0001). PA markedly reduced obesity risk across GRSBMI Qs. Inactive participants' (≥90 min/week) mean BMI was higher in GRSBMI Q3-Q4 compared to Q1 (p = 0.003 and p < 0.001, respectively). Scoring EHS ≥ median, SSBs (≥1 cup/day), and non-wine drinking were associated with higher BMI within all GRSBMI Qs compared to EHS < median, non-SSBs, and non-wine drinkers. Mean BMI was higher in GRSBMI Q4 compared to other quartiles (p < 0.0001) in non-wine drinkers and compared to Q1 for SSB's consumers (p = 0.07). A higher GRSBMI augmented the impact of lifestyle factors on obesity. The interplay between GRSBMI and modifiable lifestyle factors provides a tailored personalized prevention and treatment for obesity management.
Collapse
Affiliation(s)
| | - Ruth Birk
- Nutrition Department, Health Science Faculty, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
70
|
Næss M, Kvaløy K, Sørgjerd EP, Sætermo KS, Norøy L, Røstad AH, Hammer N, Altø TG, Vikdal AJ, Hveem K. Data Resource Profile: The HUNT Biobank. Int J Epidemiol 2024; 53:dyae073. [PMID: 38836303 PMCID: PMC11150882 DOI: 10.1093/ije/dyae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/23/2024] [Indexed: 06/06/2024] Open
Affiliation(s)
- Marit Næss
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Kirsti Kvaløy
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
- Department of Community Medicine, Center for Sami Health Research, Arctic University of Norway, Tromso, Norway
| | - Elin P Sørgjerd
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Kristin S Sætermo
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Lise Norøy
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ann Helen Røstad
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Nina Hammer
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Trine Govasli Altø
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Anne Jorunn Vikdal
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology), Trondheim, Norway
- Department of Research, St Olav’s Hospital, Trondheim, Norway
| |
Collapse
|
71
|
Yu S, Ren H, Liu T, Han X, Guo H, Ning Q, Li Y, Zhou H, Chen M, Hu T. Metformin suppresses NFE2L1 pathway activation to inhibit gap junction beta protein expression in NSCLC. Cancer Med 2024; 13:e7021. [PMID: 38562019 PMCID: PMC10985411 DOI: 10.1002/cam4.7021] [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: 09/03/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVE Non-small-cell lung cancer (NSCLC) is a deadly form of cancer that exhibits extensive intercellular communication which contributed to chemoradiotherapy resistance. Recent evidence suggests that arrange of key proteins are involved in lung cancer progression, including gap junction proteins (GJPs). METHODS AND RESULTS In this study, we examined the expression patterns of GJPs in NSCLC, uncovering that both gap junction protein, beta 2 (GJB2) and gap junction protein, beta 2 (GJB3) are increased in LUAD and LUSC. We observed a correlation between the upregulation of GJB2, GJB3 in clinical samples and a worse prognosis in patients with NSCLC. By examining the mechanics, we additionally discovered that nuclear factor erythroid-2-related factor 1 (NFE2L1) had the capability to enhance the expression of connexin26 and connexin 31 in the NSCLC cell line A549. In addition, the use of metformin was discovered to cause significant downregulation of gap junction protein, betas (GJBs) by limiting the presence of NFE2L1 in the cytoplasm. CONCLUSION This emphasizes the potential of targeting GJBs as a viable treatment approach for NSCLC patients receiving metformin.
Collapse
Affiliation(s)
- Shuo Yu
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
- Department of General SurgeryThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Hui Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Tingting Liu
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Xiaoyan Han
- Department of General SurgeryWeifang People's HospitalWeifangShandongChina
| | - Hui Guo
- Department of OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Qian Ning
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Yang Li
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Hong Zhou
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Mingwei Chen
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Tinghua Hu
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
72
|
Harvey T, Rios M. The Role of BDNF and TrkB in the Central Control of Energy and Glucose Balance: An Update. Biomolecules 2024; 14:424. [PMID: 38672441 PMCID: PMC11048226 DOI: 10.3390/biom14040424] [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: 02/20/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
The global rise in obesity and related health issues, such as type 2 diabetes and cardiovascular disease, is alarming. Gaining a deeper insight into the central neural pathways and mechanisms that regulate energy and glucose homeostasis is crucial for developing effective interventions to combat this debilitating condition. A significant body of evidence from studies in humans and rodents indicates that brain-derived neurotrophic factor (BDNF) signaling plays a key role in regulating feeding, energy expenditure, and glycemic control. BDNF is a highly conserved neurotrophin that signals via the tropomyosin-related kinase B (TrkB) receptor to facilitate neuronal survival, differentiation, and synaptic plasticity and function. Recent studies have shed light on the mechanisms through which BDNF influences energy and glucose balance. This review will cover our current understanding of the brain regions, neural circuits, and cellular and molecular mechanisms underlying the metabolic actions of BDNF and TrkB.
Collapse
Affiliation(s)
- Theresa Harvey
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
73
|
Yeyeodu S, Hanafi D, Webb K, Laurie NA, Kimbro KS. Population-enriched innate immune variants may identify candidate gene targets at the intersection of cancer and cardio-metabolic disease. Front Endocrinol (Lausanne) 2024; 14:1286979. [PMID: 38577257 PMCID: PMC10991756 DOI: 10.3389/fendo.2023.1286979] [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: 09/01/2023] [Accepted: 12/07/2023] [Indexed: 04/06/2024] Open
Abstract
Both cancer and cardio-metabolic disease disparities exist among specific populations in the US. For example, African Americans experience the highest rates of breast and prostate cancer mortality and the highest incidence of obesity. Native and Hispanic Americans experience the highest rates of liver cancer mortality. At the same time, Pacific Islanders have the highest death rate attributed to type 2 diabetes (T2D), and Asian Americans experience the highest incidence of non-alcoholic fatty liver disease (NAFLD) and cancers induced by infectious agents. Notably, the pathologic progression of both cancer and cardio-metabolic diseases involves innate immunity and mechanisms of inflammation. Innate immunity in individuals is established through genetic inheritance and external stimuli to respond to environmental threats and stresses such as pathogen exposure. Further, individual genomes contain characteristic genetic markers associated with one or more geographic ancestries (ethnic groups), including protective innate immune genetic programming optimized for survival in their corresponding ancestral environment(s). This perspective explores evidence related to our working hypothesis that genetic variations in innate immune genes, particularly those that are commonly found but unevenly distributed between populations, are associated with disparities between populations in both cancer and cardio-metabolic diseases. Identifying conventional and unconventional innate immune genes that fit this profile may provide critical insights into the underlying mechanisms that connect these two families of complex diseases and offer novel targets for precision-based treatment of cancer and/or cardio-metabolic disease.
Collapse
Affiliation(s)
- Susan Yeyeodu
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
- Charles River Discovery Services, Morrisville, NC, United States
| | - Donia Hanafi
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - Kenisha Webb
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Nikia A. Laurie
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - K. Sean Kimbro
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
74
|
Kieler IN, Persson SM, Hagman R, Marinescu VD, Hedhammar Å, Strandberg E, Lindblad-Toh K, Arendt ML. Genome wide association study in Swedish Labrador retrievers identifies genetic loci associated with hip dysplasia and body weight. Sci Rep 2024; 14:6090. [PMID: 38480780 PMCID: PMC10937653 DOI: 10.1038/s41598-024-56060-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/01/2024] [Indexed: 03/17/2024] Open
Abstract
Genome wide association studies (GWAS) have been utilized to identify genetic risk loci associated with both simple and complex inherited disorders. Here, we performed a GWAS in Labrador retrievers to identify genetic loci associated with hip dysplasia and body weight. Hip dysplasia scores were available for 209 genotyped dogs. We identified a significantly associated locus for hip dysplasia on chromosome 24, with three equally associated SNPs (p = 4.3 × 10-7) in complete linkage disequilibrium located within NDRG3, a gene which in humans has been shown to be differentially expressed in osteoarthritic joint cartilage. Body weight, available for 85 female dogs, was used as phenotype for a second analysis. We identified two significantly associated loci on chromosome 10 (p = 4.5 × 10-7) and chromosome 31 (p = 2.5 × 10-6). The most associated SNPs within these loci were located within the introns of the PRKCE and CADM2 genes, respectively. PRKCE has been shown to play a role in regulation of adipogenesis whilst CADM2 has been associated with body weight in multiple human GWAS. In summary, we identified credible candidate loci explaining part of the genetic inheritance for hip dysplasia and body weight in Labrador retrievers with strong candidate genes in each locus previously implicated in the phenotypes investigated.
