1
|
Wang W, Zhang Z, Shi Q, Wang F, Cao Y, Fan B, Yang Y. Causal effect of body mass index on herpes zoster and postherpetic neuralgia: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42775. [PMID: 40489821 DOI: 10.1097/md.0000000000042775] [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] [Indexed: 06/11/2025] Open
Abstract
Although observational studies have reported the relationship between body mass index (BMI), herpes zoster (HZ), and postherpetic neuralgia (PHN), the impacts of BMI on the incidence of HZ and PHN are still controversial. Our study aimed to explore the causal effect of BMI on HZ and PHN by a 2-sample Mendelian randomization (MR) approach. Genome-wide association studies data on BMI, HZ, and PHN were derived from publicly available genetic summary datasets. A total of 28 phenotypic single-nucleotide polymorphisms were selected as instrumental variables for BMI. Inverse-variance weighted (IVW) method was conducted as the primary MR analysis method to explore the causal effect of BMI on HZ and PHN. Several sensitivity analyses were performed to test the robustness of the MR results. Our study found no strong evidence for an effect of BMI on HZ incidence (IVW: OR = 1.018, 95% CI = 0.964-1.075, P = .524). However, it demonstrated that increased BMI was related to a higher risk of PHN (IVW: OR = 1.234, 95% CI = 1.002-1.520, P = .048). Besides, no significant heterogeneity or horizontal pleiotropy was observed in our study, and sensitivity analysis was consistent with the results. There is no causal effect of BMI on HZ risk, but it may be causally associated with a risk of PHN.
Collapse
Affiliation(s)
- Wen Wang
- Department of Pain Management, China-Japan Friendship Hospital, Beijing, China
| | - Ze Zhang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Qing Shi
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Fuquan Wang
- Department of Pain Management, China-Japan Friendship Hospital, Beijing, China
| | - Yanting Cao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Bifa Fan
- Department of Pain Management, China-Japan Friendship Hospital, Beijing, China
| | - Yang Yang
- Department of Pain Management, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
2
|
Bineid MM, Ventura EF, Samidoust A, Radha V, Anjana RM, Sudha V, Walton GE, Mohan V, Vimaleswaran KS. A Systematic Review of the Effect of Gene-Lifestyle Interactions on Metabolic-Disease-Related Traits in South Asian Populations. Nutr Rev 2025; 83:1061-1082. [PMID: 39283705 PMCID: PMC12066952 DOI: 10.1093/nutrit/nuae115] [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] [Indexed: 05/13/2025] Open
Abstract
CONTEXT Recent data from the South Asian subregion have raised concern about the dramatic increase in the prevalence of metabolic diseases, which are influenced by genetic and lifestyle factors. OBJECTIVE The aim of this systematic review was to summarize the contemporary evidence for the effect of gene-lifestyle interactions on metabolic outcomes in this population. DATA SOURCES PubMed, Web of Science, and SCOPUS databases were searched up until March 2023 for observational and intervention studies investigating the interaction between genetic variants and lifestyle factors such as diet and physical activity on obesity and type 2 diabetes traits. DATA EXTRACTION Of the 14 783 publications extracted, 15 were deemed eligible for inclusion in this study. Data extraction was carried out independently by 3 investigators. The quality of the included studies was assessed using the Appraisal Tool for Cross-Sectional Studies (AXIS), the Risk Of Bias In Non-randomized Studies-of Interventions (ROBINS-I), and the methodological quality score for nutrigenetics studies. DATA ANALYSIS Using a narrative synthesis approach, the findings were presented in textual and tabular format. Together, studies from India (n = 8), Pakistan (n = 3), Sri Lanka (n = 1), and the South Asian diaspora in Singapore and Canada (n = 3) reported 543 gene-lifestyle interactions, of which 132 (∼24%) were statistically significant. These results were related to the effects of the interaction of genetic factors with physical inactivity, poor sleep habits, smoking, and dietary intake of carbohydrates, protein, and fat on the risk of metabolic disease in this population. CONCLUSIONS The findings of this systematic review provide evidence of gene-lifestyle interactions impacting metabolic traits within the South Asian population. However, the lack of replication and correction for multiple testing and the small sample size of the included studies may limit the conclusiveness of the evidence. Note, this paper is part of the Nutrition Reviews Special Collection on Precision Nutrition. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration No. CRD42023402408.
Collapse
Affiliation(s)
- Manahil M Bineid
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, United Kingdom
| | - Eduard F Ventura
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Aryan Samidoust
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, United Kingdom
| | - Venkatesan Radha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research on Diabetes, Chennai 603103, India
| | - Ranjit Mohan Anjana
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research on Diabetes, Chennai 603103, India
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India
- Department of Diabetology, Dr Mohan’s Diabetes Specialties Centre, IDF Centre of Excellence in Diabetes Care, Chennai 600086, India
| | - Vasudevan Sudha
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India
| | - Gemma E Walton
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AP, United Kingdom
| | - Viswanathan Mohan
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research on Diabetes, Chennai 603103, India
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India
- Department of Diabetology, Dr Mohan’s Diabetes Specialties Centre, IDF Centre of Excellence in Diabetes Care, Chennai 600086, India
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, United Kingdom
- The Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading RG6 6AH, United Kingdom
| |
Collapse
|
3
|
Masip G, Han HY, Meng T, Nielsen DE. Polygenic Risk and Nutrient Intake Interactions on Obesity Outcomes: A Systematic Review and Meta-Analysis of Observational Studies. Obes Rev 2025:e13941. [PMID: 40375759 DOI: 10.1111/obr.13941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/26/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Diet is an important determinant of body weight and may modulate genetic susceptibility to obesity. OBJECTIVE This systematic review and meta-analysis aimed to synthesize evidence related to interactions between polygenic risk and nutrient intakes on obesity outcomes. METHODS MEDLINE, EMBASE, Web of Science, and Cochrane Library were systematically searched to identify observational studies that assessed interactions between polygenic risk and nutrient intakes on obesity-related outcomes. Random effects meta-analyses were performed for pooled polygenic risk score (PRS)-total fat intake and PRS-protein intake interaction coefficients on body mass index (BMI). RESULTS Twenty-six publications were retrieved with studies conducted among European, Asian, and African samples. Dietary fats (saturated fat, omega-3, and trans fat) and energy intake were most frequently reported to interact with PRS on obesity outcomes, but the total number of studies available was low. No significant interactions were identified in meta-analyses of PRS interactions with total fat intake and protein intake on BMI. Several studies were rated as low quality, heterogeneity was high, and although study samples were racially diverse, PRSs tended to be based on samples of European ancestry. CONCLUSION Evidence of interactions between polygenic risk and nutrient intakes on obesity outcomes is limited and inconsistent. Further research addressing limitations related to study quality and polygenic risk characterization is needed.
Collapse
Affiliation(s)
- Guiomar Masip
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
- Growth, Exercise, Nutrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón) Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Hannah Yang Han
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
| | - Tongzhu Meng
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
| | - Daiva E Nielsen
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Kuccukturk S, Yosunkaya S, Karaselek MA, Demirel S, Vural H. A case-control study on SH2B1 gene variants in obesity and obstructive sleep apnea severity: genetic risk factors in the leptin signaling pathway. Expert Rev Endocrinol Metab 2025; 20:241-248. [PMID: 40103410 DOI: 10.1080/17446651.2025.2478068] [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: 09/24/2024] [Accepted: 02/22/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a common sleep disorder, frequently observed in obese individuals, with shared mechanisms involving leptin and its receptor, which regulate appetite and energy expenditure. SH2B1 is a key enhancer of signaling in the leptin receptor pathway. This study aimed to investigate the association between SH2B1 variants and OSA. RESEARCH DESIGN AND METHODS This case-control study included 160 male patients with OSA and 76 healthy controls, stratified into subgroups based on BMI (≤25 kg/m2 and ≥ 30 kg/m2). Polysomnography and anthropometric measurements were performed, and genotyping of three SH2B1 variants (rs7498665, rs4788102, and rs7359397) was conducted. RESULTS Mutant genotypes of all three SH2B1 variants were significantly associated with higher BMI. Additionally, normal genotypes of rs4788102 and rs7359397 were associated with higher apnea-hypopnea index (AHI) values, indicating a potential risk for OSA. CONCLUSION The findings suggest that while SH2B1 variants are strongly associated with BMI, specific normal genotypes may independently contribute to OSA risk by increasing AHI values.
Collapse
Affiliation(s)
- Serkan Kuccukturk
- Medicine Faculty, Department of Medical Biology, Karamanoğlu Mehmetbey University, Karaman, Turkey
| | - Sebnem Yosunkaya
- Medicine Faculty, Department of Chest Diseases, Necmettin Erbakan University, Konya, Turkey
| | - Mehmet Ali Karaselek
- Medicine Faculty, Department of Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Sennur Demirel
- Medicine Faculty, Department of Medical Biology, Necmettin Erbakan University, Konya, Turkey
| | - Hasibe Vural
- Medicine Faculty, Department of Medical Biology, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
5
|
Zhang Y, Feng G, Zhang W, Liu X. Natural Compounds Exert Anti-Obesity Effects by Regulating Cytokines. Phytother Res 2025. [PMID: 40312999 DOI: 10.1002/ptr.8508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/13/2025] [Accepted: 03/29/2025] [Indexed: 05/03/2025]
Abstract
Obesity, along with its associated health risks such as hypertension, hyperlipidemia, Type 2 diabetes, stroke, metabolic syndrome, asthma, and cancer, constitutes a significant global health burden, contributing substantially to morbidity and mortality. Cytokines, a group of secreted signaling proteins, are crucial in initiating, maintaining, and resolving immune and metabolic responses. Although cytokines have unique advantages in regulating immune and metabolic functions, their therapeutic application for obesity remains limited in clinical practice. Natural compounds, known for their structural diversity and low toxicity, have become a valuable resource for drug development. Many natural compounds have shown anti-obesity effects. This review comprehensively examines the mechanisms underlying obesity, with a specific focus on the roles of cytokines, such as inflammatory cytokines, adipokines, and growth factors. Additionally, it highlights the regulatory interactions between gut microbiota and cytokines in obesity. The review critically analyzes current anti-obesity pharmacological interventions and summarizes advanced methodologies for identifying potential natural compounds. Finally, it identifies promising natural compounds that modulate cytokine activity to prevent or treat obesity and assesses their potential as complementary or alternative therapies.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Guize Feng
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
6
|
Wean J, Kowalsky AH, Laker R, Will S, Drucker DJ, Rhodes CJ, Seeley RJ. Specific loss of GIPR signaling in GABAergic neurons enhances GLP-1R agonist-induced body weight loss. Mol Metab 2025; 95:102074. [PMID: 39612941 PMCID: PMC11946504 DOI: 10.1016/j.molmet.2024.102074] [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: 08/25/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
OBJECTIVES Dual incretin agonists are among the most effective pharmaceutical treatments for obesity and type 2 diabetes to date. Such therapeutics can target two receptors, such as the glucagon-like peptide-1 (GLP-1) receptor and the glucose-dependent insulinotropic polypeptide (GIP) receptor in the case of tirzepatide, to improve glycemia and reduce body weight. Regarding body weight effects, GIPR signaling is thought to involve at least two relevant mechanisms: the enhancement of food intake reduction and the attenuation of aversive effects caused by GLP-1R agonists. Although it is known that dual GLP-1R-GIPR agonism produces greater weight loss than GLP-1R agonism alone, the precise mechanism is unknown. METHODS To address this question, we used mice lacking GIPR in the whole body, GABAergic neurons, or glutamatergic neurons. These mice were given various combinations of GLP-1R and GIPR agonist drugs with subsequent food intake and conditioned taste aversion measurements. RESULTS A GIPR knockout in either the whole body or selectively in inhibitory GABAergic neurons protects against diet-induced obesity, whereas a knockout in excitatory glutamatergic neurons had a negligible effect. Furthermore, we found that GIPR in GABAergic neurons is essential for the enhanced weight loss efficacy of dual incretin agonism, yet, surprisingly, its removal enhances the effect of GLP-1R agonism alone. Finally, GIPR knockout in GABAergic neurons prevents the anti-aversive effects of GIPR agonism. CONCLUSIONS Our findings are consistent with GIPR research at large in that both enhancement and removal of GIPR signaling are metabolically beneficial. Notably, however, our findings suggest that future obesity therapies designed to modulate GIPR signaling, whether by agonism or antagonism, would be best targeted towards GABAergic neurons.
