1
|
Watts LM, Sparkes PC, Dewhurst HF, Guilfoyle SE, Pollard AS, Komla-Ebri D, Butterfield NC, Williams GR, Bassett JHD. The GWAS candidate far upstream element binding protein 3 (FUBP3) is required for normal skeletal growth, and adult bone mass and strength in mice. Bone 2025; 195:117472. [PMID: 40139337 DOI: 10.1016/j.bone.2025.117472] [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: 12/31/2024] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Bone mineral density (BMD) and height are highly heritable traits for which hundreds of genetic loci have been linked through genome wide association studies (GWAS). FUBP3 is a DNA and RNA binding protein best characterised as a transcriptional regulator of c-Myc, but little is known about its role in vivo. Single nucleotide polymorphisms in FUBP3 at the 9q34.11 locus have been associated with BMD, fracture and height in multiple GWAS, but FUBP3 has no previously established role in the skeleton. We analysed Fubp3-deficient mice to determine the consequence of FUBP3 deficiency in vivo. Mice lacking Fubp3 had reduced survival to adulthood and impaired skeletal growth. Bone mass was decreased, most strikingly in the vertebrae, with altered trabecular micro-architecture. Fubp3 deficient bones were also weak. These data provide the first functional demonstration that Fubp3 is required for normal skeletal growth and development and maintenance of adult bone structure and strength, indicating that FUBP3 contributes to the GWAS association of 9q34.11 with variation in height, BMD and fracture.
Collapse
Affiliation(s)
- Laura M Watts
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Penny C Sparkes
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Hannah F Dewhurst
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Siobhan E Guilfoyle
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Andrea S Pollard
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Davide Komla-Ebri
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Natalie C Butterfield
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
2
|
Kong J, Han X, Wei C. Causal relationship between metabolic dysfunction-associated fatty liver disease and endotoxin biomarkers: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42311. [PMID: 40388727 PMCID: PMC12091621 DOI: 10.1097/md.0000000000042311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 04/14/2025] [Indexed: 05/21/2025] Open
Abstract
Although the relationship among lipopolysaccharides (LPS), LPS-binding proteins, and metabolic dysfunction-associated fatty liver disease (MAFLD) is widely studied, no conclusive evidence is available. In this study, we used mendelian randomization (MR) to study the causal relationship of LPS, LPS-binding proteins, and MAFLD. Using bidirectional two-sample MR method, we evaluated data from the genome wide association study; for this analysis, nonalcoholic fatty liver disease (NAFLD), liver fat percentage, and other metabolic syndromes were employed as outcomes. Furthermore, MR analysis mainly involved the inverse variance weighted method. Heterogeneity and pleiotropy tests were also conducted. LPS was found to have a causal relationship with NAFLD, obesity, high density lipoprotein cholesterol, and TG levels. Furthermore, TG levels and LBP had significant causal relationships. This study mainly concluded that LPS is a risk factor for NAFLD, obesity, high density lipoprotein cholesterol, and TG, corroborating it's the LPS role in MAFLD pathogenesis. Hence, optimizing the gut microbiota using proper diet, probiotics, or fecal microbiota transplantation may help to reduce inflammation and (IR), thereby improving lipid and glucose metabolism disorders. Although a causal relationship between TG and LBP was observed, further studies are required to determine a specific mechanism.
Collapse
Affiliation(s)
- Jingwen Kong
- Jining Medical University, Jining, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Xixi Han
- Jining Medical University, Jining, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wei
- Jining Medical University, Jining, China
| |
Collapse
|
3
|
Müller TD, Adriaenssens A, Ahrén B, Blüher M, Birkenfeld AL, Campbell JE, Coghlan MP, D'Alessio D, Deacon CF, DelPrato S, Douros JD, Drucker DJ, Figueredo Burgos NS, Flatt PR, Finan B, Gimeno RE, Gribble FM, Hayes MR, Hölscher C, Holst JJ, Knerr PJ, Knop FK, Kusminski CM, Liskiewicz A, Mabilleau G, Mowery SA, Nauck MA, Novikoff A, Reimann F, Roberts AG, Rosenkilde MM, Samms RJ, Scherer PE, Seeley RJ, Sloop KW, Wolfrum C, Wootten D, DiMarchi RD, Tschöp MH. Glucose-dependent insulinotropic polypeptide (GIP). Mol Metab 2025; 95:102118. [PMID: 40024571 PMCID: PMC11931254 DOI: 10.1016/j.molmet.2025.102118] [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: 12/06/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glucose-dependent insulinotropic polypeptide (GIP) was the first incretin identified and plays an essential role in the maintenance of glucose tolerance in healthy humans. Until recently GIP had not been developed as a therapeutic and thus has been overshadowed by the other incretin, glucagon-like peptide 1 (GLP-1), which is the basis for several successful drugs to treat diabetes and obesity. However, there has been a rekindling of interest in GIP biology in recent years, in great part due to pharmacology demonstrating that both GIPR agonism and antagonism may be beneficial in treating obesity and diabetes. This apparent paradox has reinvigorated the field, led to new lines of investigation, and deeper understanding of GIP. SCOPE OF REVIEW In this review, we provide a detailed overview on the multifaceted nature of GIP biology and discuss the therapeutic implications of GIPR signal modification on various diseases. MAJOR CONCLUSIONS Following its classification as an incretin hormone, GIP has emerged as a pleiotropic hormone with a variety of metabolic effects outside the endocrine pancreas. The numerous beneficial effects of GIPR signal modification render the peptide an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, drug-induced nausea and both bone and neurodegenerative disorders.
Collapse
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Walther-Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University Munich (LMU), Germany.
| | - Alice Adriaenssens
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Bo Ahrén
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen 72076, Germany; Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Matthew P Coghlan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - David D'Alessio
- Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Carolyn F Deacon
- School of Biomedical Sciences, Ulster University, Coleraine, UK; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefano DelPrato
- Interdisciplinary Research Center "Health Science", Sant'Anna School of Advanced Studies, Pisa, Italy
| | | | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Natalie S Figueredo Burgos
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Brian Finan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Ruth E Gimeno
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Fiona M Gribble
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Matthew R Hayes
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian Hölscher
- Neurodegeneration Research Group, Henan Academy of Innovations in Medical Science, Xinzheng, China
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Patrick J Knerr
- Indianapolis Biosciences Research Institute, Indianapolis, IN, USA
| | - Filip K Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christine M Kusminski
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Guillaume Mabilleau
- Univ Angers, Nantes Université, ONIRIS, Inserm, RMeS UMR 1229, Angers, France; CHU Angers, Departement de Pathologie Cellulaire et Tissulaire, Angers, France
| | | | - Michael A Nauck
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany
| | - Frank Reimann
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Anna G Roberts
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Ricardo J Samms
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Philip E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kyle W Sloop
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | | | - Matthias H Tschöp
- Helmholtz Munich, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
4
|
Nikkilä R, Mäkitie A, Joensuu H, Markkanen S, Elenius K, Monni O, Palotie A, Saarentaus E, Salo T, Bizaki-Vallaskangas A. Novel Genetic Risk Variants Associated with Oral Tongue Squamous Cell Carcinoma. Head Neck Pathol 2025; 19:45. [PMID: 40278994 PMCID: PMC12031715 DOI: 10.1007/s12105-025-01784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025]
Abstract
PURPOSE Limited data from genome-wide association studies (GWAS) focusing on oral tongue squamous cell carcinoma (OTSCC) are available. The present study was conducted to explore genetic associations for OTSCC. METHODS A GWAS on 376 cases of OTSCC was conducted using the FinnGen Data Freeze-12 dataset. The case-cohort included 205 males and 171 females. Cases with malignancies involving the base of the tongue or lingual tonsil were excluded from the case-cohort. Individuals with no recorded history of malignancy were used as controls (n = 407,067). A Phenome-wide association study (PheWAS) was performed for the lead variants to assess their co-associations with other cancers. RESULTS GWAS analysis identified three genome-wide significant loci associated with OTSCC (p < 5 × 10-8), located at 5p15.33 (rs27067 near gene LINC01511), 10q24 (rs1007771191 near RPS3AP36), and 20p12.3 (rs1438070080 near PLCB1), respectively. PheWAS showed associations of rs27067 mainly with prostate cancer (OR = 1.06, p = 5.41 × 10-7), and seborrheic keratosis (OR = 1.11, p = 1.51 × 10-11). A co-directional effect with melanoma was also observed (OR = 0.93, p = 6.24 × 10-5). CONCLUSION The GWAS detected two novel genetic associations with OTSCC. Further research is needed to identify the genes at these loci that contribute to the molecular pathogenesis of OTSCC.
Collapse
Affiliation(s)
- Rayan Nikkilä
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer and Research, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heikki Joensuu
- Department of Oncology, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Saara Markkanen
- Department of Otolaryngology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- The Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Klaus Elenius
- Institute of Biomedicine, and MediCity Research Laboratory, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Oncology, Turku University Hospital, Turku, Finland
| | - Outi Monni
- Department of Oncology, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland and the Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology, and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Elmo Saarentaus
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland and the Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, HUS Helsinki University Hospital, Helsinki, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Argyro Bizaki-Vallaskangas
- Department of Otolaryngology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- The Wellbeing Services County of Pirkanmaa, Tampere, Finland.
| |
Collapse
|
5
|
Zhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, et alZhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, Kooperberg C, Minster RL, Mitchell BD, Nouraie M, Psaty BM, Raffield LM, Reiner AP, Rich SS, Rotter JI, Benjamin Shoemaker M, Smith NL, Taylor KD, Telen MJ, Weiss ST, Zhang Y, Heard-Costa N, Sun YV, Lin X, Cupples LA, Lange LA, Liu CT, Loos RJF, North KE, Justice AE. Whole genome sequencing analysis of body mass index identifies novel African ancestry-specific risk allele. Nat Commun 2025; 16:3470. [PMID: 40216759 PMCID: PMC11992084 DOI: 10.1038/s41467-025-58420-2] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Obesity is a major public health crisis associated with high mortality rates. Previous genome-wide association studies (GWAS) investigating body mass index (BMI) have largely relied on imputed data from European individuals. This study leveraged whole-genome sequencing (WGS) data from 88,873 participants from the Trans-Omics for Precision Medicine (TOPMed) Program, of which 51% were of non-European population groups. We discovered 18 BMI-associated signals (P < 5 × 10-9), including two secondary signals. Notably, we identified and replicated a novel low-frequency single nucleotide polymorphism (SNP) in MTMR3 that was common in individuals of African descent. Using a diverse study population, we further identified two novel secondary signals in known BMI loci and pinpointed two likely causal variants in the POC5 and DMD loci. Our work demonstrates the benefits of combining WGS and diverse cohorts in expanding current catalog of variants and genes confer risk for obesity, bringing us one step closer to personalized medicine.
Collapse
Affiliation(s)
- Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanfei Xu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kendra R Ferrier
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Mariah Meyer
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Shreyash Gupta
- Population Health Sciences, Geisinger, Danville, PA, USA
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zilin Li
- Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
- School of Mathematics and Statistics and KLAS, Northeast Normal University, Changchun, Jilin, China
| | - Matthew A Allison
- Department of Family Medicine, Division of Preventive Medicine, The University of California San Diego, La Jolla, CA, USA
| | - Diane M Becker
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jai G Broome
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Erin J Buth
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Kyong-Mi Chang
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sameer Chavan
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yen-Feng Chiu
- Institute of Population Health Sciences, National Health Research Institutes, Taipei, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, Division of Metabolism/Endocrinology, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthew P Conomos
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Dawn L DeMeo
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravindranath Duggirala
- Life Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
- Department of Health and Behavioral Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Celeste Eng
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Alison E Fohner
- Epidemiology, Institute of Public Health Genetics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Barry I Freedman
- Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiuqing Guo
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Chris Haiman
- Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin D Heavner
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - James E Hixson
- Department of Epidemiology, School of Public Health, UTHealth Houston, Houston, TX, USA
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Brian D Hobbs
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donglei Hu
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Chii-Min Hwu
- Department of Medicine, Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, Taiwan
| | | | - Deepti Jain
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Rita R Kalyani
- Department of Medicine, Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Ethan M Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael LeNoir
- Department of Pediatrics, Bay Area Pediatrics, Oakland, CA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Merry-Lynn N McDonald
- Department of Medicine, Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caitlin P McHugh
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Take Naseri
- Naseri & Associates Public Health Consultancy Firm and Family Health Clinic, Apia, Samoa
- International Health Institute, Brown University, Providence, RI, USA
| | - Jeffrey O'Connell
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, MD, USA
| | - Christopher J O'Donnell
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James A Perry
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D C Rao
- Center for Biostatistics and Data Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Elizabeth A Regan
- Department of Medicine, Rheumatology, National Jewish Health, Denver, CO, USA
| | | | - Dan M Roden
- Medicine, Pharmacology, and Biomedical Informatics, Clinical Pharmacology and Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | | | - Zeyuan Wang
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Daniel E Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
- Diabaetes Translational Research Center, Indiana University, Indianapolis, IN, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Peter W F Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lisa R Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary T Yoneda
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics, Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Donna K Arnett
- Department of Epidemiology, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Esteban G Burchard
- Bioengineering and Therapeutic Sciences and Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yii-Der Ida Chen
- Department of Medical Genetics, Genomic Outcomes, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Myriam Fornage
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Victor R Gordeuk
- Department of Medicine, School of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lifang Hou
- Northwestern University, Chicago, IL, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ryan L Minster
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland, Baltimore, MD, USA
| | - Mehdi Nouraie
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - M Benjamin Shoemaker
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas L Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Kent D Taylor
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marilyn J Telen
- Department of Medicine, Division of Hematology, Duke University School of Medical, Durham, NC, USA
| | - Scott T Weiss
- Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, USA
| | - Yingze Zhang
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nancy Heard-Costa
- Framingham Heart Study, School of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - L Adrienne Cupples
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne E Justice
- Population Health Sciences, Geisinger, Danville, PA, USA.