Collapse
Affiliation(s)
- Ida Nordang Kieler
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofia Malm Persson
- Department for Breeding and Health, Swedish Kennel Club, Stockholm, Sweden
| | - Ragnvi Hagman
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Voichita D Marinescu
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- SciLifeLab, Uppsala, Sweden
| | - Åke Hedhammar
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Erling Strandberg
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- SciLifeLab, Uppsala, Sweden
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maja Louise Arendt
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
- SciLifeLab, Uppsala, Sweden.
| |
Collapse
|
75
|
Wan X, Ao Y, Liu X, Zhuang P, Huang Y, Shi H, Jiao J, Zhang Y. Fried food consumption, genetic risk, and incident obesity: a prospective study. Food Funct 2024; 15:2760-2771. [PMID: 38385219 DOI: 10.1039/d3fo02803h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Background and aims: Genetic and dietary factors contribute to adiposity risk, but little evidence supports genetic personalization of fried food intake recommendations for the management of obesity. This study aimed to assess the associations between fried food consumption and adiposity incidence and whether the associations were modified by an individual's genotype. Methods: We included 27 427 participants who had dietary data assessed by a validated 24 h dietary recall and available anthropometric information from the UK Biobank study. The genetic risk score (GRS) was calculated using 940 BMI associated variants. Results: With an average of 8.1 years of follow-up, 1472 and 2893 participants were defined as having overall obesity and abdominal obesity, respectively. Individuals in the highest categories of fried food consumption were positively associated with the risk of obesity (HR = 1.31; 95% CI 1.10-1.56) and abdominal obesity (HR = 1.27; 95% CI 1.12-1.45) compared with the lowest categories. Moreover, fried food consumption had a significant interatction with obesity GRS for abdominal obesity risk (P interaction = 0.016). Fried food intake was associated with a higher abdominal obesity risk (HR = 1.59, 95% CI: 1.25-2.00) among participants with a lower genetic risk. Conclusions: Our findings indicated that fried food consumption had a higher abdominal obesity risk among individuals with a lower genetic risk, suggesting the restriction of fried food intake for this group of people.
Collapse
Affiliation(s)
- Xuzhi Wan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yang Ao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yingyu Huang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Hongbo Shi
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
76
|
Dabas J, Shunmukha Priya S, Alawani A, Budhrani P. What could be the reasons for not losing weight even after following a weight loss program? JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:37. [PMID: 38429842 PMCID: PMC10908186 DOI: 10.1186/s41043-024-00516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/28/2024] [Indexed: 03/03/2024]
Abstract
INTRODUCTION Approximately four million people worldwide die annually because of obesity. Weight loss is commonly recommended as a first-line therapy in overweight and obese patients. Although many individuals attempt to lose weight, not everyone achieves optimal success. Few studies point out that weight loss eventually slows down, stagnates or reverses in 85% of the cases. RESEARCH QUESTION What could be the reasons for not losing weight even after following a weight loss program? METHODS A scoping review of the literature was performed using weight loss-related search terms such as 'Obesity,' 'Overweight,' 'Lifestyle,' 'weight loss,' 'Basal Metabolism,' 'physical activity,' 'adherence,' 'energy balance,' 'Sleep' and 'adaptations. The search involved reference tracking and database and web searches (PUBMED, Science Direct, Elsevier, Web of Science and Google Scholar). Original articles and review papers on weight loss involving human participants and adults aged > 18 years were selected. Approximately 231 articles were reviewed, and 185 were included based on the inclusion criteria. DESIGN Scoping review. RESULTS In this review, the factors associated with not losing weight have broadly been divided into five categories. Studies highlighting each subfactor were critically reviewed and discussed. A wide degree of interindividual variability in weight loss is common in studies even after controlling for variables such as adherence, sex, physical activity and baseline weight. In addition to these variables, variations in factors such as previous weight loss attempts, sleep habits, meal timings and medications can play a crucial role in upregulating or downregulating the association between energy deficit and weight loss results. CONCLUSION This review identifies and clarifies the role of several factors that may hinder weight loss after the exploration of existing evidence. Judging the effectiveness of respective lifestyle interventions by simply observing the 'general behavior of the groups' is not always applicable in clinical practice. Each individual must be monitored and advised as per their requirements and challenges.
Collapse
Affiliation(s)
- Jyoti Dabas
- Institute of Nutrition and Fitness Sciences, Platinum Square, 4th floor, Office, 403, Opp. WNS, Sakore Nagar, Viman Nagar, Pune, Maharashtra, 411014, India
| | - S Shunmukha Priya
- Institute of Nutrition and Fitness Sciences, Platinum Square, 4th floor, Office, 403, Opp. WNS, Sakore Nagar, Viman Nagar, Pune, Maharashtra, 411014, India.
| | - Akshay Alawani
- Institute of Nutrition and Fitness Sciences, Platinum Square, 4th floor, Office, 403, Opp. WNS, Sakore Nagar, Viman Nagar, Pune, Maharashtra, 411014, India
| | - Praveen Budhrani
- Institute of Nutrition and Fitness Sciences, Platinum Square, 4th floor, Office, 403, Opp. WNS, Sakore Nagar, Viman Nagar, Pune, Maharashtra, 411014, India
| |
Collapse
|
77
|
Ferreira SRG, Macotela Y, Velloso LA, Mori MA. Determinants of obesity in Latin America. Nat Metab 2024; 6:409-432. [PMID: 38438626 DOI: 10.1038/s42255-024-00977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024]
Abstract
Obesity rates are increasing almost everywhere in the world, although the pace and timing for this increase differ when populations from developed and developing countries are compared. The sharp and more recent increase in obesity rates in many Latin American countries is an example of that and results from regional characteristics that emerge from interactions between multiple factors. Aware of the complexity of enumerating these factors, we highlight eight main determinants (the physical environment, food exposure, economic and political interest, social inequity, limited access to scientific knowledge, culture, contextual behaviour and genetics) and discuss how they impact obesity rates in Latin American countries. We propose that initiatives aimed at understanding obesity and hampering obesity growth in Latin America should involve multidisciplinary, global approaches that consider these determinants to build more effective public policy and strategies, accounting for regional differences and disease complexity at the individual and systemic levels.
Collapse
Affiliation(s)
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM Campus-Juriquilla, Querétaro, Mexico
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil.
| |
Collapse
|
78
|
Mikołajczyk-Stecyna J, Zuk E, Seremak-Mrozikiewicz A, Kurzawińska G, Wolski H, Drews K, Chmurzynska A. Genetic risk score for gestational weight gain. Eur J Obstet Gynecol Reprod Biol 2024; 294:20-27. [PMID: 38184896 DOI: 10.1016/j.ejogrb.2023.12.031] [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: 06/06/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
Gestational weight gain (GWG) involves health consequences for both mother and offspring. Genetic factors seem to play a role in the GWG trait. For small effect sizes of a single genetic polymorphism (SNP), a genetic risk score (GRS) summarizing risk-associated variation from multiple SNPs can serve as an effective approach to genetic association analysis. The aim of the study was to analyze the association between genetic risk score (GRS) and gestational weight gain (GWG). GWG was calculated for a total of 342 healthy Polish women of Caucasian origin, aged 19 to 45 years. The SNPs rs9939609 (FTO), rs6548238 (TMEM18), rs17782313 (MC4R), rs10938397 (GNPDA2), rs10913469 (SEC16B), rs1137101 (LEPR), rs7799039 (LEP), and rs5443 (GNB3) were genotyped using commercial TaqMan SNP assays. A simple genetic risk score was calculated into two ways: GRS1 based on the sum of risk alleles from each of the SNPs, while GRS2 based on the sum of risk alleles of FTO, LEPR, LEP, and GNB3. Positive association between GRS2 and GWG (β = 0.12, p = 0.029) was observed. Genetic risk variants of TMEM18 (p = 0.006, OR = 2.6) and GNB3 (p < 0.001, OR = 3.3) are more frequent in women with increased GWG, but a risk variant of GNPDA2 (p < 0.001, OR = 2.7) is more frequent in women with adequate GWG, and a risk variant of LEPR (p = 0.011, OR = 3.1) in women with decreased GWG. GRS2 and genetic variants of TMEM18, GNB3, GNPDA2, and LEPR are associated with weight gain during pregnancy.