Collapse
Affiliation(s)
- Jordan Wean
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Rhianna Laker
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sarah Will
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Daniel J Drucker
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Canada
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Giuranna J, Zheng Y, Brandt M, Jall S, Mukherjee A, Shankhwar S, Renner S, Kurapati NK, May C, Peters T, Herpertz-Dahlmann B, Seitz J, de Zwaan M, Herzog W, Ehrlich S, Zipfel S, Giel K, Egberts K, Burghardt R, Föcker M, Marcus K, Keyvani K, Müller TD, Schmitz F, Rajcsanyi LS, Hinney A. Genetic and functional analyses of CTBP2 in anorexia nervosa and body weight regulation. Mol Psychiatry 2025; 30:1836-1846. [PMID: 39511451 PMCID: PMC12014503 DOI: 10.1038/s41380-024-02791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024]
Abstract
The C-terminal binding protein 2 (CTBP2) gene (translational isoforms: CTBP2-L/S, RIBEYE) had been identified by a cross-trait analysis of genome-wide association studies for anorexia nervosa (AN) and body mass index (BMI). Here, we did a mutation analysis in CTBP2 by performing polymerase chain reactions with subsequent Sanger-sequencing to identify variants relevant for AN and body weight regulation and ensued functional studies. Analysis of the coding regions of CTBP2 in 462 female patients with AN (acute or recovered), 490 children and adolescents with severe obesity, 445 healthy-lean adult individuals and 168 healthy adult individuals with normal body weight detected 24 variants located in the specific exon of RIBEYE. In the initial analysis, three of these were rare non-synonymous variants (NSVs) detected heterozygously in patients with AN (p.Arg72Trp - rs146900874; p.Val289Met -rs375685611 and p.Gly362Arg - rs202010294). Four NSVs and one heterozygous frameshift variant were exclusively detected in children and adolescents with severe obesity (p.Pro53Ser - rs150867595; p.Gln175ArgfsTer45 - rs141864737; p.Leu310Val - rs769811964; p.Pro397Ala - rs76134089 and p.Pro402Ser - rs113477585). Ribeye mRNA was detected in mouse hypothalamus. No effect of fasting or overfeeding on murine hypothalamic Ribeye expression was determined. Yet, increased Ribeye expression was detected in hypothalami of leptin-treated Lepob/ob mice. This increase was not related to reduced food intake and leptin-induced weight loss. We detected rare and frequent variants in the RIBEYE specific exon in both patients with AN and in children and adolescents with severe obesity. Our data suggest RIBEYE as a relevant gene for weight regulation.
Collapse
Affiliation(s)
- Johanna Giuranna
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Yiran Zheng
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | | | - Sigrid Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Amrita Mukherjee
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, Homburg, Germany
| | - Soni Shankhwar
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, Homburg, Germany
| | - Simone Renner
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilian University Munich (LMU), Munich, Germany
| | - Nirup Kumar Kurapati
- Institute of Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Caroline May
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Triinu Peters
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany
- Institute of Sex and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Martina de Zwaan
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Wolfgang Herzog
- Department of Internal Medicine II, General Internal and Psychosomatic Medicine, University of Heidelberg, Heidelberg, Germany
| | - Stefan Ehrlich
- Eating Disorders Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Germany
| | - Stephan Zipfel
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
- Center of Excellence in Eating Disorders KOMET, Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Katrin Giel
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
- Center of Excellence in Eating Disorders KOMET, Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Karin Egberts
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Roland Burghardt
- Child and Adolescent Psychiatry Clinic, Oberberg Fachklinik Fasanenkiez Berlin, Berlin, Germany
| | - Manuel Föcker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Münster, Münster, Germany
- LWL-University Hospital Hamm for Child and Adolescent Psychiatry, Ruhr-University Bochum, Hamm, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Kathy Keyvani
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Institute of Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Walther-Straub-Insitute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich (LMU), Munich, Germany
| | - Frank Schmitz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, Homburg, Germany
| | - Luisa Sophie Rajcsanyi
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany.
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany.
- Institute of Sex and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany.
| | - Anke Hinney
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany
- Institute of Sex and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
8
|
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
|
9
|
Deng L, Gòdia M, Derks MFL, Harlizius B, Farhangi S, Tang Z, Groenen MAM, Madsen O. Comprehensive expression genome-wide association study of long non-coding RNAs in four porcine tissues. Genomics 2025; 117:111026. [PMID: 40049421 DOI: 10.1016/j.ygeno.2025.111026] [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: 08/29/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs), a type of non-coding RNA molecules, are known to play critical regulatory roles in various biological processes. However, the functions of the majority of lncRNAs remain largely unknown, and little is understood about the regulation of lncRNA expression. In this study, high-throughput DNA genotyping and RNA sequencing were applied to investigate genomic regions associated with lncRNA expression, commonly referred to as lncRNA expression quantitative trait loci (eQTLs). We analyzed the liver, lung, spleen, and muscle transcriptomes of 100 three-way crossbred sows to identify lncRNA transcripts, explore genomic regions that might influence lncRNA expression, and identify potential regulators interacting with these regions. RESULT We identified 6380 lncRNA transcripts and 3733 lncRNA genes. Correlation tests between the expression of lncRNAs and protein-coding genes were performed. Subsequently, functional enrichment analyses were carried out on protein-coding genes highly correlated with lncRNAs. Our correlation results of these protein-coding genes uncovered terms that are related to tissue specific functions. Additionally, heatmaps of lncRNAs and protein-coding genes at different correlation levels revealed several distinct clusters. An expression genome-wide association study (eGWAS) was conducted using 535,896 genotypes and 1829, 1944, 2089, and 2074 expressed lncRNA genes for liver, spleen, lung, and muscle, respectively. This analysis identified 520,562 significant associations and 6654, 4525, 4842, and 7125 eQTLs for the respective tissues. Only a small portion of these eQTLs were classified as cis-eQTLs. Fifteen regions with the highest eQTL density were selected as eGWAS hotspots and potential mechanisms of lncRNA regulation in these hotspots were explored. However, we did not identify any interactions between the transcription factors or miRNAs in the hotspots and the lncRNAs, nor did we observe a significant enrichment of regulatory elements in these hotspots. While we could not pinpoint the key factors regulating lncRNA expression, our results suggest that the regulation of lncRNAs involves more complex mechanisms. CONCLUSION Our findings provide insights into several features and potential functions of lncRNAs in various tissues. However, the mechanisms by which lncRNA eQTLs regulate lncRNA expression remain unclear. Further research is needed to explore the regulation of lncRNA expression and the mechanisms underlying lncRNA interactions with small molecules and regulatory proteins.
Collapse
Affiliation(s)
- Liyan Deng
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands; Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Marta Gòdia
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands
| | - Martijn F L Derks
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands; Topigs Norsvin Research Center, 's-Hertogenbosch, the Netherlands
| | | | - Samin Farhangi
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands
| | - Zhonglin Tang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Martien A M Groenen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands.
| |
Collapse
|
10
|
Zhang M, Ward J, Strawbridge RJ, Anderson JJ, Celis-Morales C, Pell JP, Ho FK, Lyall DM. Genetic predisposition to adiposity, and type 2 diabetes: the role of lifestyle and phenotypic adiposity. Eur J Endocrinol 2025; 192:549-557. [PMID: 40315335 PMCID: PMC12056655 DOI: 10.1093/ejendo/lvaf084] [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: 01/21/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
AIMS Genetic predisposition to adiposity is associated with type 2 diabetes (T2D), even in the absence of phenotypic adiposity (obesity and central obesity). We aimed to quantify the overall contribution of obesity and modifiable lifestyle factors to the association between genetic predisposition to adiposity and the development of T2D. METHODS This prospective cohort study involved 220 703 White British participants from the UK Biobank. It examined the associations between genetic predisposition to adiposity [body mass index polygenic risk (BMI-PRS) and waist-hip ratio polygenic risk (WHR-PRS)] and incident T2D, as well as interactions and mediation via lifestyle factors (diet quality, physical activity levels, total energy intake, sleep duration, and smoking and alcohol intake) and phenotypic adiposity. RESULTS People with high phenotypic adiposity and high adiposity PRS values (>1 SD above the mean) had the highest risk of incident T2D (versus non-obese/central obese and non-high PRS). This was the case for BMI-PRS [hazard ratio (HR) = 3.72] and WHR-PRS (HR = 4.17). Lifestyle factors explained 30.5% of the BMI-PRS/T2D association (2.0% mediation; 28.5% effect modification), and lifestyle and obesity together explained 92.1% (78.8% mediation; 13.3% effect modification). Lifestyle factors explained 20.4% of the WHR-PRS/T2D association (3.4% mediation; 17.0% effect modification), and lifestyle and central obesity together explained 72.8% (41.1% mediation; 31.7% effect modification). CONCLUSIONS Whilst phenotypic adiposity explains a large proportion of the association between BMI-PRS/WHR-PRS and T2D, modifiable lifestyle factors also make contributions. Promoting healthy lifestyles among people prone to adiposity is important in reducing the global burden of T2D.