| |
Collapse
|
6
|
Qi M, Wei J, Zhang M, Jiao C, He C, Sui L, Ma S, Mao Z, Pan X, Zhu X. THE CAUSAL ASSOCIATION OF CARDIOMETABOLIC DISEASES AND SEPSIS-RELATED OUTCOMES: A MENDELIAN RANDOMIZATION AND POPULATION STUDY. Shock 2025; 63:579-586. [PMID: 39965633 DOI: 10.1097/shk.0000000000002538] [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: 02/20/2025]
Abstract
ABSTRACT Objective: The causality between cardiometabolic disease (CMD) and sepsis has remained largely unknown. To elucidate this, we conducted a Mendelian randomization (MR) and population study. Methods: First, we used univariable and multivariable MR analyses to investigate causal associations between CMD and sepsis-related outcomes. We obtained genome-wide association study summary from both the MRC Integrative Epidemiology Unit and the FinnGen consortium. Subsequently, a two-step mediation MR analysis was performed to explore mediators. Afterward, we conducted an observational study using the Medical Information Mart for Intensive Care IV database, in which multivariable logistic regression models were utilized to examine the relationship between CMD and sepsis-related outcomes. Results: In the MR study, type 2 diabetes mellitus (OR = 1.058, 95% CI = 1.017-1.100, P = 0.005), obesity (OR = 1.113, 95% CI = 1.057-1.172, P < 0.001), and heart failure (HF) (OR = 1.178, 95% CI = 1.063-1.305, P = 0.002) were independently causally related to sepsis. Obesity (OR = 1.215, 95% CI = 1.027-1.437, P = 0.023) and HF (OR = 1.494, 95% CI = 1.080-2.065, P = 0.015) also showed independent causal associations with sepsis critical care admission. Mediation MR analysis identified 23 blood metabolites potentially causally linked to sepsis ( P < 0.05), yet none mediated the relationship between CMD and sepsis. In the observational study, we found associations between sepsis and several conditions including type 2 diabetes mellitus, obesity, hypertension, stroke, HF, and hyperlipidemia after adjusting for confounding factors. Moreover, hypertension, stroke, HF, coronary artery disease, and hyperlipidemia were linked to sepsis critical care admission. Conclusion: This study has, for the first time, revealed indicative evidence of a causal relationship between CMD and sepsis through observational and genetic evidence. Taken together, clinical attention to sepsis may be warranted among patients with CMD.
Collapse
Affiliation(s)
- Mengmeng Qi
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chucheng Jiao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liutao Sui
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhi Mao
- Department of Critical Care Medicine, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
7
|
Wuni R, Curi-Quinto K, Liu L, Espinoza D, Aquino AI, Del Valle-Mendoza J, Aguilar-Luis MA, Murray C, Nunes R, Methven L, Lovegrove JA, Penny M, Favara M, Sánchez A, Vimaleswaran KS. Interaction between genetic risk score and dietary carbohydrate intake on high-density lipoprotein cholesterol levels: Findings from the study of obesity, nutrition, genes and social factors (SONGS). Clin Nutr ESPEN 2025; 66:83-92. [PMID: 39800136 DOI: 10.1016/j.clnesp.2024.12.027] [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: 09/27/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS Cardiometabolic traits are complex interrelated traits that result from a combination of genetic and lifestyle factors. This study aimed to assess the interaction between genetic variants and dietary macronutrient intake on cardiometabolic traits [body mass index, waist circumference, total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol, triacylglycerol, systolic blood pressure, diastolic blood pressure, fasting serum glucose, fasting serum insulin, and glycated haemoglobin]. METHODS This cross-sectional study consisted of 468 urban young adults aged 20 ± 1 years, and it was conducted as part of the Study of Obesity, Nutrition, Genes and Social factors (SONGS) project, a sub-study of the Young Lives study. Thirty-nine single nucleotide polymorphisms (SNPs) known to be associated with cardiometabolic traits at a genome-wide significance level (P < 5 × 10-8) were used to construct a genetic risk score (GRS). RESULTS There were no significant associations between the GRS and any of the cardiometabolic traits. However, a significant interaction was observed between the GRS and carbohydrate intake on HDL-C concentration (Pinteraction = 0.0007). In the first tertile of carbohydrate intake (≤327 g/day), participants with a high GRS (>37 risk alleles) had a higher concentration of HDL-C than those with a low GRS (≤37 risk alleles) [Beta = 0.06 mmol/L, 95 % confidence interval (CI), 0.01-0.10; P = 0.018]. In the third tertile of carbohydrate intake (>452 g/day), participants with a high GRS had a lower concentration of HDL-C than those with a low GRS (Beta = -0.04 mmol/L, 95 % CI -0.01 to -0.09; P = 0.027). A significant interaction was also observed between the GRS and glycaemic load (GL) on the concentration of HDL-C (Pinteraction = 0.002). For participants with a high GRS, there were lower concentrations of HDL-C across tertiles of GL (Ptrend = 0.017). There was no significant interaction between the GRS and glycaemic index on the concentration of HDL-C, and none of the other GRS∗macronutrient interactions were significant. CONCLUSIONS Our results suggest that young adults who consume a higher carbohydrate diet and have a higher GRS have a lower HDL-C concentration, which in turn is linked to cardiovascular diseases, and indicate that personalised nutrition strategies targeting a reduction in carbohydrate intake might be beneficial for these individuals.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Katherine Curi-Quinto
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru.
| | - Litai Liu
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Dianela Espinoza
- Group for the Analysis of Development (GRADE), Lima, 15063, Peru.
| | - Anthony I Aquino
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru
| | - Juana Del Valle-Mendoza
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru; Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, 15087, Peru.
| | - Miguel Angel Aguilar-Luis
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru; Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, 15087, Peru.
| | - Claudia Murray
- Department of Real Estate and Planning, University of Reading, Reading, RG6 6UD, UK.
| | - Richard Nunes
- Department of Real Estate and Planning, University of Reading, Reading, RG6 6UD, UK.
| | - Lisa Methven
- Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK; Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, RG6 6AP, UK.
| | - Mary Penny
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru.
| | - Marta Favara
- Oxford Department of International Development, University of Oxford, Oxford, OX1 3TB, UK.
| | - Alan Sánchez
- Group for the Analysis of Development (GRADE), Lima, 15063, Peru; Oxford Department of International Development, University of Oxford, Oxford, OX1 3TB, UK.
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK; Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, RG6 6AP, UK.
| |
Collapse
|
8
|
Zhu H, Wang Y, Li L, Wang L, Zhang H, Jin X. Cell-free DNA from clinical testing as a resource of population genetic analysis. Trends Genet 2025; 41:330-344. [PMID: 39578178 DOI: 10.1016/j.tig.2024.10.007] [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: 07/15/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
As a noninvasive biomarker, cell-free DNA (cfDNA) has achieved remarkable success in clinical applications. Notably, cfDNA is essentially DNA, and conducting whole-genome sequencing (WGS) can yield a wealth of genetic information. These invaluable data should not be confined to one-time use; instead, they should be leveraged for more comprehensive population genetic analysis, including genetic variation spectrum, population structure and genetic selection, and genome-wide association studies (GWASs), among others. Such research findings can, in turn, facilitate clinical practice, enabling more advanced and accurate disease predictions. This review explores the advantages, challenges, and current research areas of cfDNA in population genetics. We hope that this review can serve as a new chapter in the repurposing of cfDNA sequence data generated from clinical testing in population genetics.
Collapse
Affiliation(s)
- Huanhuan Zhu
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Yu Wang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China; College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Wang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Haiqiang Zhang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China; School of Medicine, South China University of Technology, Guangzhou 510641, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
9
|
Zhou R, Zhang Y, Wang J, Huang H, Liao T, Lai W, Ju Y, Ouyang M. Establishing the relationships between obesity and genetically predicted serum micronutrient levels: a multivariable Mendelian randomization analysis. Eat Weight Disord 2025; 30:33. [PMID: 40158042 PMCID: PMC11954692 DOI: 10.1007/s40519-025-01730-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/10/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Previous observational studies have indicated that circulating micronutrients may influence obesity risk. This study aimed to explore the causal relationship between micronutrient levels and obesity through multivariable Mendelian randomization (MR) analysis. METHODS Single nucleotide polymorphisms (SNPs) significantly associated with 15 micronutrients (selenium, zinc, copper, calcium, beta-carotene, folate, iron, magnesium, potassium, and vitamins A, B6, B12, C, D, and E) from published genome-wide association studies (GWAS) were used as instrumental variables (IVs). Three obesity-related datasets were obtained from the GWAS. Inverse variance weighted (IVW) is the main method used for MR analysis. Leave-one-out analysis, MR-Pleiotropy Residual Sum and Outlier method (MR-PRESSO), weighted median, and MR-Egger method were used to assess pleiotropy and heterogeneity. RESULTS Genetically predicted levels of circulating selenium and calcium are causally related to the risk of obesity (calcium odds ratio [OR]: 1.478, 95% confidence interval [CI] 1.128-1.935, p = 0.005; selenium OR: 1.478, 95% CI 1.128-1.935, p = 0.005). Multivariate MR analysis suggested a causal relationship between circulating selenium and calcium levels and obesity risk (calcium OR: 1.625, 95% CI 1.260-2.097; selenium OR: 1.080, 95% CI 1.003-1.163, p = 0.041). The p-value obtained in the Cochrane Q test, MR-Egger intercept test, and MR-PRESSO were > 0.05, suggesting no significant evidence of pleiotropy or heterogeneity. CONCLUSION Our study revealed, for the first time, a positive correlation between elevated circulating calcium and selenium levels and an increased obesity risk. These findings provide valuable insights into obesity's underlying mechanisms. Nevertheless, further large-scale clinical studies are required to confirm our results. LEVEL OF EVIDENCE Level III, Mendelian randomization.
Collapse
Affiliation(s)
- Rui Zhou
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China
| | - Yanxiang Zhang
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China
| | - Jiazhi Wang
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China
| | - Huacong Huang
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China
| | - Tianyou Liao
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China
| | - Weisheng Lai
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China
| | - Yongle Ju
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China.
| | - Manzhao Ouyang
- Surgical Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, No. 1 Jiazi Road, Shunde District, Foshan, 528399, Guangdong, China.
| |
Collapse
|
10
|
Lee GW, Chen JJ, Wang CH, Chang SN, Chiu FC, Huang PS, Chua SK, Chuang EY, Tsai CT. Identification of a new genetic locus associated with atrial fibrillation in the Taiwanese population by genome-wide and transcriptome-wide association studies. Europace 2025; 27:euaf042. [PMID: 40036802 PMCID: PMC11952963 DOI: 10.1093/europace/euaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
AIMS Genome-wide association studies (GWASs) identified common single-nucleotide polymorphisms (SNPs) in more than 100 genomic regions associated with atrial fibrillation (AF). We aimed to identify novel AF genes in Taiwanese population by multi-stage GWAS. METHODS AND RESULTS In exploratory stage, we did GWAS with whole-genome genotypes (4 512 191 SNPs) in 516 patients with AF from the National Taiwan University AF Registry and 5160 normal sinus rhythm controls from the Taiwan Biobank. Significant loci were replicated in 1002 independent patients and 2003 controls and in the UK Biobank. Expression quantitative trait locus (eQTL) mapping and transcriptome-wide association study (TWAS) were performed to implicate functional significance. Stage I GWAS revealed three loci associated with AF with a genome-wide significance level, including one close to PITX2 gene (chromosome 4q25, rs2723329, minor allele frequency [MAF] 0.50 vs. 0.41, P = 1.53 × 10-10), another close to RAP1A gene (also to previous KCND3; chromosome 1p13.2, rs7525578, MAF 0.17 vs. 0.07, P = 1.24 × 10-26), and one novel locus close to HNF4G gene (chromosome 8q21.13, rs2980218, MAF 0.44 vs. 0.35, P = 2.19 × 10-9). They were validated in Stage II population. The eQTL analyses showed significant colocalization of 1p13.2 locus with RAP1A gene expression in fibroblasts and 8q21.13 locus with HNF4G expression in lymphocytes. There is a significant association of RAP1A gene expression in fibroblasts and HNF4G in lymphocytes and brain with AF in TWAS. CONCLUSION Genome-wide association study in Taiwan revealed PITX2 and RAP1A/KCND3 loci and novel AF locus (HNF4G) with the most significant locus in the RAP1A locus. RAP1A and HNF4G genes may implicate fibrosis, metabolic, and neurogenic pathways in pathogenesis of AF.