Collapse
Affiliation(s)
- Joanna Mikołajczyk-Stecyna
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Wojska Polskiego 31, 60-624 Poznań, Poland
| | - Ewelina Zuk
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Wojska Polskiego 31, 60-624 Poznań, Poland
| | - Agnieszka Seremak-Mrozikiewicz
- Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland; Laboratory of Molecular Biology, Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland
| | - Grażyna Kurzawińska
- Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland; Laboratory of Molecular Biology, Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland
| | - Hubert Wolski
- Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland; Podhale State College of Applied Sciences in Nowy Targ, Kokoszków 71, 34-400 Nowy Targ, Poland
| | - Krzysztof Drews
- Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland; Laboratory of Molecular Biology, Division of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland
| | - Agata Chmurzynska
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Wojska Polskiego 31, 60-624 Poznań, Poland.
| |
Collapse
|
79
|
McEwan AR, Hing B, Erickson JC, Hutchings G, Urama C, Norton-Hughes E, D'Ippolito M, Berry S, Delibegovic M, Grassmann F, MacKenzie A. An ancient polymorphic regulatory region within the BDNF gene associated with obesity modulates anxiety-like behaviour in mice and humans. Mol Psychiatry 2024; 29:660-670. [PMID: 38228888 PMCID: PMC11153140 DOI: 10.1038/s41380-023-02359-7] [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: 06/20/2023] [Revised: 11/10/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024]
Abstract
Obesity and anxiety are morbidities notable for their increased impact on society during the recent COVID-19 pandemic. Understanding the mechanisms governing susceptibility to these conditions will increase our quality of life and resilience to future pandemics. In the current study, we explored the function of a highly conserved regulatory region (BE5.1) within the BDNF gene that harbours a polymorphism strongly associated with obesity (rs10767664; p = 4.69 × 10-26). Analysis in primary cells suggested that the major T-allele of BE5.1 was an enhancer, whereas the obesity-associated A-allele was not. However, CRISPR/CAS9 deletion of BE5.1 from the mouse genome (BE5.1KO) produced no significant effect on the expression of BDNF transcripts in the hypothalamus, no change in weight gain after 28 days and only a marginally significant increase in food intake. Nevertheless, transcripts were significantly increased in the amygdala of female mice and elevated zero maze and marble-burying tests demonstrated a significant increase in anxiety-like behaviour that could be reversed by diazepam. Consistent with these observations, human GWAS cohort analysis demonstrated a significant association between rs10767664 and anxiousness in human populations. Intriguingly, interrogation of the human GTEx eQTL database demonstrated no effect on BDNF mRNA levels associated with rs10767664 but a highly significant effect on BDNF-antisense (BDNF-AS) gene expression and splicing. The subsequent observation that deletion of BE5.1 also significantly reduced BDNF-AS expression in mice suggests a novel mechanism in the regulation of BDNF expression common to mice and humans, which contributes to the modulation of mood and anxiety in both species.
Collapse
Affiliation(s)
- Andrew R McEwan
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Benjamin Hing
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Johanna C Erickson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Greg Hutchings
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Charity Urama
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Emily Norton-Hughes
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Mariam D'Ippolito
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Susan Berry
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Mirela Delibegovic
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK
| | - Felix Grassmann
- Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Alasdair MacKenzie
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB24 2ZD, UK.
| |
Collapse
|
80
|
Sokary S, Almaghrbi H, Bawadi H. The Interaction Between Body Mass Index Genetic Risk Score and Dietary Intake on Weight Status: A Systematic Review. Diabetes Metab Syndr Obes 2024; 17:925-941. [PMID: 38435632 PMCID: PMC10908334 DOI: 10.2147/dmso.s452660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/15/2024] [Indexed: 03/05/2024] Open
Abstract
Background The escalating global obesity epidemic and the emergence of personalized medicine strategies point to the pressing need to investigate the interplay between genetic risk scores (GRSs), dietary intake, and their combined impact on weight status. This systematic review synthesizes evidence from diverse studies to elucidate how dietary patterns and individual foods interact with genetic predisposition to obesity. Methods Literature searches were conducted in the PubMed, Embase, Science Direct, and Scopus databases until August 2023, following PRISMA guidelines. Out of 575 articles, 15 articles examining the interaction between genetic risk score for body mass index and dietary intake on weight outcomes met the inclusion criteria. All included studies were cross-sectional in design and were assessed for quality using the Newcastle‒Ottawa Scale. Results Unhealthy dietary intake exacerbated the genetic predisposition to obesity, evident in studies assessing Western diet, sulfur microbial diet, and individual macronutrients, including saturated fatty acids, sugar-sweetened beverages and fried foods. Conversely, adhering to healthier dietary intake mitigated the genetic predisposition to obesity, as observed in studies involving Alternative Healthy Eating Index, Alternative Mediterranean Diet, Dietary Approach to Stop Hypertension scores, healthy plant-based diets, and specific foods such as fruits, vegetables, and n-3 polyunsaturated fatty acids. Conclusion This is the first systematic review to explore the interaction between genetics and dietary intake in shaping obesity outcomes. The findings have implications for tailored interventions; however, more controlled clinical trials with robust designs are needed to be able to recommend personalized nutrition based on nutrition for obesity prevention and management.
Collapse
Affiliation(s)
- Sara Sokary
- Department of Human Nutrition, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Heba Almaghrbi
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hiba Bawadi
- Department of Human Nutrition, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
81
|
Reshetnikov E, Churnosova M, Reshetnikova Y, Stepanov V, Bocharova A, Serebrova V, Trifonova E, Ponomarenko I, Sorokina I, Efremova O, Orlova V, Batlutskaya I, Ponomarenko M, Churnosov V, Aristova I, Polonikov A, Churnosov M. Maternal Age at Menarche Genes Determines Fetal Growth Restriction Risk. Int J Mol Sci 2024; 25:2647. [PMID: 38473894 PMCID: PMC10932237 DOI: 10.3390/ijms25052647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
We aimed to explore the potential link of maternal age at menarche (mAAM) gene polymorphisms with risk of the fetal growth restriction (FGR). This case (FGR)-control (FGR free) study included 904 women (273 FGR and 631 control) in the third trimester of gestation examined/treated in the Departments of Obstetrics. For single nucleotide polymorphism (SNP) multiplex genotyping, 50 candidate loci of mAAM were chosen. The relationship of mAAM SNPs and FGR was appreciated by regression procedures (logistic/model-based multifactor dimensionality reduction [MB-MDR]) with subsequent in silico assessment of the assumed functionality pithy of FGR-related loci. Three mAAM-appertain loci were FGR-linked to genes such as KISS1 (rs7538038) (effect allele G-odds ratio (OR)allelic = 0.63/pperm = 0.0003; ORadditive = 0.61/pperm = 0.001; ORdominant = 0.56/pperm = 0.001), NKX2-1 (rs999460) (effect allele A-ORallelic = 1.37/pperm = 0.003; ORadditive = 1.45/pperm = 0.002; ORrecessive = 2.41/pperm = 0.0002), GPRC5B (rs12444979) (effect allele T-ORallelic = 1.67/pperm = 0.0003; ORdominant = 1.59/pperm = 0.011; ORadditive = 1.56/pperm = 0.009). The haplotype ACA FSHB gene (rs555621*rs11031010*rs1782507) was FRG-correlated (OR = 0.71/pperm = 0.05). Ten FGR-implicated interworking models were founded for 13 SNPs (pperm ≤ 0.001). The rs999460 NKX2-1 and rs12444979 GPRC5B interplays significantly influenced the FGR risk (these SNPs were present in 50% of models). FGR-related mAAM-appertain 15 polymorphic variants and 350 linked SNPs were functionally momentous in relation to 39 genes participating in the regulation of hormone levels, the ovulation cycle process, male gonad development and vitamin D metabolism. Thus, this study showed, for the first time, that the mAAM-appertain genes determine FGR risk.