Collapse
Affiliation(s)
- Mengrong Zhang
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
- Department of Medicine Solna, Karolinska Institute, Stockholm 17177, Sweden
| | - Jana J Anderson
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Carlos Celis-Morales
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, United Kingdom
- Human Performance Lab, Education, Physical Activity, and Health Research Unit, Universidad Católica del Maule, Talca 115 3745, Chile
- Centro de Investigación en Medicina de Altura (CEIMA), Universidad Arturo Prat, Iquique 1100012, Chile
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Frederick K Ho
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Donald M Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| |
Collapse
|
11
|
Verde L, Galasso M, Coletta DK, Savastano S, Mandarino LJ, Colao A, Barrea L, Muscogiuri G. The Interplay of UCP3 and PCSK1 Variants in Severe Obesity. Curr Obes Rep 2025; 14:38. [PMID: 40281302 PMCID: PMC12031958 DOI: 10.1007/s13679-025-00631-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Obesity is a heterogeneous and multifactorial disease with a strong genetic component. While polygenic obesity accounts for most common cases, rare monogenic variants contribute, particularly in severe, early-onset obesity. Among the lesser-studied candidates are UCP3 and PCSK1, genes involved in key metabolic pathways. RECENT FINDINGS: The UCP3 p.Val192Ile (c.574G > A) and PCSK1 p.Asn221Asp (c.661 A > G) variants have been independently associated with metabolic pathways, including fatty acid oxidation and hormone processing, as well as a modestly increased risk of obesity. Clinical and genetic characterization of two patients with severe early-onset obesity revealed the co-occurrence of these variants, which were associated with metabolic disturbances such as insulin resistance. PURPOSE OF THE REVIEW: This narrative review examined the functional and clinical significance of UCP3 and PCSK1 variants in severe obesity, presenting two case reports to illustrate their potential impact. Our findings support a potential model in which rare variants in distinct metabolic genes may interact synergistically to exacerbate disease severity. Further studies are needed to elucidate their combined functional effects and contributions to obesity pathogenesis.
Collapse
Affiliation(s)
- Ludovica Verde
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
| | - Martina Galasso
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Dawn K Coletta
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
- Department of Physiology, University of Arizona, Tucson, AZ, USA
- Center for Disparities in Diabetes, Obesity and Metabolism, University of Arizona, Tucson, AZ, USA
| | - Silvia Savastano
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Lawrence J Mandarino
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
- Center for Disparities in Diabetes, Obesity and Metabolism, University of Arizona, Tucson, AZ, USA
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy
| | - Luigi Barrea
- Dipartimento di Psicologia e Scienze della Salute, , Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, Isola F2, 80143, Naples, Italy
| | - Giovanna Muscogiuri
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA.
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy.
| |
Collapse
|
12
|
Kim H, Chen J, Prescott B, Walker ME, Grams ME, Yu B, Vasan RS, Floyd J, Sotoodehnia N, Smith NL, Arking DE, Coresh J, Rebholz CM. Plant-Based Diets and Cardiovascular Events: A Proteomics Approach to Examine the Underlying Pathways. J Nutr 2025:S0022-3166(25)00195-6. [PMID: 40228715 DOI: 10.1016/j.tjnut.2025.04.011] [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: 12/26/2024] [Revised: 03/19/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Plant-based diets are associated with a lower risk of cardiovascular disease (CVD). Proteomics may improve our understanding of the biological pathways underlying these associations. OBJECTIVES Using large-scale proteomics, we aimed to examine if plant-based diet-related proteins, which have been previously identified, are associated with incident CVD and subtypes of CVD in the Atherosclerosis Risk in Communities (ARIC) Study and Framingham Heart Study (FHS) Offspring cohort. METHODS Discovery analyses were based on 9078 participants free of CVD at ARIC visit 3 (1993-1995). Cox proportional hazards regression was used to evaluate the associations between plant-based diet-related proteins and incident CVD, coronary artery disease, heart failure, and stroke. Replication analyses were based on 1279 participants without CVD in the FHS Offspring cohort. RESULTS In the ARIC study, over a median follow-up of 21 y, there were 3167 CVD events. At a false discovery rate <0.05, 26 of 73 plant-based diet-related proteins were significantly associated with incident CVD, after adjusting for important confounders. Eighteen, 1, and 0 proteins were associated with heart failure, stroke, and coronary artery disease, respectively. Three and 2 additional proteins were associated with CVD and heart failure risk in the FHS Offspring cohort at the nominal threshold (P < 0.05). In the ARIC Study and FHS Offspring cohort, soluble advanced glycosylation end product-specific receptor was inversely associated with incident CVD whereas thrombospondin-2 (THBS2) and N-terminal pro-BNP was positively associated with incident CVD. THBS2 was positively associated with incident heart failure, whereas neuronal growth factor regulator 1 and insulin-like growth factor-binding protein 1 was inversely associated. CONCLUSIONS These proteins highlight several pathways that could explain plant-based diets-CVD associations.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology, University of Washington, Seattle, WA, United States; Cardiovascular Health Research Unit, Department of Medicine, University of Washington School of Public Health, Seattle, WA, United States.
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Brenton Prescott
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Maura E Walker
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States; Department of Health Sciences, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA, United States
| | - Morgan E Grams
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Bing Yu
- Department of Epidemiology, University of Texas Health Sciences Center at Houston School of Public Health, Houston, TC, United States
| | - Ramachandran S Vasan
- Department of Quantitative and Qualitative Health Sciences, University of Texas School of Public Health in San Antonio, San Antonio, TX, United States
| | - James Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington School of Public Health, Seattle, WA, United States; Division of Cardiology, University of Washington, Seattle, WA, United States
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington School of Public Health, Seattle, WA, United States; Division of Cardiology, University of Washington, Seattle, WA, United States
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA, United States; Cardiovascular Health Research Unit, Department of Medicine, University of Washington School of Public Health, Seattle, WA, United States
| | - Dan E Arking
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; Optimal Aging Institute and Division of Epidemiology, Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
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
|
14
|
Mack KL, Landino NP, Tertyshnaia M, Longo TC, Vera SA, Crew LA, McDonald K, Phifer-Rixey M. Gene-by-environment Interactions and Adaptive Body Size Variation in Mice From the Americas. Mol Biol Evol 2025; 42:msaf078. [PMID: 40172935 PMCID: PMC12015161 DOI: 10.1093/molbev/msaf078] [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: 10/04/2024] [Revised: 02/14/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025] Open
Abstract
The relationship between genotype and phenotype is often mediated by the environment. Moreover, gene-by-environment (GxE) interactions can contribute to variation in phenotypes and fitness. In the last 500 yr, house mice have invaded the Americas. Despite their short residence time, there is evidence of rapid climate adaptation, including shifts in body size and aspects of metabolism with latitude. Previous selection scans have identified candidate genes for metabolic adaptation. However, environmental variation in diet as well as GxE interactions likely impact body mass variation in wild populations. Here, we investigated the role of the environment and GxE interactions in shaping adaptive phenotypic variation. Using new locally adapted inbred strains from North and South America, we evaluated response to a high-fat diet, finding that sex, strain, diet, and the interaction between strain and diet contributed significantly to variation in body size. We also found that the transcriptional response to diet is largely strain-specific, indicating that GxE interactions affecting gene expression are pervasive. Next, we used crosses between strains from contrasting climates to characterize gene expression regulatory divergence on a standard diet and on a high-fat diet. We found that gene regulatory divergence is often condition-specific, particularly for trans-acting changes. Finally, we found evidence for lineage-specific selection on cis-regulatory variation involved in diverse processes, including lipid metabolism. Overlap with scans for selection identified candidate genes for environmental adaptation with diet-specific effects. Together, our results underscore the importance of environmental variation and GxE interactions in shaping adaptive variation in complex traits.
Collapse
Affiliation(s)
- Katya L Mack
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, USA
| | - Nico P Landino
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | | | - Tiffany C Longo
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Sebastian A Vera
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Lilia A Crew
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Kristi McDonald
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Megan Phifer-Rixey
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
- Department of Biology, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
15
|
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
|
16
|
James-Okoro PP, Lewis JE, Gribble FM, Reimann F. The role of GIPR in food intake control. Front Endocrinol (Lausanne) 2025; 16:1532076. [PMID: 40166681 PMCID: PMC11955450 DOI: 10.3389/fendo.2025.1532076] [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: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is one of two incretin hormones playing key roles in the control of food intake, nutrient assimilation, insulin secretion and whole-body metabolism. Recent pharmacological advances and clinical trials show that unimolecular co-agonists that target the receptors for the incretins - GIP and glucagon-like peptide 1 (GLP-1) - offer more effective treatment strategies for obesity and type 2 diabetes mellitus (T2D) compared with GLP-1 receptor (GLP1R) agonists alone, suggesting previously underappreciated roles of GIP in regulating food intake and body weight. The mechanisms by which GIP regulates energy balance remain controversial as both agonism and antagonism of the GIP receptor (GIPR) produce weight loss and improve metabolic outcomes in preclinical models. Recent studies have shown that GIPR signalling in the central nervous system (CNS), especially in regions of the brain that regulate energy balance, is essential for its action on appetite regulation. This finding has sparked interest in understanding the mechanisms by which GIP engages brain circuits to reduce food intake and body weight. In this review, we present key knowledge around the actions of GIP on food intake regulation and the potential mechanisms by which GIPR and GIPR/GLP1R agonists may regulate energy balance.