Collapse
Affiliation(s)
- Guan-Wei Lee
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Jien-Jiun Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital Yun-Ling Branch, Dou-Liu City, Taiwan
| | - Chih-Hsien Wang
- Division of Cardiovascular Surgery, Department of Surgery, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Nan Chang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital Yun-Ling Branch, Dou-Liu City, Taiwan
| | - Fu-Chun Chiu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital Yun-Ling Branch, Dou-Liu City, Taiwan
| | - Pang-Shuo Huang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital Yun-Ling Branch, Dou-Liu City, Taiwan
| | - Su-Kiat Chua
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Eric Y Chuang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
- Bioinformatics and Biostatistics Core, Centers of Genomic and Precision Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
- Research and Development Center for Medical Devices, National Taiwan University, No. 7, Chung-Shan S Rd, Taipei 100, Taiwan
| | - Chia-Ti Tsai
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Geriatrics and Gerontology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| |
Collapse
|
11
|
Kitamoto T, Kitamoto A. Integrative proteomic and lipidomic analysis of GNB1 and SCARB2 knockdown in human subcutaneous adipocytes. PLoS One 2025; 20:e0319163. [PMID: 40127054 PMCID: PMC11932494 DOI: 10.1371/journal.pone.0319163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/28/2025] [Indexed: 03/26/2025] Open
Abstract
Obesity, a global public health concern, is influenced by various factors, including genetic predispositions. Although many obesity-associated genes have been identified through genome-wide association studies (GWAS), the molecular mechanisms linking these genes to adipose tissue function remain largely unexplored. This study integrates proteomic data on adipocyte fat accumulation with GWAS data on obesity to unravel the roles of the identified key candidate genes - G protein subunit beta 1 (GNB1) and scavenger receptor class B member 2 (SCARB2) - involved in fat accumulation. We utilized RNA interference to knock down GNB1 and SCARB2 in human subcutaneous adipocytes, followed by lipidome and proteome analyses using mass spectrometry. Knockdown of these genes resulted in a reduction in lipid droplet accumulation, indicating their role in adipocyte lipid storage. Digital PCR confirmed effective gene knockdown, with GNB1 and SCARB2 mRNA levels significantly reduced. In total, the lipidomic analysis identified 96 lipid species with significant alterations. GNB1 knockdown resulted in a decrease in cholesterol esters and an increase in phosphatidylcholines, phosphatidylinositols, and ceramides. SCARB2 knockdown also led to an increase in phosphatidylcholines, with a trend towards decreased triacylglycerols. Proteomic analysis revealed significant changes in proteins involved in lipid metabolism and adipocyte function, including PLPP1 and CDH13, which were upregulated following GNB1 knockdown, and HSPA8, which was downregulated. Conversely, SCARB2 knockdown resulted in the downregulation of PLPP1 and METTL7A, and the upregulation of PLIN2, HSPA8, NPC2, and SQSTM1. Our findings highlight the significant roles of GNB1 and SCARB2 in lipid metabolism and adipocyte function, providing insights that could inform therapeutic strategies targeting these regulatory genes in obesity.
Collapse
Affiliation(s)
- Takuya Kitamoto
- Advanced Research Facilities and Services, Division of Preeminent Research Supports, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Aya Kitamoto
- Advanced Research Facilities and Services, Division of Preeminent Research Supports, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
12
|
Chan M, Ogawa S. GPR139, an Ancient Receptor and an Emerging Target for Neuropsychiatric and Behavioral Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04828-2. [PMID: 40102345 DOI: 10.1007/s12035-025-04828-2] [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: 10/26/2024] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
GPR139 is an orphan G-protein-coupled receptor that is predominantly expressed in several midbrain regions, e.g., the habenula, striatum, and hypothalamus. GPR139 gene is highly conserved across vertebrate phylogenetic taxa, suggesting its fundamental importance in neurophysiology. Evidence from both animal studies and human genetic association studies has demonstrated that dysregulation of GPR139 expression and function is linked to aberrant behaviors, cognitive deficits, alterations in sleep and alertness, and substance abuse and withdrawal. Animal knockout models suggest that GPR139 plays an anti-opioid role by modulating the signaling activity of the μ-opioid receptor (MOR), as well as the intensity of withdrawal symptoms and nociception in behavioral paradigms. Modulation of GPR139 activity by surrogate agonists such as TAK-041 and JNJ-63533054 has shown promising results in experimental models; however, the use of TAK-041 in clinical trials has produced heterogeneous effects and has not met the intended primary endpoint. Here, we highlight current in vitro and in vivo studies of GPR139, its potential physiological roles, and therapeutic potential in the pathophysiology of neuropsychiatric and behavioral disorders. This review aims to focus on the current knowledge gaps to facilitate future studies that will contribute to the understanding of GPR139 as a therapeutic target for neuropsychiatric and behavioral disorders.
Collapse
Affiliation(s)
- Minyu Chan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia
| | - Satoshi Ogawa
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
13
|
Bazzazzadehgan S, Shariat-Madar Z, Mahdi F. Distinct Roles of Common Genetic Variants and Their Contributions to Diabetes: MODY and Uncontrolled T2DM. Biomolecules 2025; 15:414. [PMID: 40149950 PMCID: PMC11940602 DOI: 10.3390/biom15030414] [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: 12/30/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) encompasses a range of clinical manifestations, with uncontrolled diabetes leading to progressive or irreversible damage to various organs. Numerous genes associated with monogenic diabetes, exhibiting classical patterns of inheritance (autosomal dominant or recessive), have been identified. Additionally, genes involved in complex diabetes, which interact with environmental factors to trigger the disease, have also been discovered. These genetic findings have raised hopes that genetic testing could enhance diagnostics, disease surveillance, treatment selection, and family counseling. However, the accurate interpretation of genetic data remains a significant challenge, as variants may not always be definitively classified as either benign or pathogenic. Research to date, however, indicates that periodic reevaluation of genetic variants in diabetes has led to more consistent findings, with biases being steadily eliminated. This has improved the interpretation of variants across diverse ethnicities. Clinical studies suggest that genetic risk information may motivate patients to adopt behaviors that promote the prevention or management of T2DM. Given that the clinical features of certain monogenic diabetes types overlap with T2DM, and considering the significant role of genetic variants in diabetes, healthcare providers caring for prediabetic patients should consider genetic testing as part of the diagnostic process. This review summarizes current knowledge of the most common genetic variants associated with T2DM, explores novel therapeutic targets, and discusses recent advancements in the pharmaceutical management of uncontrolled T2DM.
Collapse
Affiliation(s)
- Shadi Bazzazzadehgan
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Zia Shariat-Madar
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| | - Fakhri Mahdi
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
14
|
Mohammadi A, Karimian A, Shokri K, Mohammadi A, Hazhir-Karzar N, Bahar R, Radfar A, Pakyari M, Tehrani B. RNA Therapies in Cardio-Kidney-Metabolic Syndrome: Advancing Disease Management. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10603-4. [PMID: 40080261 DOI: 10.1007/s12265-025-10603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Cardio-Kidney-Metabolic (CKM) Syndrome involves metabolic, kidney, and cardiovascular dysfunction, disproportionately affecting disadvantaged groups. Its staging (0-4) highlights the importance of early intervention. While current management targets hypertension, heart failure, dyslipidemia, and diabetes, RNA-based therapies offer innovative solutions by addressing molecular mechanisms of CKM Syndrome. Emerging RNA treatments, including antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs), show promise in slowing disease progression across CKM stages. For example, ASOs and siRNAs targeting ApoC-III and ANGPTL3 reduce triglycerides and LDL cholesterol, while siRNAs improve blood pressure control by targeting the renin-angiotensin-aldosterone system. Obesity treatments leveraging miRNAs and circRNAs tackle a key CKM risk factor. In heart failure and diabetes, RNA-based therapies improve cardiac function and glucose control, while early kidney disease trials show potential for RNAi in acute injury. Further research is essential to refine these therapies and ensure equitable access.
Collapse
Affiliation(s)
- Abbas Mohammadi
- Department of Internal Medicine, Valley Health System, Las Vegas, NV, USA.
| | - Azin Karimian
- Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kasra Shokri
- Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | - Rayeheh Bahar
- Department of Internal Medicine, Valley Health System, Las Vegas, NV, USA
| | - Azar Radfar
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mohammadreza Pakyari
- Department of Pathology, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
15
|
Gurung RL, Nangia C, Cai T, FitzGerald LM, McComish BJ, Liu E, Kaidonis G, Ridge B, Hewitt AW, Vote B, Verma N, Craig JE, Palmer CNA, Burdon KP, Meng W. Genome-Wide Association Study to Identify Genetic Variants Associated With Diabetic Maculopathy. Invest Ophthalmol Vis Sci 2025; 66:55. [PMID: 40136285 PMCID: PMC11951052 DOI: 10.1167/iovs.66.3.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose Diabetic maculopathy (including diabetic macular edema [DME]) is the leading cause of vision loss in people with diabetes. We aimed to identify the genetic determinants of diabetic maculopathy. Methods We conducted a genome-wide association study (GWAS) in two cohorts with a meta-analysis. The Australian cohort comprised 551 cases of DME and 599 controls recruited from the states of South Australia and Tasmania. The Scottish cohort comprised 1951 cases of diabetic maculopathy and 6541 controls from the Genetics of Diabetes Audit and Research in Tayside Scotland study (GoDARTS). Genotyping, imputation, and association analysis using logistic regression were conducted in each cohort, before combining summary statistics in a meta-analysis using the GWAMA package. Results A locus on chromosome 7 reached genome-wide significance in GoDARTS but showed the opposite direction of effect in the Australian cohort. The meta-analysis identified two suggestive associations (P < 5 × 10-6) for diabetic maculopathy risk with similar effect direction; one at chromosome 1 close to the RNU5E-1 gene and one at chromosome 13 upstream of the ERICH6B gene. The two loci were evaluated in silico for potential functional links to diabetic maculopathy. Both are located in regulatory regions and have annotations indicating regulatory functions. They are also expression quantitative trait locus (eQTLs) for genes plausibly involved in diabetic maculopathy pathogenesis, with links to folate metabolism and the regulation of VEGF. Conclusions The study suggests several promising SNPs and genes related to diabetic maculopathy risk. Despite being the largest genetic study of diabetic maculopathy to date, larger, homogeneous cohorts will be required to identify robust genetic risk loci for the disease.
Collapse
Affiliation(s)
- Rajya L. Gurung
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Charvi Nangia
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Tengda Cai
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham - Ningbo China, Ningbo, China
| | - Liesel M. FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Bennet J. McComish
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Ebony Liu
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Georgia Kaidonis
- Department of Ophthalmology, Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia
| | - Bronwyn Ridge
- Department of Ophthalmology, Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia
| | - Alex W. Hewitt
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Brendan Vote
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Nitin Verma
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia
| | - Colin N. A. Palmer
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Kathryn P. Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Weihua Meng
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, United Kingdom
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham - Ningbo China, Ningbo, China
| |
Collapse
|
16
|
Li H, Yin G, Zhang Y, Wang Z, Lv F, Li R, Qin J, Ye X. Effect of fat distribution on left ventricular structure and function in different sexes: a Mendelian randomization study. Front Endocrinol (Lausanne) 2025; 16:1355968. [PMID: 40070582 PMCID: PMC11893391 DOI: 10.3389/fendo.2025.1355968] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Background Although there is an interaction between sex, body fat distribution, and cardiac structure and function, these relationships have not been fully elucidated yet. This study aims to reveal the causal relationship between genetic determinants of fat distribution pattern and function of the left ventricular structure in different sexes. Methods Genetic variants for waist circumference, hip circumference, waist-to-hip ratio (WHR), and body mass index (BMI) were selected from genome-wide association studies conducted in European samples. The dataset for left ventricular (LV) parameters was obtained from over 35,000 European samples in the UK Biobank Cardiovascular Magnetic Resonance sub-study. Two-sample Mendelian randomization (MR) analysis was employed to explore causal relationships. Results After adjusting for BMI, WHR shows a positive causal relationship with LV hypertrophy and a significant negative causal relationship with LV volume and diastolic function. In further subgroup analysis, we only found similar results in WHR among the female population (FWHR), while in the male population, there was no significant causal relationship between MWHR and LV hypertrophy and diastolic function. Additionally, in our MR analysis, no causal relationship was found between WHR and LVEF. Conclusions This study indicates that the fat distribution pattern has unique effects on the structure and function of the LV, and these effects vary by sex. This study provides evidence for a causal relationship between fat distribution and LV structure and function across both sexes.