Collapse
Affiliation(s)
- Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Yuliya Reshetnikova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - 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; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Olga Efremova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Valentina Orlova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Irina Batlutskaya
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Marina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Vladimir Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| |
Collapse
|
82
|
Viinikainen J, Böckerman P, Willage B, Elovainio M, Kari JT, Lehtimäki T, Pehkonen J, Pitkänen N, Raitakari O. Effect of weight on depression using multiple genetic instruments. PLoS One 2024; 19:e0297594. [PMID: 38394117 PMCID: PMC10889664 DOI: 10.1371/journal.pone.0297594] [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: 03/16/2023] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
A striking global health development over the past few decades has been the increasing prevalence of overweight and obesity. At the same time, depression has become increasingly common in almost all high-income countries. We investigated whether body weight, measured by body mass index (BMI), has a causal effect on depression symptoms in Finland. Using data drawn from the Cardiovascular Risk in Young Finns Study (N = 1,523, mean age 41.9, SD 5), we used linear regression to establish the relationship between BMI and depression symptoms measured by 21-item Beck's Depression Inventory. To identify causal relationships, we used the Mendelian randomization (MR) method with weighted sums of genetic markers (single nucleotide polymorphisms, SNPs) as instruments for BMI. We employ instruments (polygenic risk scores, PGSs) with varying number of SNPs that are associated with BMI to evaluate the sensitivity of our results to instrument strength. Based on linear regressions, higher BMI was associated with a higher prevalence of depression symptoms among females (b = 0.238, p = 0.000) and males (b = 0.117, p = 0.019). However, the MR results imply that the positive link applies only to females (b = 0.302, p = 0.007) but not to males (b = -0.070, p = 0.520). Poor instrument strength may explain why many previous studies that have utilized genetic instruments have been unable to identify a statistically significant link between BMI and depression-related traits. Although the number of genetic markers in the instrument had only a minor effect on the point estimates, the standard errors were much smaller when more powerful instruments were employed.
Collapse
Affiliation(s)
- Jutta Viinikainen
- Jyväskylä University School of Business and Economics, University of Jyväskylä, Jyväskylä, Finland
| | - Petri Böckerman
- Jyväskylä University School of Business and Economics, University of Jyväskylä, Jyväskylä, Finland
- Labour Institute for Economic Research LABORE, Helsinki, Finland
- IZA Institute of Labor Economics, Bonn, Germany
| | - Barton Willage
- Department of Economics, University of Colorado—Denver, Denver, Colorado, United States of America
| | - Marko Elovainio
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jaana T. Kari
- Jyväskylä University School of Business and Economics, University of Jyväskylä, Jyväskylä, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
| | - Jaakko Pehkonen
- Jyväskylä University School of Business and Economics, University of Jyväskylä, Jyväskylä, Finland
| | - Niina Pitkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
83
|
Nagayoshi M, Hishida A, Shimizu T, Kato Y, Kubo Y, Okada R, Tamura T, Otonari J, Ikezaki H, Hara M, Nishida Y, Oze I, Koyanagi YN, Nakamura Y, Kusakabe M, Ibusuki R, Shibuya K, Suzuki S, Nishiyama T, Koyama T, Ozaki E, Kuriki K, Takashima N, Nakamura Y, Katsuura-Kamano S, Arisawa K, Nakatochi M, Momozawa Y, Takeuchi K, Wakai K. BMI and Cardiometabolic Traits in Japanese: A Mendelian Randomization Study. J Epidemiol 2024; 34:51-62. [PMID: 36709979 PMCID: PMC10751192 DOI: 10.2188/jea.je20220154] [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: 05/26/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Although many observational studies have demonstrated significant relationships between obesity and cardiometabolic traits, the causality of these relationships in East Asians remains to be elucidated. METHODS We conducted individual-level Mendelian randomization (MR) analyses targeting 14,083 participants in the Japan Multi-Institutional Collaborative Cohort Study and two-sample MR analyses using summary statistics based on genome-wide association study data from 173,430 Japanese. Using 83 body mass index (BMI)-related loci, genetic risk scores (GRS) for BMI were calculated, and the effects of BMI on cardiometabolic traits were examined for individual-level MR analyses using the two-stage least squares estimator method. The β-coefficients and standard errors for the per-allele association of each single-nucleotide polymorphism as well as all outcomes, or odds ratios with 95% confidence intervals were calculated in the two-sample MR analyses. RESULTS In individual-level MR analyses, the GRS of BMI was not significantly associated with any cardiometabolic traits. In two-sample MR analyses, higher BMI was associated with increased risks of higher blood pressure, triglycerides, and uric acid, as well as lower high-density-lipoprotein cholesterol and eGFR. The associations of BMI with type 2 diabetes in two-sample MR analyses were inconsistent using different methods, including the directions. CONCLUSION The results of this study suggest that, even among the Japanese, an East Asian population with low levels of obesity, higher BMI could be causally associated with the development of a variety of cardiometabolic traits. Causality in those associations should be clarified in future studies with larger populations, especially those of BMI with type 2 diabetes.
Collapse
Affiliation(s)
- Mako Nagayoshi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonori Shimizu
- Undergraduate Course, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasufumi Kato
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Kubo
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Tamura
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Otonari
- Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Psychosomatic Medicine, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Hiroaki Ikezaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Comprehensive General Internal Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, Japan
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yuriko N. Koyanagi
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yohko Nakamura
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Miho Kusakabe
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Rie Ibusuki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Keiichi Shibuya
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- Department of Emergency, Kagoshima Prefectural Oshima Hospital, Kagoshima, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takeshi Nishiyama
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Etsuko Ozaki
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Naoyuki Takashima
- Department of Public Health, Faculty of Medicine, Kindai University, Osaka, Japan
- Department of Public Health, Shiga University of Medical Science, Otsu, Japan
| | - Yasuyuki Nakamura
- Department of Public Health, Shiga University of Medical Science, Otsu, Japan
- Yamashina Racto Clinic and Medical Examination Center, Kyoto, Japan
| | - Sakurako Katsuura-Kamano
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kokichi Arisawa
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Kenji Takeuchi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
84
|
Véniant MM, Lu SC, Atangan L, Komorowski R, Stanislaus S, Cheng Y, Wu B, Falsey JR, Hager T, Thomas VA, Ambhaikar M, Sharpsten L, Zhu Y, Kurra V, Jeswani R, Oberoi RK, Parnes JR, Honarpour N, Neutel J, Strande JL. A GIPR antagonist conjugated to GLP-1 analogues promotes weight loss with improved metabolic parameters in preclinical and phase 1 settings. Nat Metab 2024; 6:290-303. [PMID: 38316982 PMCID: PMC10896721 DOI: 10.1038/s42255-023-00966-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
Obesity is a major public health crisis. Multi-specific peptides have emerged as promising therapeutic strategies for clinical weight loss. Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are endogenous incretins that regulate weight through their receptors (R). AMG 133 (maridebart cafraglutide) is a bispecific molecule engineered by conjugating a fully human monoclonal anti-human GIPR antagonist antibody to two GLP-1 analogue agonist peptides using amino acid linkers. Here, we confirm the GIPR antagonist and GLP-1R agonist activities in cell-based systems and report the ability of AMG 133 to reduce body weight and improve metabolic markers in male obese mice and cynomolgus monkeys. In a phase 1, randomized, double-blind, placebo-controlled clinical study in participants with obesity ( NCT04478708 ), AMG 133 had an acceptable safety and tolerability profile along with pronounced dose-dependent weight loss. In the multiple ascending dose cohorts, weight loss was maintained for up to 150 days after the last dose. These findings support continued clinical evaluation of AMG 133.
Collapse
Affiliation(s)
- Murielle M Véniant
- Amgen Research, Department of Cardiometabolic Disorders, Thousand Oaks, CA, USA.
| | - Shu-Chen Lu
- Amgen Research, Department of Cardiometabolic Disorders, Thousand Oaks, CA, USA
| | - Larissa Atangan
- Amgen Research, Department of Cardiometabolic Disorders, Thousand Oaks, CA, USA
| | - Renee Komorowski
- Amgen Research, Department of Cardiometabolic Disorders, Thousand Oaks, CA, USA
| | - Shanaka Stanislaus
- Amgen Research, Department of Cardiometabolic Disorders, Thousand Oaks, CA, USA
| | - Yuan Cheng
- Amgen Research, Department of Therapeutic Discovery, Thousand Oaks, CA, USA
| | - Bin Wu
- Amgen Research, Department of Therapeutic Discovery, Thousand Oaks, CA, USA
| | - James R Falsey
- Amgen Research, Department of Therapeutic Discovery, Thousand Oaks, CA, USA
| | - Todd Hager
- Amgen Research, Department of Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA
| | - Veena A Thomas
- Amgen Research, Department of Pharmacokinetics and Drug Metabolism, South San Francisco, CA, USA
| | - Malhar Ambhaikar
- Pre-pivotal Drug Substance Technologies, Amgen, Thousand Oaks, CA, USA
| | | | - Yineng Zhu
- Amgen Early Development, Amgen, Thousand Oaks, CA, USA
| | - Vamsi Kurra
- Amgen Research, Department of Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA
| | - Rohini Jeswani
- Amgen Research, Department of Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA
| | | | - Jane R Parnes
- Amgen Early Development, Amgen, Thousand Oaks, CA, USA
| | | | - Joel Neutel
- Orange County Research Center, Tustin, CA, USA
| | | |
Collapse
|
85
|
Wit M, Belykh A, Sumara G. Protein kinase D (PKD) on the crossroad of lipid absorption, synthesis and utilization. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119653. [PMID: 38104800 DOI: 10.1016/j.bbamcr.2023.119653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 10/19/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
Inappropriate lipid levels in the blood, as well as its content and composition in different organs, underlie multiple metabolic disorders including obesity, non-alcoholic fatty liver disease, type 2 diabetes, and atherosclerosis. Multiple processes contribute to the complex metabolism of triglycerides (TGs), fatty acids (FAs), and other lipid species. These consist of digestion and absorption of dietary lipids, de novo FAs synthesis (lipogenesis), uptake of TGs and FAs by peripheral tissues, TGs storage in the intracellular depots as well as lipid utilization for β-oxidation and their conversion to lipid-derivatives. A majority of the enzymatic reactions linked to lipogenesis, TGs synthesis, lipid absorption, and transport are happening at the endoplasmic reticulum, while β-oxidation takes place in mitochondria and peroxisomes. The Golgi apparatus is a central sorting, protein- and lipid-modifying organelle and hence is involved in lipid metabolism as well. However, the impact of the processes taking part in the Golgi apparatus are often overseen. The protein kinase D (PKD) family (composed of three members, PKD1, 2, and 3) is the master regulator of Golgi dynamics. PKDs are also a sensor of different lipid species in distinct cellular compartments. In this review, we discuss the roles of PKD family members in the regulation of lipid metabolism including the processes executed by PKDs at the Golgi apparatus. We also discuss the role of PKDs-dependent signaling in different cellular compartments and organs in the context of the development of metabolic disorders.