Collapse
Affiliation(s)
| | | | - Fiona Mary Gribble
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Ayesh H, Nasser SA, Ferdinand KC, Carranza Leon BG. Sex-Specific Factors Influencing Obesity in Women: Bridging the Gap Between Science and Clinical Practice. Circ Res 2025; 136:594-605. [PMID: 40080532 DOI: 10.1161/circresaha.124.325535] [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: 10/16/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 03/15/2025]
Abstract
Obesity in women is a significant public health issue with serious implications for cardiovascular-kidney-metabolic syndrome and cardiovascular disease. This complex challenge is influenced by physiological, hormonal, socioeconomic, and cultural factors. Women face unique weight management challenges due to hormonal changes during pregnancy, perimenopause, and menopause, which affect fat distribution and increase cardiovascular-kidney-metabolic syndrome risk. Current clinical guidelines often overlook these sex-specific factors, potentially limiting the effectiveness of obesity management strategies in women. This review explores the sex-specific aspects of obesity's pathophysiology, epidemiological trends, and associated comorbidities, focusing on cardiovascular and metabolic complications. This review synthesizes literature on obesity in women, emphasizing sex-specific factors influencing its development and progression. It examines the limitations of body mass index as an obesity measure and explores alternative classification methods. Additionally it investigates the relationship between obesity and comorbidities such as diabetes, hypertension, and dyslipidemia, with a focus on postmenopausal women. Obesity in women is linked to increased risks of cardiovascular-kidney-metabolic syndrome and cardiovascular disease. Hormonal fluctuations throughout life contribute to weight gain and fat distribution patterns specific to women, increasing cardiovascular disease risk. Effective obesity management strategies in women must account for these sex-specific variations. Postmenopausal women are particularly affected by obesity-related complications. Lifestyle interventions, pharmacotherapy, and bariatric surgery have shown efficacy in weight management, though success rates vary. Addressing obesity in women requires a comprehensive approach that considers sex-specific physiological factors, life-stage challenges, and sociocultural barriers. Integrating precision medicine and emerging therapies offers potential for more personalized and effective interventions. Personalized strategies that consider women's biological and life-stage challenges can enhance obesity management and improve cardiovascular outcomes. Future research and clinical practice should focus on developing tailored strategies that address women's unique vulnerabilities to obesity and its associated health risks and on validating sex-specific interventions to improve obesity management in women.
Collapse
Affiliation(s)
- Hazem Ayesh
- Deaconess Clinic Endocrinology, Deaconess Health System, Evansville, IN (H.A.)
| | - Samar A Nasser
- Department of Clinical Research and Leadership, School of Medicine and Health Sciences, The George Washington University, Washington, DC (S.A.N.)
| | - Keith C Ferdinand
- Section of Cardiology, Tulane University School of Medicine, New Orleans, LA (K.C.F.)
| | - Barbara Gisella Carranza Leon
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN (B.G.C.L.)
| |
Collapse
|
18
|
Teixeira MR, Silva T, Felício RDFM, Bozza PT, Zembrzuski VM, de Mello Neto CB, da Fonseca ACP, Kohlrausch FB, Salum KCR. Exploring the genetic contribution in obesity: An overview of dopaminergic system genes. Behav Brain Res 2025; 480:115401. [PMID: 39689745 DOI: 10.1016/j.bbr.2024.115401] [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: 08/26/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
Obesity is a widespread global health concern that affects a significant portion of the population and is associated with reduced quality of life, morbidity, and mortality. It is considered a pandemic, with its prevalence constantly rising in Western countries. As a result, numerous studies have focused on understanding the elements that contribute to obesity. Researchers have focused on neurotransmitters in the brain to develop weight management drugs that regulate food intake. This review explores the literature on genetic influences on dopaminergic processes to determine whether genetic variation has an association with obesity in reward-responsive regions, including mesolimbic efferent and mesocortical areas. Various neurotransmitters play an essential role in regulating food intake, such as dopamine which controls through mesolimbic circuits in the brain that modulate food reward. Appetite stimulation, including primary reinforcers such as food, leads to an increase in dopamine release in the reward centers of the brain. This release is related to motivation and reinforcement, which determines the motivational weighting of the reinforcer. Changes in dopamine expression can lead to hedonic eating behaviors and contribute to the development of obesity. Genetic polymorphisms have been investigated due to their potential role in modulating the risk of obesity and eating behaviors. Therefore, it is crucial to assess the impact of genetic alterations that disrupt this pathway on the obesity phenotype.
Collapse
Affiliation(s)
- Myrela Ribeiro Teixeira
- Human Genetics Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil; Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil; Postgraduate Program in Science and Biotechnology, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil
| | - Tamara Silva
- Genetics Laboratory, Grande Rio University/AFYA, Professor José de Souza Herdy Street, 1160 - Jardim Vinte e Cinco de Agosto, Duque de Caxias, RJ 25071-202, Brazil
| | - Rafaela de Freitas Martins Felício
- Congenital Malformation Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil
| | - Patrícia Torres Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Rio de Janeiro, RJ 21040‑360, Brazil
| | - Verônica Marques Zembrzuski
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil
| | - Cicero Brasileiro de Mello Neto
- Human Genetics Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil; Postgraduate Program in Science and Biotechnology, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil
| | - Ana Carolina Proença da Fonseca
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil; Genetics Laboratory, Grande Rio University/AFYA, Professor José de Souza Herdy Street, 1160 - Jardim Vinte e Cinco de Agosto, Duque de Caxias, RJ 25071-202, Brazil; Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Rio de Janeiro, RJ 21040‑360, Brazil; Postgraduate Program in Translational Biomedicine, Grande Rio University/AFYA, Professor José de Souza Herdy Street, 1160 - Jardim Vinte e Cinco de Agosto, Duque de Caxias, RJ 25071-202, Brazil
| | - Fabiana Barzotto Kohlrausch
- Human Genetics Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Professor Marcos Waldemar de Freitas Reis Street, Niterói, RJ 24210-201, Brazil
| | - Kaio Cezar Rodrigues Salum
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), 4365 Brazil Avenue, Leônidas Deane Pavilion, Rio de Janeiro, RJ 21040-360, Brazil; Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Professor Rodolpho Paulo Rocco Street, 255, University City, Rio de Janeiro, RJ 21941-617, Brazil.
| |
Collapse
|
19
|
Halder SK, Melkani GC. The Interplay of Genetic Predisposition, Circadian Misalignment, and Metabolic Regulation in Obesity. Curr Obes Rep 2025; 14:21. [PMID: 40024983 PMCID: PMC11872776 DOI: 10.1007/s13679-025-00613-3] [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] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF REVIEW This review explores the complex interplay between genetic predispositions to obesity, circadian rhythms, metabolic regulation, and sleep. It highlights how genetic factors underlying obesity exacerbate metabolic dysfunction through circadian misalignment and examines promising interventions to mitigate these effects. RECENT FINDINGS Genome-wide association Studies (GWAS) have identified numerous Single Nucleotide Polymorphisms (SNPs) associated with obesity traits, attributing 40-75% heritability to body mass index (BMI). These findings illuminate critical links between genetic obesity, circadian clocks, and metabolic processes. SNPs in clock-related genes influence metabolic pathways, with disruptions in circadian rhythms-driven by poor sleep hygiene or erratic eating patterns-amplifying metabolic dysfunction. Circadian clocks, synchronized with the 24-h light-dark cycle, regulate key metabolic activities, including glucose metabolism, lipid storage, and energy utilization. Genetic mutations or external disruptions, such as irregular sleep or eating habits, can destabilize circadian rhythms, promoting weight gain and metabolic disorders. Circadian misalignment in individuals with genetic predispositions to obesity disrupts the release of key metabolic hormones, such as leptin and insulin, impairing hunger regulation and fat storage. Interventions like time-restricted feeding (TRF) and structured physical activity offer promising strategies to restore circadian harmony, improve metabolic health, and mitigate obesity-related risks.
Collapse
Affiliation(s)
- Sajal Kumar Halder
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Girish C Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- UAB Nathan Shock Center, Birmingham, AL, 35294, USA.
| |
Collapse
|
20
|
Mazzaferro E, Mujica E, Zhang H, Emmanouilidou A, Jenseit A, Evcimen B, Metzendorf C, Dethlefsen O, Loos RJ, Vienberg SG, Larsson A, Allalou A, den Hoed M. Functionally characterizing obesity-susceptibility genes using CRISPR/Cas9, in vivo imaging and deep learning. Sci Rep 2025; 15:5408. [PMID: 39948378 PMCID: PMC11825957 DOI: 10.1038/s41598-025-89823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 02/07/2025] [Indexed: 02/16/2025] Open
Abstract
Hundreds of loci have been robustly associated with obesity-related traits, but functional characterization of candidate genes remains a bottleneck. Aiming to systematically characterize candidate genes for a role in accumulation of lipids in adipocytes and other cardiometabolic traits, we developed a pipeline using CRISPR/Cas9, non-invasive, semi-automated fluorescence imaging and deep learning-based image analysis in live zebrafish larvae. Results from a dietary intervention show that 5 days of overfeeding is sufficient to increase the odds of lipid accumulation in adipocytes by 10 days post-fertilization (dpf, n = 275). However, subsequent experiments show that across 12 to 16 established obesity genes, 10 dpf is too early to detect an effect of CRISPR/Cas9-induced mutations on lipid accumulation in adipocytes (n = 1014), and effects on food intake at 8 dpf (n = 1127) are inconsistent with earlier results from mammals. Despite this, we observe effects of CRISPR/Cas9-induced mutations on ectopic accumulation of lipids in the vasculature (sh2b1 and sim1b) and liver (bdnf); as well as on body size (pcsk1, pomca, irs1); whole-body LDLc and/or total cholesterol content (irs2b and sh2b1); and pancreatic beta cell traits and/or glucose content (pcsk1, pomca, and sim1a). Taken together, our results illustrate that CRISPR/Cas9- and image-based experiments in zebrafish larvae can highlight direct effects of obesity genes on cardiometabolic traits, unconfounded by their - not yet apparent - effect on excess adiposity.