Collapse
Affiliation(s)
- Hang Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guangjiao Yin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Yanfang Zhang
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ziwei Wang
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fang Lv
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juanjuan Qin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, Hubei, China
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, Hubei, China
| |
Collapse
|
17
|
Ghosh S, Bouchard C. Considerations on efforts needed to improve our understanding of the genetics of obesity. Int J Obes (Lond) 2025; 49:206-210. [PMID: 38849463 PMCID: PMC11805711 DOI: 10.1038/s41366-024-01528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Affiliation(s)
- Sujoy Ghosh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Claude Bouchard
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
18
|
Hu M, Li X, Wu J, Li B, Xia J, Yang Y, Yin C. Phenome-Wide Investigation of the Causal Associations Between Pre-Pregnancy Obesity Traits and Gestational Diabetes: A Two-Sample Mendelian Randomization Analyses. Reprod Sci 2025; 32:395-403. [PMID: 38789873 DOI: 10.1007/s43032-024-01577-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: 02/08/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024]
Abstract
Pre-pregnancy obesity was associated with gestational diabetes in observational studies, but whether this relationship is causal remains to be determined. To evaluate whether pre-pregnancy obesity traits causally affect gestational diabetes risk, a two-sample Mendelian randomization (MR) analysis was performed utilizing summary-level statistics from published genome-wide association studies (GWAS). Obesity-related traits included body mass index (BMI), overweight, obesity, obesity class 1, obesity class 2, obesity class 3, childhood obesity, waist circumference (WC), hip circumference (HC), waist-to-hip ratio (WHR), percent liver fat, visceral adipose tissue volume, abdominal subcutaneous adipose tissue volume. Effect estimates were evaluated using the inverse-variance weighting method. Weighted median, MR-Egger, simple mode, and weighted mode were performed as sensitivity analyses. Genetically predicted pre-pregnancy BMI [odds ratio (OR) = 1.68; 95% confidence interval (CI): 1.45-1.95; P = 9.13 × 10-12], overweight (OR = 1.49; 95% CI: 1.21-1.85; P = 2.06 × 10-4), obesity (OR = 1.25; 95% CI: 1.18-1.33; P = 8.01 × 10-13), obesity class 1 (OR = 1.31; 95% CI: 1.17-1.46; P = 1.49 × 10-6), obesity class 2 (OR = 1.26; 95% CI: 1.16-1.37; P = 5.23 × 10-8), childhood obesity (OR = 1.33; 95% CI: 1.23-1.44; P = 4.06 × 10-12), and WHR (OR = 2.35; 95% CI: 1.44-3.83; P = 5.89 × 10-4) were associated with increased risk of gestational diabetes. No significant association was observed with obesity class 3, WC, HC, percent liver fat, visceral adipose tissue volume, or abdominal subcutaneous adipose tissue volume. Similar results were observed in sensitivity analyses. Therefore, genetically predicted pre-pregnancy obesity traits may increase the risk of gestational diabetes. Weight control before pregnancy may be beneficial to prevent gestational diabetes.
Collapse
Affiliation(s)
- Mengjin Hu
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiaosong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Jiangong Wu
- Fenyang Center for Disease Control and Prevention, Fenyang, 032200, Shanxi Province, China
| | - Boyu Li
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jinggang Xia
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China.
| | - Chunlin Yin
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
19
|
Chen S, Zhang Q, Zhang X, Xie P, Guo H, Lu F, Zhou C, Dong F. Casual correlation between overweight, obesity, and severe COVID-19 infection with respiratory failure: A two-sample Mendelian randomization. Medicine (Baltimore) 2025; 104:e41006. [PMID: 40184103 PMCID: PMC11709160 DOI: 10.1097/md.0000000000041006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 04/05/2025] Open
Abstract
This study aimed to detect the causal association of overweight and obesity on severe COVID-19 infection with respiratory failure through a two-sample Mendelian randomization (MR) method based on the genome-wide association studies datasets. All genome-wide association studies summary data of exposures and outcome used in this study were obtained from the IEU database derived from Europeans. The study mainly used the inverse variance weighted method to test causal relationship. Simultaneously, MR-PRESSO and MR-EGGER were used to detect the pleiotropy, and sensitivity analysis was performed using leave-one-out analysis. In the inverse variance weighted analyses, we found no causal association between obesity (e.g., OR = 1.15, 95% CIs = 0.96-1.37, P = .13 for obesity-ebi-a-GCST90000255), obesity subtypes (e.g., OR = 1.93, 95% CIs = 0.90-4.14, P = .10 for obesity and other hyperalimentation) as well as overweight (OR = 0.90, 95% CIs = 0.64-1.27, P = .54) and severe COVID-19 infection with respiratory failure. The findings showed no causal association between obesity or overweight and severe COVID-19 infection with respiratory failure. Further validation is needed regarding whether obesity or overweight is a risk factor for it.
Collapse
Affiliation(s)
- Shiqiang Chen
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Qiang Zhang
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Xiaobing Zhang
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Peiyao Xie
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Hua Guo
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Fengling Lu
- Department of Emergency and Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Chaoyang Zhou
- Department of Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| | - Fubo Dong
- Department of Critical Care Medicine, Yuhuan People’s Hospital, Taizhou, China
| |
Collapse
|
20
|
Quan L, Liu J, Wang Y, Yang F, Yang Z, Ju J, Shuai Y, Wei T, Yue J, Wang X, Meng J, Yuan P. Exploring risk factors for endocrine-related immune-related adverse events: Insights from meta-analysis and Mendelian randomization. Hum Vaccin Immunother 2024; 20:2410557. [PMID: 39377304 PMCID: PMC11469449 DOI: 10.1080/21645515.2024.2410557] [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: 06/24/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
This study utilized meta-analysis and Mendelian randomization (MR) to identify risk factors for endocrine-related immune-related adverse events (EirAEs) and to ascertain whether EirAEs confer better prognosis of immunotherapy. The meta-analysis identified several risk factors for EirAEs, including elevated baseline TSH (OR = 1.30, 95% CI 1.10-1.53), positive TgAb (OR = 14.23, p < .001), positive TPOAb (OR = 3.75, p < .001), prior thyroid-related medical history (OR = 4.19), increased BMI (OR = 1.11), combination immune checkpoint inhibitors (ICIs) therapy with targeted treatment (OR = 2.71, 95% CI 2.11-3.47), and dual ICI therapy (OR = 3.26, 95% CI 2.22-4.79). MR analysis further supported causalities between extreme BMI, hypothyroidism, and irAEs from a genetic perspective. In addition, cancer patients who experienced EirAEs exhibited significantly prolonged PFS (HR = 0.84, 95% CI 0.73-0.97) and OS (HR = 0.59, 95% CI 0.45-0.76) compared to those without. These findings provide valuable insights for clinical decision-making among healthcare professionals and offer direction for future research in this field.
Collapse
Affiliation(s)
- Liuliu Quan
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinsong Liu
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fan Yang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zixuan Yang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Ju
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - You Shuai
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tong Wei
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Yue
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Wang
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Meng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Peng Yuan
- Department of VIP Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Uttam V, Vohra V, Chhotaray S, Santhosh A, Diwakar V, Patel V, Gahlyan RK. Exome-wide comparative analyses revealed differentiating genomic regions for performance traits in Indian native buffaloes. Anim Biotechnol 2024; 35:2277376. [PMID: 37934017 DOI: 10.1080/10495398.2023.2277376] [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: 11/08/2023]
Abstract
In India, 20 breeds of buffalo have been identified and registered, yet limited studies have been conducted to explore the performance potential of these breeds, especially in the Indian native breeds. This study is a maiden attempt to delineate the important variants and unique genes through exome sequencing for milk yield, milk composition, fertility, and adaptation traits in Indian local breeds of buffalo. In the present study, whole exome sequencing was performed on Chhattisgarhi (n = 3), Chilika (n = 4), Gojri (n = 3), and Murrah (n = 4) buffalo breeds and after stringent quality control, 4333, 6829, 4130, and 4854 InDels were revealed, respectively. Exome-wide FST along 100-kb sliding windows detected 27, 98, 38, and 35 outlier windows in Chhattisgarhi, Chilika, Gojri, and Murrah, respectively. The comparative exome analysis of InDels and subsequent gene ontology revealed unique breed specific genes for milk yield (CAMSAP3), milk composition (CLCN1, NUDT3), fertility (PTGER3) and adaptation (KCNA3, TH) traits. Study provides insight into mechanism of how these breeds have evolved under natural selection, the impact of these events on their respective genomes, and their importance in maintaining purity of these breeds for the traits under study. Additionally, this result will underwrite to the genetic acquaintance of these breeds for breeding application, and in understanding of evolution of these Indian local breeds.
Collapse
Affiliation(s)
- Vishakha Uttam
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Vikas Vohra
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Supriya Chhotaray
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Ameya Santhosh
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Vikas Diwakar
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Vaibhav Patel
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Rajesh Kumar Gahlyan
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
22
|
Zhou Z, Xu J, Zhao Y, Niu Y. Relationships of dietary habits with prostate cancer risk: results from Mendelian randomization analyses and the National Health and Nutrition Examination Survey. Food Funct 2024; 15:10823-10837. [PMID: 39404821 DOI: 10.1039/d4fo03859b] [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: 10/29/2024]
Abstract
Background: Prior investigations identified correlations between dietary habits and the risk of prostate cancer (PCa); however, the causative dynamics are unclear. Methods: Utilizing the Mendelian randomization (MR) framework, we investigated the causal links between dietary habits, daily nutrient intakes, and risk of PCa (79 148 cases and 61 106 controls). Exposure and outcome data were obtained from the UK Biobank and the Prostate Cancer Association Group to Investigate Cancer-Associated Alterations in the Genome (PRACTICAL) consortium, respectively. Univariable and multivariable MR analyses were employed. Sensitivity analyses were performed to detect outliers, evaluate heterogeneity, and discern potential pleiotropic effects. Utilizing data from the National Health and Nutrition Examination Survey (NHANES) database (2009-2010), we selected 1294 and 1778 men aged ≥40 years from a pool of 10 537 participants, ensuring no missing information. Regression analyses examined the associations between leafy/lettuce salad intake, daily nutrient intake, and the odds of PCa. Results: Univariable MR (UVMR) analysis reveals that the intake of pork and salad/raw vegetable correlated with an elevated PCa risk. Subsequent to confounder adjustment via multivariable MR (MVMR) analysis, a causal link was established between salad/raw vegetable intake and an increased risk of PCa (odds ratio [OR]: 1.658, 95% confidence interval [95% CI]: 1.037-2.644, P = 0.046). The analysis based on NHANES datasets demonstrated a link between leafy/lettuce salad intake and heightened odds of PCa (OR: 1.025, 95% CI: 1.003-1.049, P = 0.038). Increased daily intakes of β-carotene (original OR: 1.00006, 95% CI: 1.00001-1.00011, P = 0.024) and vitamin B1 (OR: 1.474, 95% CI: 1.104-1.967, P = 0.014) were associated with a higher likelihood of PCa. Conclusions: These MR analyses substantiate the causal nexus between salad/raw vegetable intake and PCa risk. Similarly, leafy/lettuce salad intake and the odds of PCa were significantly correlated in the cross-sectional observational study. Moreover, higher daily intakes of β-carotene and vitamin B1 were linked to an increased likelihood of PCa. These findings provide practical dietary recommendations for PCa prevention and enhance early identification and diagnosis.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jin Xu
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Yang Zhao
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| | - Yuanjie Niu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| |
Collapse
|
23
|
Chermside-Scabbo CJ, Shuster JT, Erdmann-Gilmore P, Tycksen E, Zhang Q, Townsend RR, Silva MJ. A proteomics approach to study mouse long bones: examining baseline differences and mechanical loading-induced bone formation in young-adult and old mice. Aging (Albany NY) 2024; 16:12726-12768. [PMID: 39400554 PMCID: PMC11501390 DOI: 10.18632/aging.206131] [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: 06/20/2023] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
With aging, bone mass declines and the anabolic effects of skeletal loading diminish. While much research has focused on gene transcription, how bone ages and loses its mechanoresponsiveness at the protein level remains unclear. We developed a novel proteomics approach and performed a paired mass spectrometry and RNA-seq analysis on tibias from young-adult (5-month) and old (22-month) mice. We report the first correlation estimate between the bone proteome and transcriptome (Spearman ρ = 0.40), which is in line with other tissues but indicates that a relatively low amount of variation in protein levels is explained by the variation in transcript levels. Of 71 shared targets that differed with age, eight were associated with bone mineral density in previous GWAS, including understudied targets Asrgl1 and Timp2. We used complementary RNA in situ hybridization to confirm that Asrgl1 and Timp2 had reduced expression in osteoblasts/osteocytes in old bones. We also found evidence for reduced TGF-beta signaling with aging, in particular Tgfb2. Next, we defined proteomic changes following mechanical loading. At the protein level, bone differed more with age than with loading, and aged bone had fewer loading-induced changes. Overall, our findings underscore the need for complementary protein-level assays in skeletal biology research.
Collapse
Affiliation(s)
- Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Petra Erdmann-Gilmore
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric Tycksen
- Department of Genetics, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Qiang Zhang
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
24
|
Rees TA, Buttle BJ, Tasma Z, Yang SH, Harris PWR, Walker CS. Tirzepatide, GIP(1-42) and GIP(1-30) display unique signaling profiles at two common GIP receptor variants, E354 and Q354. Front Pharmacol 2024; 15:1463313. [PMID: 39464637 PMCID: PMC11502443 DOI: 10.3389/fphar.2024.1463313] [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: 07/11/2024] [Accepted: 10/02/2024] [Indexed: 10/29/2024] Open
Abstract
Type 2 diabetes (T2D) and obesity are prevalent metabolic disorders affecting millions of individuals worldwide. A new effective therapeutic drug called tirzepatide for the treatment of obesity and T2D is a dual agonist of the GIP receptor and GLP-1 receptor. Tirzepatide is clinically more effective than GLP-1 receptor agonists but the reasons why are not well understood. Tirzepatide reportedly stimulates the GIP receptor more potently than the GLP-1 receptor. However, tirzepatide signaling has not been thoroughly investigated at the E354 (wildtype) or Q354 (E354Q) GIP receptor variants. The E354Q variant is associated increased risk of T2D and lower body mass index. To better understand GIP receptor signaling we characterized the activity of endogenous agonists and tirzepatide at both GIP receptor variants. Using Cos7 cells we examined wildtype and E354Q GIP receptor signaling, analyzing cAMP and IP1 accumulation as well as AKT, ERK1/2 and CREB phosphorylation. GIP(1-42) and GIP(1-30)NH2 displayed equipotent effects on these pathways excluding CREB phosphorylation where GIP(1-30)NH2 was more potent than GIP(1-42) at the E354Q GIP receptor. Tirzepatide favored cAMP signaling at both variants. These findings indicate that tirzepatide is a biased agonist towards Gαs signaling and suggests it equally activates the wildtype and E354Q GIP receptor variants. We also observed differences between the pharmacology of the GIP receptor variants with endogenous peptides, which may help to explain differences in phenotype. These findings contribute to a comprehensive understanding of GIP receptor signaling, and will aid development of therapies combating T2D and obesity.