Collapse
Affiliation(s)
- Magdalena Wit
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland
| | - Andrei Belykh
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland
| | - Grzegorz Sumara
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland.
| |
Collapse
|
86
|
Mosley JD, Shi M, Agamasu D, Vaitinadin NS, Murthy VL, Shah RV, Bagheri M, Ferguson JF. Branched-chain amino acids and type 2 diabetes: a bidirectional Mendelian randomization analysis. Obesity (Silver Spring) 2024; 32:423-435. [PMID: 38269471 PMCID: PMC10827349 DOI: 10.1002/oby.23951] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 01/26/2024]
Abstract
OBJECTIVE Genetic studies have suggested that the branched-chain amino acids (BCAAs) valine, leucine, and isoleucine have a causal association with type 2 diabetes (T2D). However, inferences are based on a limited number of genetic loci associated with BCAAs. METHODS Instrumental variables (IVs) for each BCAA were constructed and validated using large well-powered data sets and their association with T2D was tested using a two-sample inverse-variance weighted Mendelian randomization approach. Sensitivity analyses were performed to ensure the accuracy of the findings. A reverse association was assessed using instrumental variables for T2D. RESULTS Estimated effect sizes between BCAA IVs and T2D, excluding outliers, were as follows: valine (β = 0.14 change in log-odds per SD change in valine, 95% CI: -0.06 to 0.33, p = 0.17), leucine (β = 0.15, 95% CI: -0.02 to 0.32, p = 0.09), and isoleucine (β = 0.13, 95% CI: -0.08 to 0.34, p = 0.24). In contrast, T2D IVs were positively associated with each BCAA, i.e., valine (β = 0.08 per SD change in levels per log-odds change in T2D, 95% CI: 0.05 to 0.10, p = 1.8 × 10-9 ), leucine (β = 0.06, 95% CI: 0.04 to 0.09, p = 4.5 × 10-8 ), and isoleucine (β = 0.06, 95% CI: 0.04 to 0.08, p = 2.8 × 10-8 ). CONCLUSIONS These data suggest that the BCAAs are not mediators of T2D risk but are biomarkers of diabetes.
Collapse
Affiliation(s)
- Jonathan D. Mosley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mingjian Shi
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Venkatesh L. Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Ravi V. Shah
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Minoo Bagheri
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jane F. Ferguson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
87
|
Lopez-Cortes OD, Trujillo-Sánchez F, Sierra-Ruelas E, Martinez-Lopez E, Di Marzo V, Vizmanos B. Association between the FAAH C385A variant (rs324420) and obesity-related traits: a systematic review. Int J Obes (Lond) 2024; 48:188-201. [PMID: 38114812 DOI: 10.1038/s41366-023-01428-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Overweight and obesity are the consequence of a sustained positive energy balance. Twin studies show high heritability rates pointing to genetics as one of the principal risk factors. By 2022, genomic studies led to the identification of almost 300 obesity-associated variants that could help to fill the gap of the high heritability rates. The endocannabinoid system is a critical regulator of metabolism for its effects on the central nervous system and peripheral tissues. Fatty acid amide hydrolase (FAAH) is a key enzyme in the inactivation of one of the two endocannabinoids, anandamide, and of its congeners. The rs324420 variant within the FAAH gene is a nucleotide missense change at position 385 from cytosine to adenine, resulting in a non-synonymous amino acid substitution from proline to threonine in the FAAH enzyme. This change increases sensitivity to proteolytic degradation, leading to reduced FAAH levels and increased levels of anandamide, associated with obesity-related traits. However, association studies of this variant with metabolic parameters have found conflicting results. This work aims to perform a systematic review of the existing literature on the association of the rs324420 variant in the FAAH gene with obesity and its related traits. METHODS A literature search was conducted in PubMed, Web of Science, and Scopus. A total of 645 eligible studies were identified for the review. RESULTS/CONCLUSIONS After the identification, duplicate elimination, title and abstract screening, and full-text evaluation, 28 studies were included, involving 28 183 individuals. We show some evidence of associations between the presence of the variant allele and higher body mass index, waist circumference, fat mass, and waist-to-hip ratio levels and alterations in glucose and lipid homeostasis. However, this evidence should be taken with caution, as many included studies did not report a significant difference between genotypes. These discordant results could be explained mainly by the pleiotropy of the endocannabinoid system, the increase of other anandamide-like mediators metabolized by FAAH, and the influence of gene-environment interactions. More research is necessary to study the endocannabinoidomic profiles and their association with metabolic diseases.
Collapse
Affiliation(s)
- Oscar David Lopez-Cortes
- Licenciatura en Medicina, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
- Canada Excellence Research Chair in Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec, QC, G1V 4G5, Canada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Université Laval, Québec, QC, G1V 4G5, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Québec, QC, G1V 4G5, Canada
| | - Francisco Trujillo-Sánchez
- Licenciatura en Medicina, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
| | - Erika Sierra-Ruelas
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
- Doctorado en Ciencias de la Nutrición Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
| | - Erika Martinez-Lopez
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair in Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec, QC, G1V 4G5, Canada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Université Laval, Québec, QC, G1V 4G5, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Québec, QC, G1V 4G5, Canada
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche (CNR), Pozzuoli, Italy
| | - Barbara Vizmanos
- Instituto de Nutrigenética y Nutrigenómica Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico.
- Doctorado en Ciencias de la Nutrición Traslacional, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico.
- Departamento de Clínicas de Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico.
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44320, Mexico.
| |
Collapse
|
88
|
Carvalho NRG, He Y, Smadbeck P, Flannick J, Mercader JM, Udler M, Manrai AK, Moreno J, Patel CJ. Assessing the genetic contribution of cumulative behavioral factors associated with longitudinal type 2 diabetes risk highlights adiposity and the brain-metabolic axis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.30.24302019. [PMID: 38352440 PMCID: PMC10863013 DOI: 10.1101/2024.01.30.24302019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
While genetic factors, behavior, and environmental exposures form a complex web of interrelated associations in type 2 diabetes (T2D), their interaction is poorly understood. Here, using data from ~500K participants of the UK Biobank, we identify the genetic determinants of a "polyexposure risk score" (PXS) a new risk factor that consists of an accumulation of 25 associated individual-level behaviors and environmental risk factors that predict longitudinal T2D incidence. PXS-T2D had a non-zero heritability (h2 = 0.18) extensive shared genetic architecture with established clinical and biological determinants of T2D, most prominently with body mass index (genetic correlation [rg] = 0.57) and Homeostatic Model Assessment for Insulin Resistance (rg = 0.51). Genetic loci associated with PXS-T2D were enriched for expression in the brain. Biobank scale data with genetic information illuminates how complex and cumulative exposures and behaviors as a whole impact T2D risk but whose biology have been elusive in genome-wide studies of T2D.