Collapse
Affiliation(s)
- Eugenia Mazzaferro
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Endrina Mujica
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Hanqing Zhang
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Anastasia Emmanouilidou
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Anne Jenseit
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Bade Evcimen
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Christoph Metzendorf
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden
| | - Olga Dethlefsen
- Science for Life Laboratory, National Bioinformatics Infrastructure, Stockholm University, Stockholm, Sweden
| | - Ruth Jf Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala , Sweden
| | - Amin Allalou
- Department of Information Technology, Division of Visual Information and Interaction, Uppsala University, Uppsala , Sweden
- BioImage Informatics Facility at SciLifeLab, Uppsala, Sweden
| | - Marcel den Hoed
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala , Sweden.
| |
Collapse
|
21
|
Križanac AM, Reimer C, Heise J, Liu Z, Pryce JE, Bennewitz J, Thaller G, Falker-Gieske C, Tetens J. Sequence-based GWAS in 180,000 German Holstein cattle reveals new candidate variants for milk production traits. Genet Sel Evol 2025; 57:3. [PMID: 39905301 PMCID: PMC11796172 DOI: 10.1186/s12711-025-00951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Milk production traits are complex and influenced by many genetic and environmental factors. Although extensive research has been performed for these traits, with many associations unveiled thus far, due to their crucial economic importance, complex genetic architecture, and the fact that causal variants in cattle are still scarce, there is a need for a better understanding of their genetic background. In this study, we aimed to identify new candidate loci associated with milk production traits in German Holstein cattle, the most important dairy breed in Germany and worldwide. For that purpose, 180,217 cattle were imputed to the sequence level and large-scale genome-wide association study (GWAS) followed by fine-mapping and evolutionary and functional annotation were carried out to identify and prioritize new association signals. RESULTS Using the imputed sequence data of a large cattle dataset, we identified 50,876 significant variants, confirming many known and identifying previously unreported candidate variants for milk (MY), fat (FY), and protein yield (PY). Genome-wide significant signals were fine-mapped with the Bayesian approach that determines the credible variant sets and generates the probability of causality for each signal. The variants with the highest probabilities of being causal were further classified using external information about the function and evolution, making the prioritization for subsequent validation experiments easier. The top potential causal variants determined with fine-mapping explained a large percentage of genetic variance compared to random ones; 178 variants explained 11.5%, 104 explained 7.7%, and 68 variants explained 3.9% of the variance for MY, FY, and PY, respectively, demonstrating the potential for causality. CONCLUSIONS Our findings proved the power of large samples and sequence-based GWAS in detecting new association signals. In order to fully exploit the power of GWAS, one should aim at very large samples combined with whole-genome sequence data. These can also come with both computational and time burdens, as presented in our study. Although milk production traits in cattle are comprehensively investigated, the genetic background of these traits is still not fully understood, with the potential for many new associations to be revealed, as shown. With constantly growing sample sizes, we expect more insights into the genetic architecture of milk production traits in the future.
Collapse
Affiliation(s)
- Ana-Marija Križanac
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany.
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany.
| | - Christian Reimer
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Johannes Heise
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Zengting Liu
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Jennie E Pryce
- Agriculture Victoria Research, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Jörn Bennewitz
- Institute of Animal Science, University of Hohenheim, 70599, Stuttgart, Germany
| | - Georg Thaller
- Institute of Animal Breeding and Husbandry, Christian-Albrechts-University, 24118, Kiel, Germany
| | - Clemens Falker-Gieske
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Jens Tetens
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| |
Collapse
|
22
|
Song M, Bai Y, Song F. High-fat diet and neuroinflammation: The role of mitochondria. Pharmacol Res 2025; 212:107615. [PMID: 39842474 DOI: 10.1016/j.phrs.2025.107615] [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: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
In recent years, increasing evidence has supported that high-fat diet (HFD) can induce the chronic, low-grade neuroinflammation in the brain, which is closely associated with the impairment of cognitive function. As the key organelles responsible for energy metabolism in the cell, mitochondria are believed to involved in the pathogenesis of a variety of neurological disorders. This review summarizes the current progress in the field of the relationship between HFD exposure and neurodegenerative diseases, and outline the major routines of HFD induced neuroinflammation and its pathological significance in the pathogenesis of neurodegenerative diseases. Furthermore, the article highlights the pivotal role of mitochondrial dysfunction in driving the neuroinflammation in the setting of HFD. Danger-associated molecular patterns (DAMPs) from damaged mitochondria can activate innate immune signaling pathways, while mitochondrial dysfunction itself can lead to metabolic remodeling of inflammatory cells, thus inducing neuroinflammation. More importantly, mitochondrial damage, neuroinflammation, and insulin resistance caused by HFD form a mutually reinforcing vicious cycle, ultimately leading to the death of neurons and promoting the progression of neurodegenerative diseases. Thus, in-depth elucidation of the role and underlying mechanisms of mitochondrial dysfunction in HFD-induced metabolic disorders may not only expand our understanding of the mechanistic linkages between HFD and etiology of neurodegenerative diseases, but also help develop the specific strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Yao Bai
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China.
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
23
|
Chourasia S, Petucci C, Shoffler C, Abbasian D, Wang H, Han X, Sivan E, Brandis A, Mehlman T, Malitsky S, Itkin M, Sharp A, Rotkopf R, Dassa B, Regev L, Zaltsman Y, Gross A. MTCH2 controls energy demand and expenditure to fuel anabolism during adipogenesis. EMBO J 2025; 44:1007-1038. [PMID: 39753955 PMCID: PMC11832942 DOI: 10.1038/s44318-024-00335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 02/19/2025] Open
Abstract
Mitochondrial carrier homolog 2 (MTCH2) is a regulator of apoptosis, mitochondrial dynamics, and metabolism. Loss of MTCH2 results in mitochondrial fragmentation, an increase in whole-body energy utilization, and protection against diet-induced obesity. In this study, we used temporal metabolomics on HeLa cells to show that MTCH2 deletion results in a high ATP demand, an oxidized cellular environment, and elevated utilization of lipids, amino acids, and carbohydrates, accompanied by a decrease in several metabolites. Lipidomics analysis revealed a strategic adaptive reduction in membrane lipids and an increase in storage lipids in MTCH2 knockout cells. Importantly, MTCH2 knockout cells showed an increase in mitochondrial oxidative function, which may explain the higher energy demand. Interestingly, this imbalance in energy metabolism and reductive potential triggered by MTCH2-deletion prevents NIH3T3L1 preadipocytes from differentiating into mature adipocytes, an energy consuming reductive biosynthetic process. In summary, the loss of MTCH2 leads to increased mitochondrial oxidative activity and energy demand, creating a catabolic and oxidative environment that fails to fuel the anabolic processes required for lipid accumulation and adipocyte differentiation.
Collapse
Affiliation(s)
- Sabita Chourasia
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel.
| | - Christopher Petucci
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Clarissa Shoffler
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dina Abbasian
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hu Wang
- Barshop Institute for Longevity and Aging Studies, and Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, and Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Ehud Sivan
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Sergey Malitsky
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Maxim Itkin
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Ayala Sharp
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Ron Rotkopf
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Bareket Dassa
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Limor Regev
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Yehudit Zaltsman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Atan Gross
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
24
|
Salum KCR, Assis ISDS, Kopke ÚDA, Palhinha L, Abreu GDM, Gouvêa LW, Teixeira MR, Mattos FCC, Nogueira Neto JF, Felício RDFM, Rosado EL, Zembrzuski VM, Campos Junior M, Maya-Monteiro CM, Cabello PH, Carneiro JRI, Bozza PT, Kohlrausch FB, da Fonseca ACP. FTO rs17817449 Variant Increases the Risk of Severe Obesity in a Brazilian Cohort: A Case-Control Study. Diabetes Metab Syndr Obes 2025; 18:283-303. [PMID: 39906696 PMCID: PMC11792641 DOI: 10.2147/dmso.s451401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/17/2024] [Indexed: 02/06/2025] Open
Abstract
Purpose Obesity is a complex disease caused by a combination of genetic, environmental, and epigenetic factors, and is associated with an increased risk of chronic diseases. The leptin-melanocortin pathway integrates peripheral signals about the body's energy stores with a central neuronal circuit in the hypothalamus. This pathway has been extensively studied over the years, as genetic variations in genes related to it may play a crucial role in determining an individual's susceptibility to obesity. Therefore, we analyzed the association between obesity and specific polymorphisms in leptin-melanocortin-related genes such as LEPR rs1137101, POMC rs1042571, LEP rs7799039, BDNF rs6265, FTO rs17817449, CART rs121909065, and NPY rs16147/rs5574. Patients and Methods The study enrolled 501 participants from Rio de Janeiro, Brazil, with obesity class II or greater (BMI ≥ 35 kg/m2) and normal weight controls (18.5≤ BMI ≤24.9 kg/m2). We collected demographic, body composition, biochemical, and genotyping data by real-time PCR, and performed logistic and linear regression analyses to investigate the association of polymorphisms with severe obesity status and obesity-related quantitative parameters. Results Individuals with severe obesity had significantly higher anthropometric measures, blood pressure, and biochemical levels. The FTO rs17817449 TT genotype was associated with a significantly higher risk of developing severe obesity, and distinct cytokine expression was observed across the FTO rs17817449 genotypes. The BDNF rs6265 dominant-model and NPY rs16147 CC genotypes were associated with triglyceride levels and childhood obesity, respectively. Finally, individuals with obesity were more likely to carry a greater number of risk alleles than those without obesity. Conclusion Our study observed an important association between FTO rs17817449 polymorphism with obesity and obesity-related traits. Additionally, BDNF rs6265 dominant-model was associated with triglyceride serum levels, and NPY rs16147 may have a role in obesity onset.