Collapse
Affiliation(s)
- Tayla A. Rees
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
- Headache Group, Wolfson Sensory Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Benjamin J. Buttle
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Zoe Tasma
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Sung-Hyun Yang
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Paul W. R. Harris
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Christopher S. Walker
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
Ziyatdinov A, Mbatchou J, Marcketta A, Backman J, Gaynor S, Zou Y, Joseph T, Geraghty B, Herman J, Watanabe K, Ghosh A, Kosmicki J, Locke A, Thornton T, Kang HM, Ferreira M, Baras A, Abecasis G, Marchini J. Joint testing of rare variant burden scores using non-negative least squares. Am J Hum Genet 2024; 111:2139-2149. [PMID: 39366334 PMCID: PMC11480795 DOI: 10.1016/j.ajhg.2024.08.021] [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: 03/18/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 10/06/2024] Open
Abstract
Gene-based burden tests are a popular and powerful approach for analysis of exome-wide association studies. These approaches combine sets of variants within a gene into a single burden score that is then tested for association. Typically, a range of burden scores are calculated and tested across a range of annotation classes and frequency bins. Correlation between these tests can complicate the multiple testing correction and hamper interpretation of the results. We introduce a method called the sparse burden association test (SBAT) that tests the joint set of burden scores under the assumption that causal burden scores act in the same effect direction. The method simultaneously assesses the significance of the model fit and selects the set of burden scores that best explain the association at the same time. Using simulated data, we show that the method is well calibrated and highlight scenarios where the test outperforms existing gene-based tests. We apply the method to 73 quantitative traits from the UK Biobank, showing that SBAT is a valuable additional gene-based test when combined with other existing approaches. This test is implemented in the REGENIE software.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuxin Zou
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | | | - Adam Locke
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | |
Collapse
|
26
|
Gupta MK, Gouda G, Vadde R. Relation Between Obesity and Type 2 Diabetes: Evolutionary Insights, Perspectives and Controversies. Curr Obes Rep 2024; 13:475-495. [PMID: 38850502 DOI: 10.1007/s13679-024-00572-1] [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] [Accepted: 05/13/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE OF REVIEW Since the mid-twentieth century, obesity and its related comorbidities, notably insulin resistance (IR) and type 2 diabetes (T2D), have surged. Nevertheless, their underlying mechanisms remain elusive. Evolutionary medicine (EM) sheds light on these issues by examining how evolutionary processes shape traits and diseases, offering insights for medical practice. This review summarizes the pathogenesis and genetics of obesity-related IR and T2D. Subsequently, delving into their evolutionary connections. Addressing limitations and proposing future research directions aims to enhance our understanding of these conditions, paving the way for improved treatments and prevention strategies. RECENT FINDINGS Several evolutionary hypotheses have been proposed to unmask the origin of obesity-related IR and T2D, e.g., the "thrifty genotype" hypothesis suggests that certain "thrifty genes" that helped hunter-gatherer populations efficiently store energy as fat during feast-famine cycles are now maladaptive in our modern obesogenic environment. The "drifty genotype" theory suggests that if thrifty genes were advantageous, they would have spread widely, but proposes genetic drift instead. The "behavioral switch" and "carnivore connection" hypotheses propose insulin resistance as an adaptation for a brain-dependent, low-carbohydrate lifestyle. The thrifty phenotype theory suggests various metabolic outcomes shaped by genes and environment during development. However, the majority of these hypotheses lack experimental validation. Understanding why ancestral advantages now predispose us to diseases may aid in drug development and prevention of disease. EM helps us to understand the evolutionary relation between obesity-related IR and T2D. But still gaps and contradictions persist. Further interdisciplinary research is required to elucidate complete mechanisms.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa, 516005, Andhra Pradesh, India.
| | - Gayatri Gouda
- ICAR-National Rice Research Institute, Cuttack, 753 006, Odisha, India
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa, 516005, Andhra Pradesh, India
| |
Collapse
|
27
|
Nishito Y, Fujishiro H, Nagamatsu S, Kambe T. Reduced Mn uptake of pleiotropic ZIP8 SNP is caused by its loss of Mn-responsive accumulation on the cell-surface. Biosci Biotechnol Biochem 2024; 88:1019-1026. [PMID: 38821503 DOI: 10.1093/bbb/zbae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Zrt/Irt-like protein 8 (ZIP8), which is a Zn transporter, plays a pivotal role as a Mn transporter. Recent studies have shown that a ZIP8 SNP (rs13107325 C→T, A391T) is associated with multiple diseases, likely by causing systemic Mn deficiency. However, the underlying molecular mechanisms remain unclear. We attempted to address this issue in cell-based experiments using Madin-Darby canine kidney cells stably expressing ZIP8 WT or the A391T SNP mutant under the control of the Tet-regulatable promoter. We showed that the A391T mutant lost the property of Mn-responsive accumulation on the cell surface, which was observed in WT ZIP8. We also showed that the loss of Mn-responsive accumulation of A391T mutant was associated with its reduced Mn uptake, compared with WT ZIP8, in the Mn uptake assay using the radioisotope 54Mn. Our results potentially explain how the ZIP8 A391T substitution is associated with disease pathogenesis caused by Mn deficiency.
Collapse
Affiliation(s)
- Yukina Nishito
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Shino Nagamatsu
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
28
|
Zhang J, Jiang H, Fu G, Wu Z, Yao Y, Sun J. Relationship between serum vitamin C and serum uric acid in people with different BMIs: results from the NHANES 2017-2018 and Mendelian randomization study. Front Nutr 2024; 11:1429123. [PMID: 39246399 PMCID: PMC11380155 DOI: 10.3389/fnut.2024.1429123] [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: 05/07/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
Objective To examine the association of overweight/obesity and serum vitamin C (serum VC) with serum uric acid (SUA) and to assess causality using Mendelian randomization (MR). Methods 4,772 participants from the National Health and Nutrition Examination Survey (NHANES), 2017-2018 were included in this study. Multivariate linear regression, variance inflation factor and quantile regression were used to analyze the relationships between overweight/obesity and serum VC and SUA levels. Secondly, Mendelian randomization (MR) was utilized to mitigate bias and prevent reverse causality in the observational study. Genetic variants associated with obesity (N = 13,848), vitamin C levels (N = 64,979) and serum uric acid levels (N = 343,836) were sourced from the most extensive genome-wide association studies (GWAS). The primary analytical method employed was inverse variance weighted (IVW). Results Based on the observational study, BMI was positively associated with SUA (β = 0.06, 95% CI: 0.05 to 0.07, p < 0.001) and serum VC was negatively associated with SUA (β = -0.14, 95% CI: -0.23 to -0.04, p = 0.005). In individuals with overweight/obesity (BMI > =25), the negative effects of serum VC on SUA enhanced with increasing serum VC. High serum VC level (Q4 level, above 1.19 mg/dL) reduced SUA (β = -0.30, 95% CI: -0.47 to -0.14, p < 0.001) in individuals with overweight/obesity compared to low serum VC level (Q1 level, below 0.54 mg/dL). IVW-MR analysis revealed a significant association between SUA levels and genetically elevated levels of VC (β = -0.03, 95% CI: -0.06 to -0.00, p = 0.029) and obesity (β = 0.06, 95% CI: 0.04 to 0.07, p < 0.001). Conclusion Cross-sectional observational analysis revealed that BMI exhibited a positive correlation with SUA levels and that serum VC was negatively correlated with SUA levels; moreover, moderate serum VC can reduce SUA, especially in individuals with overweight/obesity. There was evidence indicating a causal effect of VC and obesity on SUA. It highlights the importance of VC in the management of SUA levels, particularly in overweight/obese individuals. The findings might be helpful for the management of high SUA levels.
Collapse
Affiliation(s)
- Jiajie Zhang
- Department of Urology, National Children's Medical Center & Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hejun Jiang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanghui Fu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zou Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yukai Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Sun
- Department of Urology, National Children's Medical Center & Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne) 2024; 15:1431292. [PMID: 39114288 PMCID: PMC11304055 DOI: 10.3389/fendo.2024.1431292] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are two incretins that bind to their respective receptors and activate the downstream signaling in various tissues and organs. Both GIP and GLP-1 play roles in regulating food intake by stimulating neurons in the brain's satiety center. They also stimulate insulin secretion in pancreatic β-cells, but their effects on glucagon production in pancreatic α-cells differ, with GIP having a glucagonotropic effect during hypoglycemia and GLP-1 exhibiting glucagonostatic effect during hyperglycemia. Additionally, GIP directly stimulates lipogenesis, while GLP-1 indirectly promotes lipolysis, collectively maintaining healthy adipocytes, reducing ectopic fat distribution, and increasing the production and secretion of adiponectin from adipocytes. Together, these two incretins contribute to metabolic homeostasis, preventing both hyperglycemia and hypoglycemia, mitigating dyslipidemia, and reducing the risk of cardiovascular diseases in individuals with type 2 diabetes and obesity. Several GLP-1 and dual GIP/GLP-1 receptor agonists have been developed to harness these pharmacological effects in the treatment of type 2 diabetes, with some demonstrating robust effectiveness in weight management and prevention of cardiovascular diseases. Elucidating the underlying cellular and molecular mechanisms could potentially usher in the development of new generations of incretin mimetics with enhanced efficacy and fewer adverse effects. The treatment guidelines are evolving based on clinical trial outcomes, shaping the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Qiyuan Keith Liu
- MedStar Medical Group, MedStar Montgomery Medical Center, Olney, MD, United States
| |
Collapse
|
30
|
Shi R, Xiang S, Jia T, Robbins TW, Kang J, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Orfanos DP, Paus T, Poustka L, Hohmann S, Millenet S, Fröhner JH, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, Lin X, Sahakian BJ, Feng J. Investigating grey matter volumetric trajectories through the lifespan at the individual level. Nat Commun 2024; 15:5954. [PMID: 39009591 PMCID: PMC11251262 DOI: 10.1038/s41467-024-50305-0] [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: 06/01/2023] [Accepted: 07/04/2024] [Indexed: 07/17/2024] Open
Abstract
Adolescents exhibit remarkable heterogeneity in the structural architecture of brain development. However, due to limited large-scale longitudinal neuroimaging studies, existing research has largely focused on population averages, and the neurobiological basis underlying individual heterogeneity remains poorly understood. Here we identify, using the IMAGEN adolescent cohort followed up over 9 years (14-23 y), three groups of adolescents characterized by distinct developmental patterns of whole-brain gray matter volume (GMV). Group 1 show continuously decreasing GMV associated with higher neurocognitive performances than the other two groups during adolescence. Group 2 exhibit a slower rate of GMV decrease and lower neurocognitive performances compared with Group 1, which was associated with epigenetic differences and greater environmental burden. Group 3 show increasing GMV and lower baseline neurocognitive performances due to a genetic variation. Using the UK Biobank, we show these differences may be attenuated in mid-to-late adulthood. Our study reveals clusters of adolescent neurodevelopment based on GMV and the potential long-term impact.
Collapse
Grants
- U24 DA041147 NIDA NIH HHS
- U54 EB020403 NIBIB NIH HHS
- R56 AG058854 NIA NIH HHS
- MR/N000390/1 Medical Research Council
- MR/S020306/1 Medical Research Council
- R01 DA049238 NIDA NIH HHS
- MR/R00465X/1 Medical Research Council
- R01 MH085772 NIMH NIH HHS
- National Key R&D Program of China (No.2023YFE0199700 [to X.L.])
- the Medical Research Foundation and Medical Research Council (grants MR/R00465X/1 and MR/S020306/1 [to S.D.]), the National Institutes of Health (NIH) funded ENIGMA (grants 5U54EB020403-05 and 1R56AG058854-01 [to S.D.])
- NSFC grant 82150710554 and environMENTAL grant. Further support was provided by grants from: - the ANR (ANR-12-SAMA-0004, AAPG2019 - GeBra [to J.-L.M.]), the Eranet Neuron (AF12-NEUR0008-01 - WM2NA; and ANR-18-NEUR00002-01 - ADORe [to J.-L.M.]), the Fondation de France (00081242 [to J.-L.M.]), the Fondation pour la Recherche Médicale (DPA20140629802 [to J.-L.M.]), the Mission Interministérielle de Lutte-contre-les-Drogues-et-les-Conduites-Addictives (MILDECA [to J.-L.M.]), Paris Sud University IDEX 2012 [to J.-L.M.]