Collapse
Affiliation(s)
- Nuno R. G. Carvalho
- School of Biological Sciences; Georgia Institute of Technology; Atlanta, GA, 30332, USA
| | - Yixuan He
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Patrick Smadbeck
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Jason Flannick
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Josep M. Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Miriam Udler
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Arjun K Manrai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Jordi Moreno
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Chirag J. Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
89
|
Nemoto S, Kubota T, Ishikura T, Nakayama M, Kobayashi A, Yazaki J, Uchida K, Matsuda M, Kondo T, Ohara O, Koseki H, Koyasu S, Ohno H. Characterization of metabolic phenotypes and distinctive genes in mice with low-weight gain. FASEB J 2024; 38:e23339. [PMID: 38069905 DOI: 10.1096/fj.202301565r] [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: 08/03/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Being overweight exacerbates various metabolic diseases, necessitating the identification of target molecules for obesity control. In the current study, we investigated common physiological features related to metabolism in mice with low weight gain: (1) G protein-coupled receptor, family C, group 5, member B-knockout; (2) gastric inhibitory polypeptide receptor-knockout; and (3) Iroquois-related homeobox 3-knockout. Moreover, we explored genes involved in metabolism by analyzing differentially expressed genes (DEGs) between low-weight gain mice and the respective wild-type control mice. The common characteristics of the low-weight gain mice were low inguinal white adipose tissue (iWAT) and liver weight despite similar food intake along with lower blood leptin levels and high energy expenditure. The DEGs of iWAT, epididymal (gonadal) WAT, brown adipose tissue, muscle, liver, hypothalamus, and hippocampus common to these low-weight gain mice were designated as candidate genes associated with metabolism. One such gene tetraspanin 7 (Tspan7) from the iWAT was validated using knockout and overexpressing mouse models. Mice with low Tspan7 expression gained more weight, while those with high Tspan7 expression gained less weight, confirming the involvement of the Tspan7 gene in weight regulation. Collectively, these findings suggest that the candidate gene list generated in this study contains potential target molecules for obesity regulation. Further validation and additional data from low-weight gain mice will aid in understanding the molecular mechanisms associated with obesity.
Collapse
Affiliation(s)
- Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tetsuya Kubota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Ishikura
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Manabu Nakayama
- Laboratory of Medical Omics Research, Department of Frontier Research and Development, Kazusa DNA Research Institute, Kisarazu, Chiba, Japan
| | - Atsuo Kobayashi
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Junshi Yazaki
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kazuyo Uchida
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masashi Matsuda
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takashi Kondo
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Chiba, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Immune Regulation, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
90
|
Queen NJ, Huang W, Zou X, Mo X, Cao L. AAV-BDNF gene therapy ameliorates a hypothalamic neuroinflammatory signature in the Magel2-null model of Prader-Willi syndrome. Mol Ther Methods Clin Dev 2023; 31:101108. [PMID: 37766791 PMCID: PMC10520877 DOI: 10.1016/j.omtm.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Individuals with Prader-Willi syndrome (PWS) exhibit several metabolic and behavioral abnormalities associated with excessive food-seeking activity. PWS is thought to be driven in part by dysfunctional hypothalamic circuitry and blunted responses to peripheral signals of satiety. Previous work described a hypothalamic transcriptomic signature of individuals with PWS. Notably, PWS patients exhibited downregulation of genes involved in neuronal development and an upregulation of neuroinflammatory genes. Deficiencies of brain-derived neurotrophic factor (BDNF) and its receptor were identified as potential drivers of PWS phenotypes. Our group recently applied an adeno-associated viral (AAV)-BDNF gene therapy within a preclinical PWS model, Magel2-null mice, to improve metabolic and behavioral function. While this proof-of-concept project was promising, it remained unclear how AAV-BDNF was influencing the hypothalamic microenvironment and how its therapeutic effect was mediated. To investigate, we hypothalamically injected AAV-BDNF to wild type and Magel2-null mice and performed mRNA sequencing on hypothalamic tissue. Here, we report that (1) Magel2 deficiency is associated with neuroinflammation in the hypothalamus and (2) AAV-BDNF gene therapy reverses this neuroinflammation. These data newly reveal Magel2-null mice as a valid model of PWS-related neuroinflammation and furthermore suggest that AAV-BDNF may modulate obesity-related neuroinflammatory phenotypes through direct or indirect means.
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
| | - Xunchang Zou
- 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
| | - Xiaokui Mo
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 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
|
91
|
Prone-Olazabal D, Davies I, González-Galarza FF. Metabolic Syndrome: An Overview on Its Genetic Associations and Gene-Diet Interactions. Metab Syndr Relat Disord 2023; 21:545-560. [PMID: 37816229 DOI: 10.1089/met.2023.0125] [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] [Indexed: 10/12/2023] Open
Abstract
Metabolic syndrome (MetS) is a cluster of cardiometabolic risk factors that includes central obesity, hyperglycemia, hypertension, and dyslipidemias and whose inter-related occurrence may increase the odds of developing type 2 diabetes and cardiovascular diseases. MetS has become one of the most studied conditions, nevertheless, due to its complex etiology, this has not been fully elucidated. Recent evidence describes that both genetic and environmental factors play an important role on its development. With the advent of genomic-wide association studies, single nucleotide polymorphisms (SNPs) have gained special importance. In this review, we present an update of the genetics surrounding MetS as a single entity as well as its corresponding risk factors, considering SNPs and gene-diet interactions related to cardiometabolic markers. In this study, we focus on the conceptual aspects, diagnostic criteria, as well as the role of genetics, particularly on SNPs and polygenic risk scores (PRS) for interindividual analysis. In addition, this review highlights future perspectives of personalized nutrition with regard to the approach of MetS and how individualized multiomics approaches could improve the current outlook.
Collapse
Affiliation(s)
- Denisse Prone-Olazabal
- Postgraduate Department, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Ian Davies
- Research Institute of Sport and Exercise Science, The Institute for Health Research, Liverpool John Moores University, Liverpool, United Kingdom
| | | |
Collapse
|
92
|
Wang L, Wang S, Song C, Yu Y, Jiang Y, Wang Y, Li X. Bibliometric analysis of residual cardiovascular risk: trends and frontiers. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2023; 42:132. [PMID: 38017531 PMCID: PMC10683255 DOI: 10.1186/s41043-023-00478-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND The presence of residual cardiovascular risk is an important cause of cardiovascular events. Despite the significant advances in our understanding of residual cardiovascular risk, a comprehensive analysis through bibliometrics has not been performed to date. Our objective is to conduct bibliometric studies to analyze and visualize the current research hotspots and trends related to residual cardiovascular risk. This will aid in understanding the future directions of both basic and clinical research in this area. METHODS The literature was obtained from the Web of Science Core Collection database. The literature search date was September 28, 2022. Bibliometric indicators were analyzed using CiteSpace, VOSviewer, Bibliometrix (an R package), and Microsoft Excel. RESULT A total of 1167 papers were included, and the number of publications is increasing rapidly in recent years. The United States and Harvard Medical School are the leading country and institution, respectively, in the study of residual cardiovascular risk. Ridker PM and Boden WE are outstanding investigators in this field. According to our research results, the New England Journal of Medicine is the most influential journal in the field of residual cardiovascular risk, whereas Atherosclerosis boasts the highest number of publications on this topic. Analysis of keywords and landmark literature identified current research hotspots including complications of residual cardiovascular risk, risk factors, and pharmacological prevention strategies. CONCLUSION In recent times, global attention toward residual cardiovascular risk has significantly increased. Current research is focused on comprehensive lipid-lowering, residual inflammation risk, and dual-pathway inhibition strategies. Future efforts should emphasize strengthening international communication and cooperation to promote the comprehensive evaluation and management of residual cardiovascular risk.