Collapse
Affiliation(s)
- Kaio Cezar Rodrigues Salum
- Medical Clinic Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Human Genetics Laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Izadora Sthephanie da Silva Assis
- Medical Clinic Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Human Genetics Laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | | | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Gabriella de Medeiros Abreu
- Human Genetics Laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laura Wendling Gouvêa
- Medical Clinic Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Human Genetics Laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Myrela Ribeiro Teixeira
- Human Genetics Laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Human Genetics Laboratory, Institute of Biology, Federal Fluminense University Niterói, Rio de Janeiro, Brazil
- Postgraduate in Sciences and Biotechnology, Fluminense Federal University Niterói, Rio de Janeiro, Brazil
| | | | | | - Rafaela de Freitas Martins Felício
- Birth Defect Epidemiology laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Health Care Network for Congenital Anomalies of the Central Nervous System, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Eliane Lopes Rosado
- Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | - Fabiana Barzotto Kohlrausch
- Human Genetics Laboratory, Institute of Biology, Federal Fluminense University Niterói, Rio de Janeiro, Brazil
| | - Ana Carolina Proença da Fonseca
- Human Genetics Laboratory, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Genetics Laboratory, Grande Rio University, Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Miksza U, Bauer W, Roszkowska J, Moroz M, Buczynska A, Wiatr A, Gorska M, Adamska-Patruno E, Kretowski A. The BDNF Protein is Associated With Glucose Homeostasis and Food Intake in Carriers of Common BDNF Gene Variants. J Clin Endocrinol Metab 2025; 110:e487-e496. [PMID: 38478378 DOI: 10.1210/clinem/dgae165] [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: 01/03/2024] [Indexed: 01/22/2025]
Abstract
CONTEXT Brain-derived neurotrophic factor (BDNF) concentrations may differ between BDNF genotype carriers. These changes occur in individuals with metabolic and mental disorders. OBJECTIVE The aim of this study was to assess the associations of glucose homeostasis parameters and the frequency of food consumption with BDNF protein concentrations based on BDNF single nucleotide polymorphisms (SNPs). METHODS Among the 439 participants, some common rs10835211 BDNF gene variants were analyzed. We evaluated BDNF concentrations, and measured glucose and insulin after fasting and during oral glucose tolerance tests. Anthropometric measurements, body composition, and body fat distribution were assessed, and a 3-day food intake diary and food frequency questionnaire were completed. RESULTS We observed significant differences in BDNF concentration between AA and AG genotype rs10835211 carriers (P = .018). The group of AA genotype holders were older, and positive correlation was found between age and BDNF in the whole study population (P = .012) and in the GG genotype carriers (P = .023). Moreover, BDNF protein correlated with fasting insulin (P = .015), HOMA-IR (P = .031), HOMA-B (P = .010), and the visceral/subcutaneous adipose tissue (VAT/SAT) ratio (P = .026) in the GG genotype individuals. Presence of the GG genotype was negatively correlated with nut and seed (P = .047) and lean pork consumption (P = .015), and the BDNF protein. Moreover, we observed correlations between the frequency of chicken (P = .028), pasta (P = .033), and sweet food intake (P = .040) with BDNF concentration in the general population. Among carriers of the AA genotype, we observed a positive correlation between the consumption of rice (P = .048) and sweet food (P = .028) and the BDNF protein level. CONCLUSION Peripheral BDNF may be associated with VAT content and insulin concentrations in GG genotype carriers and may vary with particular food intake, which warrants further investigation.
Collapse
Affiliation(s)
- Urszula Miksza
- Department of Nutriomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
- Clinical Research Support Centre, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Witold Bauer
- Department of Nutriomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Joanna Roszkowska
- Department of Nutriomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Monika Moroz
- Department of Nutriomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Angelika Buczynska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Aleksandra Wiatr
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Edyta Adamska-Patruno
- Department of Nutriomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
- Clinical Research Support Centre, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Adam Kretowski
- Department of Nutriomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
- Clinical Research Support Centre, Medical University of Bialystok, 15-274 Bialystok, Poland
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
26
|
Lin B, Pan L, He H, Hu Y, Tu J, Zhang L, Cui Z, Ren X, Wang X, Nai J, Shan G. Heritability and genetic correlations of obesity indices and cardiometabolic traits in the Northern Chinese families. Ann Hum Genet 2025; 89:1-11. [PMID: 39239922 DOI: 10.1111/ahg.12578] [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: 05/23/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE This study aimed to investigate the heritability of various obesity indices and their shared genetic factors with cardiometabolic traits in the Chinese nuclear family. METHODS A total of 1270 individuals from 538 nuclear families were included in this cross-sectional study. Different indices were used to quantify fat mass and distribution, including body index mass (BMI), visceral fat index (VFI), and body fat percent (BFP). Heritability and genetic correlations for all quantitative traits were estimated using variance component models. The susceptibility-threshold model was utilized to estimate the heritability for binary traits. RESULTS Heritability estimates for obesity indices were highest for BMI (59%), followed by BFP (49%), and VFI (40%). Heritability estimates for continuous cardiometabolic traits varied from 24% to 50%. All obesity measures exhibited consistently significant positive genetic correlations with blood pressure, fasting blood glucose, and uric acid (rG range: 0.26-0.57). However, diverse genetic correlations between various obesity indices and lipid profiles were observed. Significant genetic correlations were limited to specific pairs: BFP and total cholesterol (rG = 0.24), BFP and low-density lipoprotein cholesterol (rG = 0.25), and VFI and triglyceride (rG = 0.33). CONCLUSION The genetic overlap between various obesity indices and cardiometabolic traits underscores the importance of pleiotropic genes. Further studies are warranted to investigate specific shared genetic and environmental factors between obesity and cardiometabolic diseases.
Collapse
Affiliation(s)
- Binbin Lin
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Li Pan
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Huijing He
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Yaoda Hu
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Ji Tu
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Ze Cui
- Department of Chronic and Noncommunicable Disease Prevention and Control, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, Hebei, China
| | - Xiaolan Ren
- Department of Chronic and Noncommunicable Disease Prevention and Control, Gansu Provincial Center for Disease Control and Prevention, Lanzhou, Gansu, China
| | - Xianghua Wang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Tianjin, China
| | - Jing Nai
- Clinical Laboratory, Beijing Hepingli Hospital, Beijing, China
| | - Guangliang Shan
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Su F, Pfundstein G, Sah S, Zhang S, Keable R, Hagan DW, Sharpe LJ, Clemens KJ, Begg D, Phelps EA, Brown AJ, Leshchyns'ka I, Sytnyk V. Neuronal growth regulator 1 (NEGR1) promotes the synaptic targeting of glutamic acid decarboxylase 65 (GAD65). J Neurochem 2025; 169:e16279. [PMID: 39676071 DOI: 10.1111/jnc.16279] [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: 03/26/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024]
Abstract
Neuronal growth regulator 1 (NEGR1) is a synaptic plasma membrane localized cell adhesion molecule implicated in a wide spectrum of psychiatric disorders. By RNAseq analysis of the transcriptomic changes in the brain of NEGR1-deficient mice, we found that NEGR1 deficiency affects the expression of the Gad2 gene. We show that glutamic acid decarboxylase 65 (GAD65), the Gad2 - encoded enzyme synthesizing the inhibitory neurotransmitter GABA on synaptic vesicles, accumulates non-synaptically in brains of NEGR1-deficient mice. The density of non-synaptic GAD65 accumulations is also increased in NEGR1 deficient cultured hypothalamic neurons, and this effect is rescued by re-expression of NEGR1. By using a novel biosensor of the plasma membrane attachment of GAD65, we demonstrate that GAD65 attaches to the plasma membrane. NEGR1 promotes palmitoylation-dependent clearance of GAD65 from the plasma membrane and targeting of GAD65 to plasma membrane-derived endocytic vesicles. In NEGR1 deficient cultured hypothalamic neurons, the synaptic and extrasynaptic levels of the plasma membrane attached GAD65 are increased, and the synaptic levels of GABA are reduced. NEGR1-deficient mice are characterized by reduced body weight, lower GABAergic synapse densities in the arcuate nucleus, and blunted responsiveness to the reinforcing effects of food rewards. Our results indicate that abnormalities in synaptic GABA synthesis can contribute to brain disorders associated with abnormal expression of NEGR1 in humans.
Collapse
Affiliation(s)
- Feifei Su
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Saroj Sah
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Shuyue Zhang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - D Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Kelly J Clemens
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Denovan Begg
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
28
|
Ding K, Qin X, Wang H, Wang K, Kang X, Yu Y, Liu Y, Gong H, Wu T, Chen D, Hu Y, Wang T, Wu Y. Identification of shared genetic etiology of cardiovascular and cerebrovascular diseases through common cardiometabolic risk factors. Commun Biol 2024; 7:1703. [PMID: 39730871 DOI: 10.1038/s42003-024-07417-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) and cerebrovascular diseases (CeVDs) are closely related vascular diseases, sharing common cardiometabolic risk factors (RFs). Although pleiotropic genetic variants of these two diseases have been reported, their underlying pathological mechanisms are still unclear. Leveraging GWAS summary data and using genetic correlation, pleiotropic variants identification, and colocalization analyses, we identified 11 colocalized loci for CVDs-CeVDs-BP (blood pressure), CVDs-CeVDs-LIP (lipid traits), and CVDs-CeVDs-cIMT (carotid intima-media thickness) triplets. No shared causal loci were found for CVDs-CeVDs-T2D (type 2 diabetes) or CVDs-CeVDs-BMI (body mass index) triplets. The 11 loci were mapped to 12 genes, namely CASZ1, CDKN1A, TWIST1, CDKN2B, ABO, SWAP70, SH2B3, LRCH1, FES, GOSR2, RPRML, and LDLR, where both GOSR2 and RPRML were mapped to one locus. They were enriched in pathways related to cellular response to external stimulus and regulation of the phosphate metabolic process and were highly expressed in endothelial cells, epithelial cells, and smooth muscle cells. Multi-omics analysis revealed methylation of two genes (CASZ1 and LRCH1) may play a causal role in the genetic pleiotropy. Notably, these pleiotropic loci are highly enriched in the targets of antihypertensive drugs, which further emphasizes the role of the blood pressure regulation pathway in the shared etiology of CVDs and CeVDs.
Collapse
Affiliation(s)
- Kexin Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| | - Huairong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Kun Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xiaoying Kang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Yao Yu
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Haiying Gong
- Fangshan District Center for Disease Control and Prevention, Beijing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dafang Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Tao Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yiqun Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| |
Collapse
|
29
|
Springer M, Meugnier E, Schnabl K, Hof KS, Champy MF, Sorg T, Petit-Demoulière B, Germain N, Galusca B, Estour B, Vidal H, Klingenspor M, Hager J. Loss of Sult1a1 reduces body weight and increases browning of white adipose tissue. Front Endocrinol (Lausanne) 2024; 15:1448107. [PMID: 39703864 PMCID: PMC11656314 DOI: 10.3389/fendo.2024.1448107] [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: 06/12/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Background and objective Overweight and obesity affects millions of individuals worldwide and consequently represents a major public health concern. Individuals living with overweight and obesity have difficulty maintaining a low body weight due to known physiological mechanisms which prevent further weight loss and drive weight regain. In contrast, mechanisms which promote low body weight maintenance receive less attention and are largely unknown. To uncover these intrinsic mechanisms, we investigated a human cohort of constitutionally thin (CT) individuals which maintain a low body weight and are resistant to weight gain despite exposure to an obesogenic environment. Methods To identify novel genes that contribute to low body weight maintenance, we performed transcriptomics on adipose tissue biopsies collected from CT and normal body weight (NBW) individuals and identified sulfotransferase 1A1 (SULT1A1) as a target for further investigation in mice. Sult1a1 knockout (KO) mice were fed a standard diet to assess the impact of Sult1a1 deletion on metabolic traits. To determine if high-fat feeding recapitulated the CT weight gain resistance phenotype, Sult1a1 KO mice were fed a high-fat diet for 13-weeks. A subset of wild-type and Sult1a1 KO mice from the standard diet were further analyzed for characterization of adipose tissue respiratory capacity. Results In comparison to NBW controls, adipose tissue from CT individuals expresses less SULT1A1. Sult1a1 KO mice weigh 10% less at the end of the study period and on a high-fat diet, Sult1a1 KO mice tended to gain less weight and had reduced fat mass at 14-weeks of age. These changes were associated with reduced fasting insulin and lessened adipose tissue inflammation and fibrosis. Subcutaneous adipose tissue from Sult1a1 KO mice on a standard chow diet had elevated leak respiration, uncoupling protein 1 (UCP1) expression and increased expression of a mitochondrial marker, VDAC, associating Sult1a1 deletion to adipose tissue browning. Conclusions Our results associate Sult1a1 deletion with a tendency for lower body weight through remodeling of white adipose tissue towards a brown phenotype. The presence of UCP1, the expression of an additional mitochondrial protein and increased respiratory capacity suggest browning of the subcutaneous adipose tissue depot of Sult1a1 KO mice.