- the Assistance-Publique-Hôpitaux-de-Paris and INSERM (interface grant [to M.-L.P.M.]), the Fondation de l’Avenir (grant AP-RM-17-013 [to M.-L.P.M.])
- the Fédération pour la Recherche sur le Cerveau; the National Institutes of Health, Science Foundation Ireland (16/ERCD/3797 [to R.W.])
- the European Union-funded FP6 Integrated Project IMAGEN (Reinforcement-related behaviour in normal brain function and psychopathology) (LSHM-CT- 2007-037286 [to G.S.]), the Horizon 2020 funded ERC Advanced Grant ‘STRATIFY’ (Brain network based stratification of reinforcement-related disorders) (695313 [to G.S.]), Human Brain Project (HBP SGA 2, 785907, and HBP SGA 3, 945539 [to G.S.]), the Medical Research Council Grant 'c-VEDA’ (Consortium on Vulnerability to Externalizing Disorders and Addictions) (MR/N000390/1 [to G.S.]), the National Institute of Health (NIH) (R01DA049238 [to G.S.], A decentralized macro and micro gene-by-environment interaction analysis of substance use behavior and its brain biomarkers), the National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, the Bundesministeriumfür Bildung und Forschung (BMBF grants 01GS08152; 01EV0711 [to G.S.]; Forschungsnetz AERIAL 01EE1406A, 01EE1406B; Forschungsnetz IMAC-Mind 01GL1745B [to G.S.]), the Deutsche Forschungsgemeinschaft (DFG grants SM 80/7-2, SFB 940, TRR 265, NE 1383/14-1 [to G.S.])
- National Key R&D Program of China (No.2019YFA0709502 [to J.F.], No.2018YFC1312904 [to J.F.]),No.2019YFA0709502 [to J.F.], No.2018YFC1312904 [to J.F.]), Shanghai Municipal Science and Technology Major Project (No.2018SHZDZX01 [to J.F.], ZJ Lab [to J.F.], and Shanghai Center for Brain Science and Brain-Inspired Technology [to J.F.]), the 111 Project (No.B18015 [to J.F.])
Collapse
Affiliation(s)
- Runye Shi
- School of Data Science, Fudan University, Shanghai, China
| | - Shitong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, China
- School of Psychology, University of Southampton, Southampton, UK
| | - Trevor W Robbins
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Jujiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 "Trajectoires développementales en psychiatrie", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 "Trajectoires développementales en psychiatrie", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
- Department of Child and Adolescent Psychiatry, AP-HP, Sorbonne Université, Pitié-Salpêtrière Hospital, Paris, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 "Trajectoires développementales en psychiatrie", Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein Kiel University, Kiel, Germany
| | | | - Tomáš Paus
- Department of Psychiatry, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC, Canada
- Departments of Psychiatry and Psychology, University of Toronto, Toronto, ON, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, von-Siebold-Str. 5, Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
| | - Sabina Millenet
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
| | - Juliane H Fröhner
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Science and Technology for Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, China
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Xiaolei Lin
- School of Data Science, Fudan University, Shanghai, China.
- Huashan Institute of Medicine, Huashan Hospital affiliated to Fudan University, Shanghai, China.
| | - Barbara J Sahakian
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
| | - Jianfeng Feng
- School of Data Science, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- Department of Computer Science, University of Warwick, Coventry, UK.
| |
Collapse
|
31
|
Wang J, Liu F, Mo J, Gong D, Zheng F, Su J, Ding S, Yang W, Guo P. Exploring the causal relationship between body mass index and keratoconus: a Mendelian randomization study. Front Med (Lausanne) 2024; 11:1402108. [PMID: 39050542 PMCID: PMC11266172 DOI: 10.3389/fmed.2024.1402108] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Background Despite reports suggesting a link between obesity and keratoconus, the causal relationship is not fully understood. Methods We used genome-wide association study (GWAS) data from public databases for a two-sample Mendelian randomization analysis to investigate the causal link between body mass index (BMI) and keratoconus. The primary method was inverse variance weighted (IVW), complemented by different analytical techniques and sensitivity analyses to ensure result robustness. A meta-analysis was also performed to bolster the findings' reliability. Results Our study identified a significant causal relationship between BMI and keratoconus. Out of 20 Mendelian randomization (MR) analyses conducted, 9 showed heterogeneity or pleiotropy. Among the 11 analyses that met all three MR assumptions, 4 demonstrated a significant causal difference between BMI and keratoconus, while the remaining 7 showed a positive trend but were not statistically significant. Meta-analysis confirmed a significant causal relationship between BMI and keratoconus. Conclusion There is a significant causal relationship between BMI and keratoconus, suggesting that obesity may be a risk factor for keratoconus.
Collapse
Affiliation(s)
- Jiaoman Wang
- The 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Fangyuan Liu
- The 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Jianhao Mo
- The 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Di Gong
- Shenzhen Eye Institute, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| | - Fang Zheng
- Department of Ophthalmology, Jinzhou Medical University, Jinzhou, China
| | - Jingjing Su
- Shenzhen Eye Institute, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| | - Sicheng Ding
- The 2nd Clinical Medical College of Jinan University, Shenzhen, China
| | - Weihua Yang
- Shenzhen Eye Institute, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| | - Ping Guo
- Shenzhen Eye Institute, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| |
Collapse
|
32
|
AlBaloul AH, Griffin J, Kopytek A, Elliott P, Frost G. Evidence of gene-nutrient interaction association with waist circumference, cross-sectional analysis. BMC Public Health 2024; 24:1842. [PMID: 38987751 PMCID: PMC11234640 DOI: 10.1186/s12889-024-19127-z] [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: 06/15/2023] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND AND AIMS Waist circumference (WC) is a significant indicator of body adiposity and is associated with increased mortality and morbidity of cardiovascular diseases. Although, single nutrient intake and candidate genes were previously associated with WC. Little is known about WC association with overall diet quality, genetic risk score and gene-nutrient interaction. This study aims to investigate the influence of overall diet quality and multiple WC-associated single nucleotide polymorphisms on WC. In addition to investigating gene-nutrient interaction association with WC. METHODS This study explored cross-sectional data from two large sample-size studies, to provide reproducible results. As a representation of the UK population, the Airwave Health Monitoring Study (n = 6,502) and the UK-Biobank Cohort Study (n = 171,129) were explored for factors associated with WC. Diet quality was evaluated based on the Mellen Index for Dietary Approaches to Stop Hypertension (Mellen-DASH). The genetic risk score for WC (GRS-Waist) was calculated by screening the population genotype for WC-associated single nucleotide polymorphisms. Multivariate linear regression models were built to explore WC association with diet quality and genetic risk score. Gene-nutrient interaction was explored by introducing the interaction term (GRS-Waist X Mellen-DASH score) to multivariate linear regression analysis. RESULTS The prevalence of high WC (Female > 80 cm, Male > 94 cm) was 46.5% and 51.7% in both populations. Diet quality and genetic risk score of WC were significantly associated with WC. There was no evidence of interaction between GRS-Waist, DASH diet scores and nutrient intake on WC. CONCLUSION This study's findings provided reproducible results on waist circumference association with diet and genetics and tested the possibility of gene-nutrient interaction. These reproducible results are successful in building the foundation for using diet and genetics for early identification of those at risk of having high WC and WC-associated diseases. In addition, evidence on gene-diet interactions on WC is limited and lacks replication, therefore our findings may guide future research in investigating this interaction and investigating its application in precision nutrition.
Collapse
Affiliation(s)
- Anwar H AlBaloul
- Department of Community Medicine and Behavioural Sciences, Faculty of Medicine, Kuwait University, Safat, Kuwait
- Section of Nutrition, Faculty of Medicine, Imperial College London, London, UK
| | - Jennifer Griffin
- Section of Nutrition, Faculty of Medicine, Imperial College London, London, UK
| | - Alexandra Kopytek
- Section of Nutrition, Faculty of Medicine, Imperial College London, London, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Gary Frost
- Section of Nutrition, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
33
|
Huang X, Liang W, Yang R, Jin L, Zhao K, Chen J, Shang X, Zhou Y, Wang X, Yu H. Variations in the LINGO2 and GLIS3 Genes and Gene-Environment Interactions Increase Gestational Diabetes Mellitus Risk in Chinese Women. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:11596-11605. [PMID: 38888423 DOI: 10.1021/acs.est.4c03221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Gestational diabetes mellitus (GDM) has been found to be a common complication in pregnant women, known to escalate the risk of negative obstetric outcomes. In our study, we genotyped 1,566 Chinese pregnant women for two single nucleotide polymorphisms (SNPs) in the LINGO2 gene and one SNP in the GLIS3 gene, utilizing targeted next-generation sequencing. The impact of two interacting genes, and the interaction of genes with the environment─including exposure to particulate matter (PM2.5), ozone (O3), and variations in prepregnancy body mass index (BMI)─on the incidence of GDM were analyzed using logistic regression. Our findings identify the variants LINGO2 rs10968576 (P = 0.022, OR = 1.224) and rs1412239 (P = 0.018, OR = 1.231), as well as GLIS3 rs10814916 (P = 0.028, OR = 1.172), as risk mutations significantly linked to increased susceptibility to GDM. Further analysis underscores the crucial role of gene-gene and gene-environment interactions in the development of GDM among Chinese women (P < 0.05). Particularly, the individuals carrying the rs10968576 G-rs1412239 G-rs10814916 C haplotype exhibit increased susceptibility to GDM during the prepregnancy period when interacting with PM2.5, O3, and BMI (P = 8.004 × 10-7, OR = 1.206; P = 6.3264 × 10-11, OR = 1.280; P = 9.928 × 10-7, OR = 1.334, respectively). In conclusion, our research emphasizes the importance of the interaction between specific gene variations─LINGO2 and GLIS3─and environmental factors in influencing GDM risk. Notably, we found significant associations between these gene variations and GDM risk across various environmental exposure periods.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Weiwei Liang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Runqiu Yang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Lei Jin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing 100091, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Juan Chen
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xuejun Shang
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210023, China
| | - Yuanzhong Zhou
- School of Public Health, Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi 563000, China
| | - Xin Wang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
34
|
Wang F, Puentes E, Behrman J, Cunha F. You are What Your Parents Expect: Height and Local Reference Points. JOURNAL OF ECONOMETRICS 2024; 243:105269. [PMID: 39328300 PMCID: PMC11424033 DOI: 10.1016/j.jeconom.2021.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Recent estimates are that about 150 million children under five years of age are stunted, with substantial negative consequences for their schooling, cognitive skills, health, and economic productivity. Therefore, understanding what determines such growth retardation is significant for designing public policies that aim to address this issue. We build a model for nutritional choices and health with reference-dependent preferences. Parents care about the health of their children relative to some reference population. In our empirical model, we use height as the health outcome that parents target. Reference height is an equilibrium object determined by earlier cohorts' parents' nutritional choices in the same village. We explore the exogenous variation in reference height produced by a protein-supplementation experiment in Guatemala to estimate our model's parameters. We use our model to decompose the impact of the protein intervention on height into price and reference-point effects. We find that the changes in reference points account for 65% of the height difference between two-year-old children in experimental and control villages in the sixth annual cohort born after the initiation of the intervention.
Collapse
Affiliation(s)
- Fan Wang
- Department of Economics, University of Houston, Houston, Texas, USA
| | - Esteban Puentes
- Department of Economics, Universidad de Chile, Santiago, Chile
| | - Jere Behrman
- Departments of Economics and Sociology and Population Studies Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Flávio Cunha
- Department of Economics, Rice University, Houston, Texas, USA
| |
Collapse
|
35
|
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
|
36
|
Chen X, Yang M, Zhao W, Tu J, Liu Q, Yuan X. Mendelian randomization unraveled: gender-specific insights into obesity-related phenotypes and colorectal cancer susceptibility. Front Endocrinol (Lausanne) 2024; 15:1322253. [PMID: 38904048 PMCID: PMC11187001 DOI: 10.3389/fendo.2024.1322253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Objective Evidence has been increasingly pointing towards a potential link between phenotypes related to obesity and the incidence of colorectal cancer. However, confirming this as a direct causal connection remains elusive. This investigation aims to elucidate the causative links between obesity-associated phenotypes and the incidence of colorectal cancer. Methods Employing the Two Sample Mendelian Randomization (TwoSampleMR) R package, analyses were conducted using Mendelian randomization (MR) to discern potential causative links between obesity categories sourced from both the Institute for Education and University (IEU) Open GWAS Project and Zenodo, and colorectal tumors (data obtained from IEU Open GWAS and FinnGen). For primary evaluations, the study utilized the Wald ratio and the Inverse Variance Weighting (IVW) methods, while the MR-Egger approach was integrated for sensitivity assessment. Bidirectional Mendelian Randomization (Bidirectional MR), as well as Linkage Disequilibrium (LD) Score Regression with well-imputed HapMap3 single nucleotide polymorphisms (SNPs), were additionally executed. Sensitivity assessments entailed IVW, MR-Egger methodologies to assess heterogeneity and pleiotropy, along with a leave-one-out strategy. Instrumental variables were chosen judiciously based on predetermined P-value thresholds and F-statistics. Results Results from MR evaluations did not identify a clear causative link between BMI and colorectal malignancy. Conversely, both measures of obesity, the Waist-Hip Ratio (WHR) and its adjusted form for BMI (WHRadjBMI), displayed a connection to increased risk of colorectal cancer, especially prominent among female subjects. Reverse MR analyses dismissed potential reverse causality between colorectal malignancies and obesity. A significant genetic interplay was observed between WHR, WHRadjBMI, and colorectal cancer instances. Ensuing MR probes spotlighted inflammatory bowel ailment as a protective factor, while salad intake was indicated as a potential risk concerning colorectal malignancies. Sensitivity reviews, which included tests for both pleiotropy and heterogeneity, validated the robustness of the MR findings. Conclusion Findings from this research indicate that specific obesity-related parameters, notably WHR and WHRadjBMI, carry a causal relationship with an elevated colorectal cancer risk. The impact is distinctly more evident among females. Such insights might be pivotal for public health deliberations, hinting that individuals boasting a high WHR might necessitate intensified colorectal cancer screenings.