Collapse
Affiliation(s)
- Lin Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Sutong Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chaoyuan Song
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Zibo Central Hospital, Zibo, China
| | - Yiding Yu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuehua Jiang
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongcheng Wang
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Li
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
93
|
Pelczyńska M, Miller-Kasprzak E, Piątkowski M, Mazurek R, Klause M, Suchecka A, Bucoń M, Bogdański P. The Role of Adipokines and Myokines in the Pathogenesis of Different Obesity Phenotypes-New Perspectives. Antioxidants (Basel) 2023; 12:2046. [PMID: 38136166 PMCID: PMC10740719 DOI: 10.3390/antiox12122046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Obesity is a characteristic disease of the twenty-first century that is affecting an increasing percentage of society. Obesity expresses itself in different phenotypes: normal-weight obesity (NWO), metabolically obese normal-weight (MONW), metabolically healthy obesity (MHO), and metabolically unhealthy obesity (MUO). A range of pathophysiological mechanisms underlie the occurrence of obesity, including inflammation, oxidative stress, adipokine secretion, and other processes related to the pathophysiology of adipose tissue (AT). Body mass index (BMI) is the key indicator in the diagnosis of obesity; however, in the case of the NWO and MONW phenotypes, the metabolic disturbances are present despite BMI being within the normal range. On the other hand, MHO subjects with elevated BMI values do not present metabolic abnormalities. The MUO phenotype involves both a high BMI value and an abnormal metabolic profile. In this regard, attention has been focused on the variety of molecules produced by AT and their role in the development of obesity. Nesfatin-1, neuregulin 4, myonectin, irisin, and brain-derived neurotrophic factor (BDNF) all seem to have protective effects against obesity. The primary mechanism underlying the action of nesfatin-1 involves an increase in insulin sensitivity and reduced food intake. Neuregulin 4 sup-presses lipogenesis, decreases lipid accumulation, and reduces chronic low-grade inflammation. Myonectin lowers the amount of fatty acids in the bloodstream by increasing their absorption in the liver and AT. Irisin stimulates the browning of white adipose tissue (WAT) and consequently in-creases energy expenditure, additionally regulating glucose metabolism. Another molecule, BDNF, has anorexigenic effects. Decorin protects against the development of hyperglycemia, but may also contribute to proinflammatory processes. Similar effects are shown in the case of visfatin and chemerin, which may predispose to obesity. Visfatin increases adipogenesis, causes cholesterol accumulation in macrophages, and contributes to the development of glucose intolerance. Chemerin induces angiogenesis, which promotes the expansion of AT. This review aims to discuss the role of adipokines and myokines in the pathogenesis of the different obesity phenotypes.
Collapse
Affiliation(s)
- Marta Pelczyńska
- Chair and Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569 Poznań, Poland; (E.M.-K.); (P.B.)
| | - Ewa Miller-Kasprzak
- Chair and Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569 Poznań, Poland; (E.M.-K.); (P.B.)
| | - Marcin Piątkowski
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Roksana Mazurek
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Mateusz Klause
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Anna Suchecka
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Magdalena Bucoń
- Faculty of Medicine, Poznan University of Medical Sciences, 70 Bukowska Street, 60-812 Poznań, Poland
| | - Paweł Bogdański
- Chair and Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 84 Szamarzewskiego Street, 60-569 Poznań, Poland; (E.M.-K.); (P.B.)
| |
Collapse
|
94
|
Mera-Charria A, Nieto-Lopez F, Francès MP, Arbex PM, Vila-Vecilla L, Russo V, Silva CCV, De Souza GT. Genetic variant panel allows predicting both obesity risk, and efficacy of procedures and diet in weight loss. Front Nutr 2023; 10:1274662. [PMID: 38035352 PMCID: PMC10687570 DOI: 10.3389/fnut.2023.1274662] [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: 08/08/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023] Open
Abstract
Purpose Obesity is a multifactorial condition with a relevant genetic correlation. Recent advances in genomic research have identified several single nucleotide polymorphisms (SNPs) in genes such as FTO, MCM6, HLA, and MC4R, associated with obesity. This study aimed to evaluate the association of 102 SNPs with BMI and weight loss treatment response in a multi-ethnic population. Methods The study analyzed 9,372 patients for the correlation between SNPs and BMI (dataset A). The correlation between SNP and weight loss was accessed in 474 patients undergoing different treatments (dataset B). Patients in dataset B were further divided into 3 categories based on the type of intervention: dietary therapy, intragastric balloon procedures, or surgeries. SNP association analysis and multiple models of inheritance were performed. Results In dataset A, ten SNPs, including rs9939609 (FTO), rs4988235 (MCM6), and rs2395182 (HLA), were significantly associated with increased BMI. Additionally, other four SNPs, rs7903146 (TCF7L2), (rs6511720), rs5400 (SLC2A2), and rs7498665 (SH2B1), showed sex-specific correlation. For dataset B, SNPs rs2016520 (PPAR-Delta) and rs2419621 (ACSL5) demonstrated significant correlation with weight loss for all treatment types. In patients who adhered to dietary therapy, SNPs rs6544713 (ABCG8) and rs762551 (CYP1A2) were strongly correlated with weight loss. Patients undergoing surgical or endoscopic procedures exhibited differential correlations with several SNPs, including rs1801725 (CASR) and rs12970134 (MC4R), and weight loss. Conclusion This study provides valuable insights into the genetic factors influencing BMI and weight loss response to different treatments. The findings highlight the potential for personalized weight management approaches based on individual genetic profiles.
Collapse
Affiliation(s)
| | - Francisco Nieto-Lopez
- Dorsia Clinics, Madrid, Spain
- Catedra UCAM Dorsia, Catholic University San Antonio of Murcia, Guadalupe, Spain
| | | | | | | | | | | | | |
Collapse
|
95
|
Du X, Yan Y, Yu J, Zhu T, Huang CC, Zhang L, Shan X, Li R, Dai Y, Lv H, Zhang XY, Feng J, Li WG, Luo Q, Li F. SH2B1 Tunes Hippocampal ERK Signaling to Influence Fluid Intelligence in Humans and Mice. RESEARCH (WASHINGTON, D.C.) 2023; 6:0269. [PMID: 38434247 PMCID: PMC10907025 DOI: 10.34133/research.0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/19/2023] [Indexed: 03/05/2024]
Abstract
Fluid intelligence is a cognitive domain that encompasses general reasoning, pattern recognition, and problem-solving abilities independent of task-specific experience. Understanding its genetic and neural underpinnings is critical yet challenging for predicting human development, lifelong health, and well-being. One approach to address this challenge is to map the network of correlations between intelligence and other constructs. In the current study, we performed a genome-wide association study using fluid intelligence quotient scores from the UK Biobank to explore the genetic architecture of the associations between obesity risk and fluid intelligence. Our results revealed novel common genetic loci (SH2B1, TUFM, ATP2A1, and FOXO3) underlying the association between fluid intelligence and body metabolism. Surprisingly, we demonstrated that SH2B1 variation influenced fluid intelligence independently of its effects on metabolism but partially mediated its association with bilateral hippocampal volume. Consistently, selective genetic ablation of Sh2b1 in the mouse hippocampus, particularly in inhibitory neurons, but not in excitatory neurons, significantly impaired working memory, short-term novel object recognition memory, and behavioral flexibility, but not spatial learning and memory, mirroring the human intellectual performance. Single-cell genetic profiling of Sh2B1-regulated molecular pathways revealed that Sh2b1 deletion resulted in aberrantly enhanced extracellular signal-regulated kinase (ERK) signaling, whereas pharmacological inhibition of ERK signaling reversed the associated behavioral impairment. Our cross-species study thus provides unprecedented insight into the role of SH2B1 in fluid intelligence and has implications for understanding the genetic and neural underpinnings of lifelong mental health and well-being.
Collapse
Affiliation(s)
- Xiujuan Du
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Yuhua Yan
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education),
School of Life Sciences, East China Normal University, Shanghai 200062, China
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science,
Fudan University, Shanghai 200032, China
| | - Juehua Yu
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Center for Experimental Studies and Research,
The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Tailin Zhu
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education),
School of Life Sciences, East China Normal University, Shanghai 200062, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| | - Chu-Chung Huang
- Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science,
East China Normal University, Shanghai 200062, China
| | - Lingli Zhang
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
| | - Xingyue Shan
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education),
School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Ren Li
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Yuan Dai
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
| | - Hui Lv
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
| | - Xiao-Yong Zhang
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Jianfeng Feng
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science,
Fudan University, Shanghai 200032, China
| | - Qiang Luo
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Fei Li
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| |
Collapse
|
96
|
Carbeck K, Arcese P, Lovette I, Pruett C, Winker K, Walsh J. Candidate genes under selection in song sparrows co-vary with climate and body mass in support of Bergmann's Rule. Nat Commun 2023; 14:6974. [PMID: 37935683 PMCID: PMC10630373 DOI: 10.1038/s41467-023-42786-2] [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: 01/10/2023] [Accepted: 10/19/2023] [Indexed: 11/09/2023] Open
Abstract
Ecogeographic rules denote spatial patterns in phenotype and environment that may reflect local adaptation as well as a species' capacity to adapt to change. To identify genes underlying Bergmann's Rule, which posits that spatial correlations of body mass and temperature reflect natural selection and local adaptation in endotherms, we compare 79 genomes from nine song sparrow (Melospiza melodia) subspecies that vary ~300% in body mass (17 - 50 g). Comparing large- and smaller-bodied subspecies revealed 9 candidate genes in three genomic regions associated with body mass. Further comparisons to the five smallest subspecies endemic to California revealed eight SNPs within four of the candidate genes (GARNL3, RALGPS1, ANGPTL2, and COL15A1) associated with body mass and varying as predicted by Bergmann's Rule. Our results support the hypothesis that co-variation in environment, body mass and genotype reflect the influence of natural selection on local adaptation and a capacity for contemporary evolution in this diverse species.