Collapse
Affiliation(s)
- Margherita Springer
- Société des Produits Nestlé S.A., Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Chair for Molecular Nutritional Medicine, Technical University of Munich (TUM) School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Emmanuelle Meugnier
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAE U1397, Université Claude Bernard Lyon 1, Institut National des Sciences Appliquées de Lyon (INSA Lyon), Oullins, France
| | - Katharina Schnabl
- Chair for Molecular Nutritional Medicine, Technical University of Munich (TUM) School of Life Sciences, Technical University of Munich, Freising, Germany
- Else Kröner Fresenius Zentrum (EKFZ) für Ernährungsmedizin, Technical University of Munich, Freising, Germany
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | | | - Marie-France Champy
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Tania Sorg
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Benoit Petit-Demoulière
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Natacha Germain
- Division of Endocrinology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
- TAPE (Eating Disorders, Addictions & Extreme Bodyweight) Research Group, University Jean Monnet, Saint Etienne, France
| | - Bogdan Galusca
- Division of Endocrinology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
- TAPE (Eating Disorders, Addictions & Extreme Bodyweight) Research Group, University Jean Monnet, Saint Etienne, France
| | - Bruno Estour
- Division of Endocrinology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
- TAPE (Eating Disorders, Addictions & Extreme Bodyweight) Research Group, University Jean Monnet, Saint Etienne, France
| | - Hubert Vidal
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAE U1397, Université Claude Bernard Lyon 1, Institut National des Sciences Appliquées de Lyon (INSA Lyon), Oullins, France
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, Technical University of Munich (TUM) School of Life Sciences, Technical University of Munich, Freising, Germany
- Else Kröner Fresenius Zentrum (EKFZ) für Ernährungsmedizin, Technical University of Munich, Freising, Germany
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Jörg Hager
- Société des Produits Nestlé S.A., Nestlé Institute of Health Sciences, Lausanne, Switzerland
| |
Collapse
|
30
|
Kosse C, Ivanov J, Knight Z, Pellegrino K, Friedman J. A subcortical feeding circuit linking an interoceptive node to jaw movement. Nature 2024; 636:151-161. [PMID: 39443799 PMCID: PMC11618074 DOI: 10.1038/s41586-024-08098-1] [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: 10/24/2022] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The brain processes an array of stimuli, enabling the selection of appropriate behavioural responses, but the neural pathways linking interoceptive inputs to outputs for feeding are poorly understood1-3. Here we delineate a subcortical circuit in which brain-derived neurotrophic factor (BDNF)-expressing neurons in the ventromedial hypothalamus (VMH) directly connect interoceptive inputs to motor centres, controlling food consumption and jaw movements. VMHBDNF neuron inhibition increases food intake by gating motor sequences of feeding through projections to premotor areas of the jaw. When food is unavailable, VMHBDNF inhibition elicits consummatory behaviours directed at inanimate objects such as wooden blocks, and inhibition of perimesencephalic trigeminal area (pMe5) projections evokes rhythmic jaw movements. The activity of these neurons is decreased during food consumption and increases when food is in proximity but not consumed. Activity is also increased in obese animals and after leptin treatment. VMHBDNF neurons receive monosynaptic inputs from both agouti-related peptide (AgRP) and proopiomelanocortin neurons in the arcuate nucleus (Arc), and constitutive VMHBDNF activation blocks the orexigenic effect of AgRP activation. These data indicate an Arc → VMHBDNF → pMe5 circuit that senses the energy state of an animal and regulates consummatory behaviours in a state-dependent manner.
Collapse
Affiliation(s)
- Christin Kosse
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Jessica Ivanov
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Zachary Knight
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Pellegrino
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Jeffrey Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
31
|
Chatterjee E, Betti MJ, Sheng Q, Lin P, Emont MP, Li G, Amancherla K, Limpitikul WB, Whittaker OR, Luong K, Azzam C, Gee D, Hutter M, Flanders K, Sahu P, Garcia-Contreras M, Gokulnath P, Flynn CR, Brown J, Yu D, Rosen ED, Jensen KVK, Gamazon ER, Shah R, Das S. The extracellular vesicle transcriptome provides tissue-specific functional genomic annotation relevant to disease susceptibility in obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.18.24317277. [PMID: 39606385 PMCID: PMC11601731 DOI: 10.1101/2024.11.18.24317277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
We characterized circulating extracellular vesicles (EVs) in obese and lean humans, identifying transcriptional cargo differentially expressed in obesity. Since circulating EVs may have broad origin, we compared this obesity EV transcriptome to expression from human visceral adipose tissue derived EVs from freshly collected and cultured biopsies from the same obese individuals. Using a comprehensive set of adipose-specific epigenomic and chromatin conformation assays, we found that the differentially expressed transcripts from the EVs were those regulated in adipose by BMI-associated SNPs from a large-scale GWAS. Using a phenome-wide association study of the regulatory SNPs for the EV-derived transcripts, we identified a substantial enrichment for inflammatory phenotypes, including type 2 diabetes. Collectively, these findings represent the convergence of the GWAS (genetics), epigenomics (transcript regulation), and EV (liquid biopsy) fields, enabling powerful future genomic studies of complex diseases.
Collapse
|
32
|
Garvey WT. Future Medications for Obesity and Clinical Implications. Diabetes Spectr 2024; 37:325-334. [PMID: 39649698 PMCID: PMC11623045 DOI: 10.2337/dsi24-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Semaglutide and tirzepatide have recently been approved for obesity and found to achieve ≥15% weight loss in clinical trials. These drugs have been referred to as second-generation medications because the unprecedented degree of weight loss they afford is sufficient to treat or prevent a broad array of obesity complications and related diseases. Many other medications are in development based on the actions of nutrient-regulated hormones (NRHs), including mono-, dual-, and triple-receptor agonists/antagonists for glucagon-like peptide 1, glucose-dependent insulinotropic polypeptide, amylin, peptide tyrosine-tyrosine, and glucagon. Clinical trial evidence is accumulating that these medications ameliorate multiple biomechanical, metabolic, and vascular complications of obesity. These tools enable a comprehensive complications-centric approach to care within the contextual framework of the diagnostic term adiposity-based chronic disease (ABCD). The potential to reduce patient suffering and the huge social burden of ABCD is profound. The current era of drug development based on NRHs could represent a landmark in the history of medicine provided that societies ensure access to these medications for the patients who need them.
Collapse
Affiliation(s)
- W. Timothy Garvey
- Department of Nutrition Sciences and the UAB Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
33
|
Drucker DJ. Efficacy and Safety of GLP-1 Medicines for Type 2 Diabetes and Obesity. Diabetes Care 2024; 47:1873-1888. [PMID: 38843460 DOI: 10.2337/dci24-0003] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/14/2024] [Indexed: 10/23/2024]
Abstract
The development of glucagon-like peptide 1 receptor agonists (GLP-1RA) for type 2 diabetes and obesity was followed by data establishing the cardiorenal benefits of GLP-1RA in select patient populations. In ongoing trials investigators are interrogating the efficacy of these agents for new indications, including metabolic liver disease, peripheral artery disease, Parkinson disease, and Alzheimer disease. The success of GLP-1-based medicines has spurred the development of new molecular entities and combinations with unique pharmacokinetic and pharmacodynamic profiles, exemplified by tirzepatide, a GIP-GLP-1 receptor coagonist. Simultaneously, investigational molecules such as maritide block the GIP and activate the GLP-1 receptor, whereas retatrutide and survodutide enable simultaneous activation of the glucagon and GLP-1 receptors. Here I highlight evidence establishing the efficacy of GLP-1-based medicines, while discussing data that inform safety, focusing on muscle strength, bone density and fractures, exercise capacity, gastrointestinal motility, retained gastric contents and anesthesia, pancreatic and biliary tract disorders, and the risk of cancer. Rapid progress in development of highly efficacious GLP-1 medicines, and anticipated differentiation of newer agents in subsets of metabolic disorders, will provide greater opportunities for use of personalized medicine approaches to improve the health of people living with cardiometabolic disorders.
Collapse
Affiliation(s)
- Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
34
|
Pérez-Gutiérrez AM, Carmona R, Loucera C, Cervilla JA, Gutiérrez B, Molina E, Lopez-Lopez D, Pérez-Florido J, Zarza-Rebollo JA, López-Isac E, Dopazo J, Martínez-González LJ, Rivera M. Mutational landscape of risk variants in comorbid depression and obesity: a next-generation sequencing approach. Mol Psychiatry 2024; 29:3553-3566. [PMID: 38806690 DOI: 10.1038/s41380-024-02609-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
Major depression (MD) and obesity are complex genetic disorders that are frequently comorbid. However, the study of both diseases concurrently remains poorly addressed and therefore the underlying genetic mechanisms involved in this comorbidity remain largely unknown. Here we examine the contribution of common and rare variants to this comorbidity through a next-generation sequencing (NGS) approach. Specific genomic regions of interest in MD and obesity were sequenced in a group of 654 individuals from the PISMA-ep epidemiological study. We obtained variants across the entire frequency spectrum and assessed their association with comorbid MD and obesity, both at variant and gene levels. We identified 55 independent common variants and a burden of rare variants in 4 genes (PARK2, FGF21, HIST1H3D and RSRC1) associated with the comorbid phenotype. Follow-up analyses revealed significantly enriched gene-sets associated with biological processes and pathways involved in metabolic dysregulation, hormone signaling and cell cycle regulation. Our results suggest that, while risk variants specific to the comorbid phenotype have been identified, the genes functionally impacted by the risk variants share cell biological processes and signaling pathways with MD and obesity phenotypes separately. To the best of our knowledge, this is the first study involving a targeted sequencing approach toward the study of the comorbid MD and obesity. The framework presented here allowed a deep characterization of the genetics of the co-occurring MD and obesity, revealing insights into the mutational and functional profile that underlies this comorbidity and contributing to a better understanding of the relationship between these two disabling disorders.