Collapse
Affiliation(s)
| | | | | | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingxu Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xianglin Yuan
- *Correspondence: Xianglin Yuan, ; Qingxu Liu, ; Jingyao Tu,
| |
Collapse
|
37
|
Sun W, Zhang X, Li N, He Y, Ji J, Zheng D. Genetic association of glycemic traits and antihyperglycemic agent target genes with the risk of lung cancer: A Mendelian randomization study. Diabetes Metab Syndr 2024; 18:103048. [PMID: 38850595 DOI: 10.1016/j.dsx.2024.103048] [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: 09/07/2023] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/10/2024]
Abstract
AIMS To evaluate the potential causal effect of glycemic traits on lung cancer and investigate the impact of antihyperglycemic agent-target genes on lung cancer risk. METHODS Genetic variants associated with glycemic traits, antihyperglycemic agent-target genes, and lung cancer were extracted from the Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC), expression quantitative trait loci (eQTLs), protein quantitative trait loci (pQTLs), and the International Lung Cancer Consortium (ILCCO), respectively. Mendelian randomization (MR) analyses were performed to examine the associations of glycemic traits and antihyperglycemic agent-target genes with lung cancer. Mediation analysis was conducted to explore whether overweight operated as a mediator between antihyperglycemic agents and lung cancer outcomes. RESULTS Genetically determined glycated hemoglobin A1c levels were associated with squamous cell lung cancer (OR = 1.78; 95 % CI, 1.08-2.92; p = 0.023). The PRKAB1 gene (the target of metformin) was associated with a lower risk of developing lung adenocarcinoma (OR = 0.85; 95 % CI, 0.76-0.96; p = 0.006). Further mediation analyses did not support overweight as a mediator between PRKAB1 activation and lung adenocarcinoma. CONCLUSION Our analyses suggest an association of genetically determined abnormal glycemic traits with squamous cell lung cancer. The potential association between PRKAB1 activation and a reduced risk of developing lung adenocarcinoma appears to be independent of the anti-obesity effects of metformin, suggesting that PRKAB1 activation may have a direct protective effect on lung adenocarcinoma development.
Collapse
Affiliation(s)
- Wen Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Xiaoyu Zhang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ning Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yan He
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| | - Jianguang Ji
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden.
| | - Deqiang Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.
| |
Collapse
|
38
|
Zomer HD, Cooke PS. Advances in Drug Treatments for Companion Animal Obesity. BIOLOGY 2024; 13:335. [PMID: 38785817 PMCID: PMC11117622 DOI: 10.3390/biology13050335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Companion animal obesity has emerged as a significant veterinary health concern globally, with escalating rates posing challenges for preventive and therapeutic interventions. Obesity not only leads to immediate health problems but also contributes to various comorbidities affecting animal well-being and longevity, with consequent emotional and financial burdens on owners. While past treatment strategies have shown limited success, recent breakthroughs in human medicine present new opportunities for addressing this complex issue in companion animals. Here, we discuss the potential of GLP-1 receptor agonists, specifically semaglutide and tirzepatide, already approved for human use, for addressing companion animal obesity. These drugs, originally developed to treat type 2 diabetes in humans and subsequently repurposed to treat obesity, have demonstrated remarkable weight loss effects in rodents, non-human primates and people. Additionally, newer drug combinations have shown even more promising results in clinical trials. Despite current cost and supply challenges, advancements in oral and/or extended-release formulations and increased production may make these drugs more accessible for veterinary use. Thus, these drugs may have utility in companion animal weight management, and future feasibility studies exploring their efficacy and safety in treating companion animal obesity are warranted.
Collapse
Affiliation(s)
| | - Paul S. Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
39
|
Zeng L, Li Y, Hong C, Wang J, Zhu H, Li Q, Cui H, Ma P, Li R, He J, Zhu H, Liu L, Xiao L. Association between fatty liver index and controlled attenuation parameters as markers of metabolic dysfunction-associated fatty liver disease and bone mineral density: observational and two-sample Mendelian randomization studies. Osteoporos Int 2024; 35:679-689. [PMID: 38221591 DOI: 10.1007/s00198-023-06996-0] [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: 05/16/2023] [Accepted: 12/04/2023] [Indexed: 01/16/2024]
Abstract
Previously observational studies did not draw a clear conclusion on the association between fatty liver diseases and bone mineral density (BMD). Our large-scale studies revealed that MAFLD and hepatic steatosis had no causal effect on BMD, while some metabolic factors were correlated with BMD. The findings have important implications for the relationship between fatty liver diseases and BMD, and may help direct the clinical management of MAFLD patients who experience osteoporosis and osteopenia. PURPOSE Liver and bone are active endocrine organs with several metabolic functions. However, the link between metabolic dysfunction-associated fatty liver disease (MAFLD) and bone mineral density (BMD) is contradictory. METHODS Using the UK Biobank and National Health and Nutrition Examination Survey (NHANES) dataset, we investigated the association between MAFLD, steatosis, and BMD in the observational analysis. We performed genome-wide association analysis to identify single-nucleotide polymorphisms associated with MAFLD. Large-scale two-sample Mendelian randomization (TSMR) analyses examined the potential causal relationship between MAFLD, hepatic steatosis, or major comorbid metabolic factors, and BMD. RESULTS After adjusting for demographic factors and body mass index, logistic regression analysis demonstrated a significant association between MAFLD and reduced heel BMD. However, this association disappeared after adjusting for additional metabolic factors. MAFLD was not associated with total body, femur neck, and lumbar BMD in the NHANES dataset. Magnetic resonance imaging-measured steatosis did not show significant associations with reduced total body, femur neck, and lumbar BMD in multivariate analysis. TSMR analyses indicated that MAFLD and hepatic steatosis were not associated with BMD. Among all MAFLD-related comorbid factors, overweight and type 2 diabetes showed a causal relationship with increased BMD, while waist circumference and hyperlipidemia had the opposite effect. CONCLUSION No causal effect of MAFLD and hepatic steatosis on BMD was observed in this study, while some metabolic factors were correlated with BMD. This has important implications for understanding the relationship between fatty liver disease and BMD, which may help direct the clinical management of MAFLD patients with osteoporosis.
Collapse
Affiliation(s)
- Lin Zeng
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chang Hong
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiaren Wang
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongbo Zhu
- Department of Medical Oncology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China
| | - Qimei Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Cui
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pengcheng Ma
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ruining Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jingzhe He
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hong Zhu
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Li Liu
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lushan Xiao
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
40
|
Li Y, Wang X, Zhang Z, Shi L, Cheng L, Zhang X. Effect of the gut microbiome, plasma metabolome, peripheral cells, and inflammatory cytokines on obesity: a bidirectional two-sample Mendelian randomization study and mediation analysis. Front Immunol 2024; 15:1348347. [PMID: 38558794 PMCID: PMC10981273 DOI: 10.3389/fimmu.2024.1348347] [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: 12/02/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Background Obesity is a metabolic and chronic inflammatory disease involving genetic and environmental factors. This study aimed to investigate the causal relationship among gut microbiota abundance, plasma metabolomics, peripheral cell (blood and immune cell) counts, inflammatory cytokines, and obesity. Methods Summary statistics of 191 gut microbiota traits (N = 18,340), 1,400 plasma metabolite traits (N = 8,299), 128 peripheral cell counts (blood cells, N = 408,112; immune cells, N = 3,757), 41 inflammatory cytokine traits (N = 8,293), and 6 obesity traits were obtained from publicly available genome-wide association studies. Two-sample Mendelian randomization (MR) analysis was applied to infer the causal links using inverse variance-weighted, maximum likelihood, MR-Egger, weighted median, weighted mode, and Wald ratio methods. Several sensitivity analyses were also utilized to ensure reliable MR results. Finally, we used mediation analysis to identify the pathway from gut microbiota to obesity mediated by plasma metabolites, peripheral cells, and inflammatory cytokines. Results MR revealed a causal effect of 44 gut microbiota taxa, 281 plasma metabolites, 27 peripheral cells, and 8 inflammatory cytokines on obesity. Among them, five shared causal gut microbiota taxa belonged to the phylum Actinobacteria, order Bifidobacteriales, family Bifidobacteriaceae, genus Lachnospiraceae UCG008, and species Eubacterium nodatum group. Furthermore, we screened 42 shared causal metabolites, 7 shared causal peripheral cells, and 1 shared causal inflammatory cytokine. Based on known causal metabolites, we observed that the metabolic pathways of D-arginine, D-ornithine, linoleic acid, and glycerophospholipid metabolism were closely related to obesity. Finally, mediation analysis revealed 20 mediation relationships, including the causal pathway from gut microbiota to obesity, mediated by 17 metabolites, 2 peripheral cells, and 1 inflammatory cytokine. Sensitivity analysis represented no heterogeneity or pleiotropy in this study. Conclusion Our findings support a causal relationship among gut microbiota, plasma metabolites, peripheral cells, inflammatory cytokines, and obesity. These biomarkers provide new insights into the mechanisms underlying obesity and contribute to its prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Ying Li
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Xin Wang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zitong Zhang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Lei Shi
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Liang Cheng
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xue Zhang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Ipas H, Gouws EB, Abell NS, Chiou PC, Devanathan SK, Hervé S, Lee S, Mercado M, Reinsborough C, Halabelian L, Arrowsmith CH, Xhemalçe B. ChemRAP uncovers specific mRNA translation regulation via RNA 5' phospho-methylation. EMBO Rep 2024; 25:1570-1588. [PMID: 38263329 PMCID: PMC10933402 DOI: 10.1038/s44319-024-00059-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/25/2024] Open
Abstract
5'-end modifications play key roles in determining RNA fates. Phospho-methylation is a noncanonical cap occurring on either 5'-PPP or 5'-P ends. We used ChemRAP, in which affinity purification of cellular proteins with chemically synthesized modified RNAs is coupled to quantitative proteomics, to identify 5'-Pme "readers". We show that 5'-Pme is directly recognized by EPRS, the central subunit of the multisynthetase complex (MSC), through its linker domain, which has previously been involved in key noncanonical EPRS and MSC functions. We further determine that the 5'-Pme writer BCDIN3D regulates the binding of EPRS to specific mRNAs, either at coding regions rich in MSC codons, or around start codons. In the case of LRPPRC (leucine-rich pentatricopeptide repeat containing), a nuclear-encoded mitochondrial protein associated with the French Canadian Leigh syndrome, BCDIN3D deficiency abolishes binding of EPRS around its mRNA start codon, increases its translation but ultimately results in LRPPRC mislocalization. Overall, our results suggest that BCDIN3D may regulate the translation of specific mRNA via RNA-5'-Pme.
Collapse
Affiliation(s)
- Hélène Ipas
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Ellen B Gouws
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Nathan S Abell
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Po-Chin Chiou
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Sravan K Devanathan
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Solène Hervé
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Sidae Lee
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Marvin Mercado
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Calder Reinsborough
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA
| | - Levon Halabelian
- Structural Genomics Consortium, and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Blerta Xhemalçe
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, 78712, Austin, TX, USA.
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
Hübel C, Abdulkadir M, Herle M, Palmos AB, Loos RJF, Breen G, Micali N, Bulik CM. Persistent thinness and anorexia nervosa differ on a genomic level. Eur J Hum Genet 2024; 32:117-124. [PMID: 37474786 PMCID: PMC10772076 DOI: 10.1038/s41431-023-01431-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
Thinness and anorexia nervosa are both characterised by persistent low weight. Individuals with anorexia nervosa concurrently report distorted perceptions of their body and engage in weight-loss behaviours, whereas individuals with thinness often wish to gain weight. Both conditions are heritable and share genomics with BMI, but are not genetically correlated with each other. Based on their pattern of genetic associations with other traits, we explored differences between thinness and anorexia nervosa on a genomic level. In Part 1, using publicly available data, we compared genetic correlations of persistent thinness/anorexia nervosa with eleven psychiatric disorders. In Part 2, we identified individuals with adolescent persistent thinness in the Avon Longitudinal Study of Parents and Children (ALSPAC) by latent class growth analysis of measured BMI from 10 to 24 years (n = 6594) and evaluated associations with psychiatric and anthropometric polygenic scores. In Part 1, in contrast to the positive genetic correlations of anorexia nervosa with various psychiatric disorders, persistent thinness showed negative genetic correlations with attention deficit hyperactivity disorder (rgAN = 0.08 vs. rgPT = -0.30), alcohol dependence (rgAN = 0.07 vs. rgPT = -0.44), major depressive disorder (rgAN = 0.27 vs. rgPT = -0.18) and post-traumatic stress disorder (rgAN = 0.26 vs. rgPT = -0.20). In Part 2, individuals with adolescent persistent thinness in the ALSPAC had lower borderline personality disorder polygenic scores (OR = 0.77; Q = 0.01). Overall, results suggest that genetic variants associated with thinness are negatively associated with psychiatric disorders and therefore thinness may be differentiable from anorexia nervosa on a genomic level.