Collapse
Affiliation(s)
- Katherine Carbeck
- Department of Forest and Conservation Sciences, University of British Columbia, Vancouver, BC, T6T 1Z4, Canada.
| | - Peter Arcese
- Department of Forest and Conservation Sciences, University of British Columbia, Vancouver, BC, T6T 1Z4, Canada
| | - Irby Lovette
- Fuller Evolutionary Biology Program, Cornell Lab of Ornithology, Cornell University, Ithaca, NY, 14850, USA
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, 14850, USA
| | - Christin Pruett
- Department of Biology, Ouachita Baptist University, Arkadelphia, AR, 71998, USA
| | - Kevin Winker
- University of Alaska Museum, University of Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Jennifer Walsh
- Fuller Evolutionary Biology Program, Cornell Lab of Ornithology, Cornell University, Ithaca, NY, 14850, USA
| |
Collapse
|
97
|
Shen W, Ren S, Hou Y, Zuo Z, Liu S, Liu Z, Fu J, Wang H, Yang B, Zhao R, Chen Y, Yamamoto M, Xu Y, Zhang Q, Pi J. Single-nucleus RNA-sequencing reveals NRF1/NFE2L1 as a key factor determining the thermogenesis and cellular heterogeneity and dynamics of brown adipose tissues in mice. Redox Biol 2023; 67:102879. [PMID: 37716088 PMCID: PMC10511808 DOI: 10.1016/j.redox.2023.102879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023] Open
Abstract
Brown adipose tissue (BAT) is a major site of non-shivering thermogenesis in mammals and plays an important role in energy homeostasis. Nuclear factor-erythroid 2-related factor 1 (NFE2L1, also known as Nrf1), a master regulator of cellular metabolic homeostasis and numerous stress responses, has been found to function as a critical driver in BAT thermogenic adaption to cold or obesity by providing proteometabolic quality control. Our recent studies using adipocyte-specific Nfe2l1 knockout [Nfe2l1(f)-KO] mice demonstrated that NFE2L1-dependent transcription of lipolytic genes is crucial for white adipose tissue (WAT) homeostasis and plasticity. In the present study, we found that Nfe2l1(f)-KO mice develop an age-dependent whitening and shrinking of BAT, with signatures of down-regulation of proteasome, impaired mitochondrial function, reduced thermogenesis, pro-inflammation, and elevated regulatory cell death (RCD). Mechanistic studies revealed that deficiency of Nfe2l1 in brown adipocytes (BAC) primarily results in down-regulation of lipolytic genes, which decelerates lipolysis, making BAC unable to fuel thermogenesis. These changes lead to BAC hypertrophy, inflammation-associated RCD, and consequently cold intolerance. Single-nucleus RNA-sequencing of BAT reveals that deficiency of Nfe2l1 induces significant transcriptomic changes leading to aberrant expression of a variety of genes involved in lipid metabolism, proteasome, mitochondrial stress, inflammatory responses, and inflammation-related RCD in distinct subpopulations of BAC. Taken together, our study demonstrated that NFE2L1 serves as a vital transcriptional regulator that controls the lipid metabolic homeostasis in BAC, which in turn determines the metabolic dynamics, cellular heterogeneity and subsequently cell fates in BAT.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Suping Ren
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Yongyong Hou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Zhuo Zuo
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Shengnan Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Zhiyuan Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Jingqi Fu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Huihui Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Bei Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; School of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Rui Zhao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; School of Forensic Medicine, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Yanyan Chen
- The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Road, Heping Area, Shenyang, Liaoning, 110001, China
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, GA, 30322, USA.
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
98
|
Agarwal T, Lyngdoh T, Khadgawat R, Prabhakaran D, Chandak GR, Walia GK. Genetic architecture of adiposity measures among Asians: Findings from GWAS. Ann Hum Genet 2023; 87:255-273. [PMID: 37671428 DOI: 10.1111/ahg.12526] [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: 02/15/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/07/2023]
Abstract
Adiposity has gradually become a global public threat over the years with drastic increase in the attributable deaths and disability adjusted life years (DALYs). Given an increased metabolic risk among Asians as compared to Europeans for any given body mass index (BMI) and considering the differences in genetic architecture between them, the present review aims to summarize the findings from genome-wide scans for various adiposity indices and related anthropometric measures from Asian populations. The search for related studies, published till February 2022, were made on PubMed and GWAS Catalog using search strategy built with relevant keywords joined by Boolean operators. It was recorded that out of a total of 47 identified studies, maximum studies are from Korean population (n = 14), followed by Chinese (n = 7), and Japanese (n = 6). Nearly 200 loci have been identified for BMI, 660 for height, 16 for weight, 28 for circumferences (waist and hip), 32 for ratios (waist hip ratio [WHR] and thoracic hip ratio [THR]), 5 for body fat, 16 for obesity, and 28 for adiposity-related blood markers among Asians. It was observed that though, most of the loci were unique for each trait, there were 3 loci in common to BMI and WHR. Apart from validation of variants identified in European setting, there were many novel loci discovered in Asian populations. Notably, 125 novel loci form Asian studies have been reported for BMI, 47 for height, 5 for waist circumference, and 2 for adiponectin level to the existing knowledge of the genetic framework of adiposity and related measures. It is necessary to examine more advanced adiposity measures, specifically of relevance to abdominal adiposity, a major risk factor for cardiometabolic disorders among Asians. Moreover, in spite of being one continent, there is diversity among different ethnicities across Asia in terms of lifestyle, climate, geography, genetic structure and consequently the phenotypic manifestations. Hence, it is also important to consider ethnic specific studies for identifying and validating reliable genetic variants of adiposity measures among Asians.
Collapse
Affiliation(s)
- Tripti Agarwal
- Indian Institute of Public Health-Delhi, Public Health Foundation of India, Delhi, India
| | | | | | | | - Giriraj Ratan Chandak
- Genomic Research in Complex diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | |
Collapse
|
99
|
Wang H, Zhao X, Wen J, Wang C, Zhang X, Ren X, Zhang J, Li H, Muhatai G, Qu L. Comparative population genomics analysis uncovers genomic footprints and genes influencing body weight trait in Chinese indigenous chicken. Poult Sci 2023; 102:103031. [PMID: 37716235 PMCID: PMC10511812 DOI: 10.1016/j.psj.2023.103031] [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: 04/04/2023] [Revised: 07/27/2023] [Accepted: 08/11/2023] [Indexed: 09/18/2023] Open
Abstract
Body weight of chicken is a typical quantitative trait, which shows phenotypic variations due to selective breeding. Despite some QTL loci have been obtained, the body weight of native chicken breeds in different geographic regions varies greatly, its genetic basis remains unresolved questions. To address this issue, we analyzed 117 Chinese indigenous chickens from 10 breeds (Huiyang Bearded, Xinhua, Hotan Black, Baicheng You, Liyang, Yunyang Da, Jining Bairi, Lindian, Beijing You, Tibetan). We applied fixation index (FST) analysis to find selected genomic regions and genes associated with body weight traits. Our study suggests that NELL1, XYLT1, and NCAPG/LCORL genes are strongly selected in the body weight trait of Chinese indigenous chicken breeds. In addition, the IL1RAPL1 gene was strongly selected in large body weight chickens, while the PCDH17 and CADM2 genes were strongly selected in small body weight chickens. This result suggests that the patterns of genetic variation of native chicken and commercial chicken, and/or distinct local chicken breeds may follow different evolutionary mechanisms.
Collapse
Affiliation(s)
- Huie Wang
- Xinjiang Production & Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, College of Animal Science and Technology, Tarim University, Alar 843300, China
| | - Xiurong Zhao
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Junhui Wen
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chengqian Wang
- Xinjiang Production & Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, College of Animal Science and Technology, Tarim University, Alar 843300, China
| | - Xinye Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xufang Ren
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jinxin Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Haiying Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830000, China
| | - Gemingguli Muhatai
- Xinjiang Production & Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, College of Animal Science and Technology, Tarim University, Alar 843300, China
| | - Lujiang Qu
- Xinjiang Production & Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, College of Animal Science and Technology, Tarim University, Alar 843300, China; State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
100
|
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
|