Collapse
Affiliation(s)
- Christopher Hübel
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK.
- National Centre for Register-based Research, Aarhus Business and Social Sciences, Aarhus University, Aarhus, Denmark.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Mohamed Abdulkadir
- National Centre for Register-based Research, Aarhus Business and Social Sciences, Aarhus University, Aarhus, Denmark
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Moritz Herle
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Biostatistics & Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Alish B Palmos
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Gerome Breen
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Nadia Micali
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Mental Health Services in the Capital Region of Denmark, Eating Disorders Research Unit, Psychiatric Centre Ballerup, Ballerup, Denmark
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Ou J, Zou L, Wu Y, Zhang Q, Fang Y, Qiu M, Tian X, Ma L, Bi H, Liu C. Causal inference between rheumatoid arthritis and prostate cancer. Clin Exp Med 2023; 23:4681-4694. [PMID: 37567983 DOI: 10.1007/s10238-023-01151-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023]
Abstract
It is unclear if the association between rheumatoid arthritis (RA) and a higher risk of prostate cancer (Pca) reflects a causal relationship. We conducted a meta-analysis and used the Mendelian randomization method (MR) to evaluate the association between RA and Pca risk. A meta-analysis and subgroup analysis of the incidence of Pca in patients with RA was conducted. To determine whether genetically elevated RA levels were causally linked to Pca, two MR samples were employed. To eliminate gender-related bias, we conducted a stratified analysis of the GWAS data for RA by gender, specifically including 140,254 males. Additional MR analysis was also performed to determine potential confounding factors influencing the association between genetically susceptible RA and Pca. In total, 409,950 participants were enrolled in 20 trials to investigate the Pca risk in patients with RA. The meta-analysis suggested that RA was unrelated to the Pca risk (SIR = 1.072, 95% CI, 0.883-1.261). However, a subgroup analysis showed that low smoking rates might increase the Pca risk in patients with RA by 24%. The MR analysis showed that increased genetic susceptibility to RA was related to a high Pca risk (OR = 36.20, 95%CI = 1.24-1053.12, P = 0.037). The causality estimation of MR-Egger, Weighted mode, Simple mode, and Weighted median method were similar in direction and magnitude. Although our meta-analysis found no correlation between RA and Pca risk, MR analyses supported a causal relationship between genetic susceptibility to RA and increased prostate risk. Early attention to Pca risk in patients with RA may be important for improving prognosis and mortality in such patients. Further research is needed to determine the etiology of RA attributed to Pca and its underlying mechanisms.
Collapse
Affiliation(s)
- Junyong Ou
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Lang Zou
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Yaqian Wu
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Qiming Zhang
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Yangyi Fang
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Min Qiu
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Xiaojun Tian
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Lulin Ma
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China
| | - Hai Bi
- Department of Urology, Peking University Third Hospital, Peking University Health Science Center, 49 North Garden Road, Beijing, 100191, China.
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China.
| | - Cheng Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, China.
| |
Collapse
|
46
|
Ma Z, Chang Y, Brito LF, Li Y, Yang T, Wang Y, Yang N. Multitrait meta-analyses identify potential candidate genes for growth-related traits in Holstein heifers. J Dairy Sci 2023; 106:9055-9070. [PMID: 37641329 DOI: 10.3168/jds.2023-23462] [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: 03/07/2023] [Accepted: 06/20/2023] [Indexed: 08/31/2023]
Abstract
Understanding the underlying pleiotropic relationships among growth and body size traits is important for refining breeding strategies in dairy cattle for optimal body size and growth rate. Therefore, we performed single-trait GWAS for monthly-recorded body weight (BW), hip height, body length, and chest girth from birth to 12 mo of age in Holstein animals, followed by stepwise multiple regression of independent or lowly-linked markers from GWAS loci using conditional and joint association analyses (COJO). Subsequently, we conducted a multitrait meta-analysis to detect pleiotropic markers. Based on the single-trait GWAS, we identified 170 significant SNPs, in which 59 of them remained significant after the COJO analyses. The most significant SNP, located at BTA7:3,676,741, explained 2.93% of the total phenotypic variance for BW6 (BW at 6 mo of age). We identified 17 SNPs with potential pleiotropic effects based on the multitrait meta-analyses, which resulted in 3 additional SNPs in comparison to those detected based on the single-trait GWAS. The identified quantitative trait loci regions overlap with genes known to influence human growth-related traits. According to positional and functional analyses, we proposed HMGA2, HNF4G, MED13L, BHLHE40, FRZB, DMP1, TRIB3, and GATAD2A as important candidate genes influencing the studied traits. The combination of single-trait GWAS and meta-analyses of GWAS results improved the efficiency of detecting associated SNPs, and provided new insights into the genetic mechanisms of growth and development in Holstein cattle.
Collapse
Affiliation(s)
- Z Ma
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China; Beijing Sunlon Livestock Development Co. Ltd., 100029, Beijing, China
| | - Y Chang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Y Li
- Beijing Sunlon Livestock Development Co. Ltd., 100029, Beijing, China
| | - T Yang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China
| | - Y Wang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China.
| | - N Yang
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
47
|
Radhika A, Burgula S, Badapanda C, Hussain T, Naushad SM. Elucidation of genetic determinants of dyslipidaemia using a global screening array for the early detection of coronary artery disease. Mamm Genome 2023; 34:632-643. [PMID: 37668737 DOI: 10.1007/s00335-023-10017-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
Dyslipidemia is a major risk factor for the development of coronary artery disease (CAD). Understanding the genetic determinants of dyslipidemia can provide valuable information on the pathogenesis of CAD and aid in the development of early detection strategies. In this study, we used a Global Screening Array (GSA) to elucidate the genetic factors associated with dyslipidemia and their potential role in the prediction of CAD. We conducted a GSA-based association study in 265 subjects to identify the genetic loci associated with dyslipidemia traits using Multiple Linear Regression (MLR) and Logistic Regression (LR), Classification and Regression Tree (CART), and Manhattan plots. We identified an association between dyslipidemia and variants identified in genes such as JCAD, GLIS3, CD38, FN1, CELSR2, MTNR1B, GIPR, DYM, APOB, APOE, ADCY5. The MLR models explained 62%, 71%, and 81% of the variability in HDL, LDL, and triglycerides, respectively. The Area Under the Curve (AUC) values in the LR models of HDL, LDL, and triglycerides were 1.00, 0.94, and 0.95, respectively. CART models identified novel gene-gene interactions influencing the risk for dyslipidemia. To conclude, we have identified the association of 12 SNVs with dyslipidemia and demonstrated their clinical utility in four different models such as MLR, LR, CART, and Manhattan plots. The identified genetic variants and associated pathways shed light on the underlying biology of dyslipidemia and offer potential avenues for precision medicine strategies in the management of CAD.
Collapse
Affiliation(s)
- Ananthaneni Radhika
- Genomics Division, Yoda Lifeline Diagnostics Pvt Ltd, 6-3-862/A, Lal Bungalow Add On, Ameerpet, Hyderabad, 500016, India
- Department of Microbiology, Osmania University, Taranaka, Hyderabad, 500007, India
| | - Sandeepta Burgula
- Department of Microbiology, Osmania University, Taranaka, Hyderabad, 500007, India.
| | - Chandan Badapanda
- Genomics Division, Yoda Lifeline Diagnostics Pvt Ltd, 6-3-862/A, Lal Bungalow Add On, Ameerpet, Hyderabad, 500016, India
| | - Tajamul Hussain
- Research Chair for Biomedical Applications of Nanomaterials, Biochemistry Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
- Center of Excellence in Biotechnology Research, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Shaik Mohammad Naushad
- Genomics Division, Yoda Lifeline Diagnostics Pvt Ltd, 6-3-862/A, Lal Bungalow Add On, Ameerpet, Hyderabad, 500016, India.
| |
Collapse
|
48
|
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
|
49
|
Yammine L, Picatoste B, Abdullah N, Leahey RA, Johnson EF, Gómez-Banoy N, Rosselot C, Wen J, Hossain T, Goncalves MD, Lo JC, Garcia-Ocaña A, McGraw TE. Spatiotemporal regulation of GIPR signaling impacts glucose homeostasis as revealed in studies of a common GIPR variant. Mol Metab 2023; 78:101831. [PMID: 37925022 PMCID: PMC10665708 DOI: 10.1016/j.molmet.2023.101831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
OBJECTIVE Glucose-dependent insulinotropic polypeptide (GIP) has a role in controlling postprandial metabolic tone. In humans, a GIP receptor (GIPR) variant (Q354, rs1800437) is associated with a lower body mass index (BMI) and increased risk for Type 2 Diabetes. To better understand the impacts of GIPR-Q354 on metabolism, it is necessary to study it in an isogeneic background to the predominant GIPR isoform, E354. To accomplish this objective, we used CRISPR-CAS9 editing to generate mouse models of GIPR-Q354 and GIPR-E354. Here we characterize the metabolic effects of GIPR-Q354 variant in a mouse model (GIPR-Q350). METHODS We generated the GIPR-Q350 mice for in vivo studies of metabolic impact of the variant. We isolated pancreatic islets from GIPR-Q350 mice to study insulin secretion ex vivo. We used a β-cell cell line to understand the impact of the GIPR-Q354 variant on the receptor traffic. RESULTS We found that female GIPR-Q350 mice are leaner than littermate controls, and male GIPR-Q350 mice are resistant to diet-induced obesity, in line with the association of the variant with reduced BMI in humans. GIPR-Q350 mice of both sexes are more glucose tolerant and exhibit an increased sensitivity to GIP. Postprandial GIP levels are reduced in GIPR-Q350 mice, revealing feedback regulation that balances the increased sensitivity of GIP target tissues to secretion of GIP from intestinal endocrine cells. The increased GIP sensitivity is recapitulated ex vivo during glucose stimulated insulin secretion assays in islets. Generation of cAMP in islets downstream of GIPR activation is not affected by the Q354 substitution. However, post-activation traffic of GIPR-Q354 variant in β-cells is altered, characterized by enhanced intracellular dwell time and increased localization to the Trans-Golgi Network (TGN). CONCLUSIONS Our data link altered intracellular traffic of the GIPR-Q354 variant with GIP control of metabolism. We propose that this change in spatiotemporal signaling underlies the physiologic effects of GIPR-Q350/4 and GIPR-E350/4 in mice and humans. These findings contribute to a more complete understanding of the impact of GIPR-Q354 variant on glucose homeostasis that could perhaps be leveraged to enhance pharmacologic targeting of GIPR for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Lucie Yammine
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Belén Picatoste
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Nazish Abdullah
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Rosemary A Leahey
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Emma F Johnson
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Nicolás Gómez-Banoy
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Carolina Rosselot
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Tahmina Hossain
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | | | - James C Lo
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA; Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
50
|
Jain PR, Burch M, Martinez M, Mir P, Fichna JP, Zekanowski C, Rizzo R, Tümer Z, Barta C, Yannaki E, Stamatoyannopoulos J, Drineas P, Paschou P. Can polygenic risk scores help explain disease prevalence differences around the world? A worldwide investigation. BMC Genom Data 2023; 24:70. [PMID: 37986041 PMCID: PMC10662565 DOI: 10.1186/s12863-023-01168-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023] Open
Abstract
Complex disorders are caused by a combination of genetic, environmental and lifestyle factors, and their prevalence can vary greatly across different populations. The extent to which genetic risk, as identified by Genome Wide Association Study (GWAS), correlates to disease prevalence in different populations has not been investigated systematically. Here, we studied 14 different complex disorders and explored whether polygenic risk scores (PRS) based on current GWAS correlate to disease prevalence within Europe and around the world. A clear variation in GWAS-based genetic risk was observed based on ancestry and we identified populations that have a higher genetic liability for developing certain disorders. We found that for four out of the 14 studied disorders, PRS significantly correlates to disease prevalence within Europe. We also found significant correlations between worldwide disease prevalence and PRS for eight of the studied disorders with Multiple Sclerosis genetic risk having the highest correlation to disease prevalence. Based on current GWAS results, the across population differences in genetic risk for certain disorders can potentially be used to understand differences in disease prevalence and identify populations with the highest genetic liability. The study highlights both the limitations of PRS based on current GWAS but also the fact that in some cases, PRS may already have high predictive power. This could be due to the genetic architecture of specific disorders or increased GWAS power in some cases.
Collapse
Affiliation(s)
- Pritesh R Jain
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Myson Burch
- Department of Computer Sciences, Purdue University, West Lafayette, IN, USA
| | - Melanie Martinez
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Instituto de Biomedicina de Sevilla (IBiS). Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jakub P Fichna
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Cezary Zekanowski
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Renata Rizzo
- Child and Adolescent Neurology and Psychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Zeynep Tümer
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Evangelia Yannaki
- Hematology Department- Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - John Stamatoyannopoulos
- Altius Institute for Biomedical Sciences, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Oncology, University of Washington, Seattle, WA, USA
| | - Petros Drineas
- Department of Computer Sciences, Purdue University, West Lafayette, IN, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|