Collapse
Affiliation(s)
- Ana M Pérez-Gutiérrez
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain
| | - Rosario Carmona
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), U715, Seville, Spain
| | - Carlos Loucera
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
| | - Jorge A Cervilla
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain
- Department of Psychiatry, Faculty of Medicine, University of Granada, Granada, Spain
| | - Blanca Gutiérrez
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain
- Department of Psychiatry, Faculty of Medicine, University of Granada, Granada, Spain
| | - Esther Molina
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain
- Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
| | - Daniel Lopez-Lopez
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
| | - Javier Pérez-Florido
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), U715, Seville, Spain
| | - Juan Antonio Zarza-Rebollo
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain
| | - Elena López-Isac
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain
| | - Joaquín Dopazo
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), U715, Seville, Spain
| | - Luis Javier Martínez-González
- Genomics Unit, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Margarita Rivera
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain.
- Institute of Neurosciences "Federico Olóriz", Biomedical Research Center (CIBM), University of Granada, Granada, Spain.
- Instituto de Investigación Biosanitaria, Ibs Granada, Granada, Spain.
| |
Collapse
|
35
|
Clemons HJ, Hogan DJ, Brown PO. Depot-specific mRNA expression programs in human adipocytes suggest physiological specialization via distinct developmental programs. PLoS One 2024; 19:e0311751. [PMID: 39401200 PMCID: PMC11472956 DOI: 10.1371/journal.pone.0311751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/24/2024] [Indexed: 10/17/2024] Open
Abstract
Adipose tissue is distributed in diverse locations throughout the human body. Not much is known about the extent to which anatomically distinct adipose depots are functionally distinct, specialized organs, nor whether depot-specific characteristics result from intrinsic developmental programs, as opposed to reversible physiological responses to differences in tissue microenvironment. We used DNA microarrays to compare mRNA expression patterns of isolated human adipocytes and cultured adipose stem cells, before and after ex vivo adipocyte differentiation, from seven anatomically diverse adipose tissue depots. Adipocytes from different depots display distinct gene expression programs, which are most closely shared with anatomically related depots. mRNAs whose expression differs between anatomically diverse groups of depots (e.g., subcutaneous vs. internal) suggest important functional specializations. These depot-specific differences in gene expression were recapitulated when adipocyte progenitor cells from each site were differentiated ex vivo, suggesting that progenitor cells from specific anatomic sites are deterministically programmed to differentiate into depot-specific adipocytes. Many developmental transcription factors show striking depot-specific patterns of expression, suggesting that adipocytes in each anatomic depot are programmed during early development in concert with anatomically related tissues and organs. Our results support the hypothesis that adipocytes from different depots are functionally distinct and that their depot-specific specialization reflects distinct developmental programs.
Collapse
Affiliation(s)
- Heather J. Clemons
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Daniel J. Hogan
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Patrick O. Brown
- Department of Biochemistry, Stanford University School of Medicine, Palo Alto, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Palo Alto, California, United States of America
| |
Collapse
|
36
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [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: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
37
|
Ling H, Raraigh KS, Pugh EW, Aksit MA, Zhang P, Pace RG, Faino AV, Bamshad MJ, Gibson RL, O'Neal W, Knowles MR, Blackman SM, Cutting GR. Genetic modifiers of body mass index in individuals with cystic fibrosis. Am J Hum Genet 2024; 111:2203-2218. [PMID: 39260370 PMCID: PMC11480786 DOI: 10.1016/j.ajhg.2024.08.004] [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: 12/12/2023] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/13/2024] Open
Abstract
To identify modifier loci underlying variation in body mass index (BMI) in persons with cystic fibrosis (pwCF), we performed a genome-wide association study (GWAS). Utilizing longitudinal height and weight data, along with demographic information and covariates from 4,393 pwCF, we calculated AvgBMIz representing the average of per-quarter BMI Z scores. The GWAS incorporated 9.8M single nucleotide polymorphisms (SNPs) with a minor allele frequency (MAF) > 0.005 extracted from whole-genome sequencing (WGS) of each study subject. We observed genome-wide significant association with a variant in FTO (FaT mass and Obesity-associated gene; rs28567725; p value = 1.21e-08; MAF = 0.41, β = 0.106; n = 4,393 individuals) and a variant within ADAMTS5 (A Disintegrin And Metalloproteinase with ThromboSpondin motifs 5; rs162500; p value = 2.11e-10; MAF = 0.005, β = -0.768; n = 4,085 pancreatic-insufficient individuals). Notably, BMI-associated variants in ADAMTS5 occur on a haplotype that is much more common in African (AFR, MAF = 0.183) than European (EUR, MAF = 0.006) populations (1000 Genomes project). A polygenic risk score (PRS) calculated using 924 SNPs (excluding 17 in FTO) showed significant association with AvgBMIz (p value = 2.2e-16; r2 = 0.03). Association between variants in FTO and the PRS correlation reveals similarities in the genetic architecture of BMI in CF and the general population. Inclusion of Black individuals in whom the single-gene disorder CF is much less common but genomic diversity is greater facilitated detection of association with variants that are in LD with functional SNPs in ADAMTS5. Our results illustrate the importance of population diversity, particularly when attempting to identify variants that manifest only under certain physiologic conditions.
Collapse
Affiliation(s)
- Hua Ling
- Center for Inherited Disease Research, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karen S Raraigh
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth W Pugh
- Center for Inherited Disease Research, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Melis A Aksit
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Peng Zhang
- Center for Inherited Disease Research, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rhonda G Pace
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna V Faino
- Children's Core for Biostatistics, Epidemiology and Analytics in Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Michael J Bamshad
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA; Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Center for Clinical and Translational Research, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Ronald L Gibson
- Center for Clinical and Translational Research, Seattle Children's Hospital, Seattle, WA 98105, USA; Department of Pediatrics, Division of Pulmonary & Sleep Medicine, University of Washington School of Medicine/Seattle Children's Hospital, Seattle, WA, USA
| | - Wanda O'Neal
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michael R Knowles
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott M Blackman
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Garry R Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
38
|
Renton MC, McGee SL, Howlett KF. The role of protein kinase D (PKD) in obesity: Lessons from the heart and other tissues. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119814. [PMID: 39128598 DOI: 10.1016/j.bbamcr.2024.119814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Obesity causes a range of tissue dysfunctions that increases the risk for morbidity and mortality. Protein kinase D (PKD) represents a family of stress-activated intracellular signalling proteins that regulate essential processes such as cell proliferation and differentiation, cell survival, and exocytosis. Evidence suggests that PKD regulates the cellular adaptations to the obese environment in metabolically important tissues and drives the development of a variety of diseases. This review explores the role that PKD plays in tissue dysfunction in obesity, with special consideration of the development of obesity-mediated cardiomyopathy, a distinct cardiovascular disease that occurs in the absence of common comorbidities and leads to eventual heart failure and death. The downstream mechanisms mediated by PKD that could contribute to dysfunctions observed in the heart and other metabolically important tissues in obesity, and the predicted cell types involved are discussed to suggest potential targets for the development of therapeutics against obesity-related disease.
Collapse
Affiliation(s)
- Mark C Renton
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia; The Fralin Biomedical Research Institute at Virginia Tech Carilion, Centre for Vascular and Heart Research, Roanoke, VA, USA.
| | - Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia.
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Australia.
| |
Collapse
|
39
|
Jovanovic VM, Narisu N, Bonnycastle LL, Tharakan R, Mesch KT, Glover HJ, Yan T, Sinha N, Sen C, Castellano D, Yang S, Blivis D, Ryu S, Bennett DF, Rosales-Soto G, Inman J, Ormanoglu P, LeClair CA, Xia M, Schneider M, Hernandez-Ochoa EO, Erdos MR, Simeonov A, Chen S, Collins FS, Doege CA, Tristan CA. Scalable Hypothalamic Arcuate Neuron Differentiation from Human Pluripotent Stem Cells Suitable for Modeling Metabolic and Reproductive Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601062. [PMID: 39005353 PMCID: PMC11244856 DOI: 10.1101/2024.06.27.601062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The hypothalamus, composed of several nuclei, is essential for maintaining our body's homeostasis. The arcuate nucleus (ARC), located in the mediobasal hypothalamus, contains neuronal populations with eminent roles in energy and glucose homeostasis as well as reproduction. These neuronal populations are of great interest for translational research. To fulfill this promise, we used a robotic cell culture platform to provide a scalable and chemically defined approach for differentiating human pluripotent stem cells (hPSCs) into pro-opiomelanocortin (POMC), somatostatin (SST), tyrosine hydroxylase (TH) and gonadotropin-releasing hormone (GnRH) neuronal subpopulations with an ARC-like signature. This robust approach is reproducible across several distinct hPSC lines and exhibits a stepwise induction of key ventral diencephalon and ARC markers in transcriptomic profiling experiments. This is further corroborated by direct comparison to human fetal hypothalamus, and the enriched expression of genes implicated in obesity and type 2 diabetes (T2D). Genome-wide chromatin accessibility profiling by ATAC-seq identified accessible regulatory regions that can be utilized to predict candidate enhancers related to metabolic disorders and hypothalamic development. In depth molecular, cellular, and functional experiments unveiled the responsiveness of the hPSC-derived hypothalamic neurons to hormonal stimuli, such as insulin, neuropeptides including kisspeptin, and incretin mimetic drugs such as Exendin-4, highlighting their potential utility as physiologically relevant cellular models for disease studies. In addition, differential glucose and insulin treatments uncovered adaptability within the generated ARC neurons in the dynamic regulation of POMC and insulin receptors. In summary, the establishment of this model represents a novel, chemically defined, and scalable platform for manufacturing large numbers of hypothalamic arcuate neurons and serves as a valuable resource for modeling metabolic and reproductive disorders.
Collapse
Affiliation(s)
- Vukasin M. Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Lori L. Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Ravi Tharakan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Kendall T. Mesch
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - Hannah J. Glover
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Hypothalamus Consortium
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Neelam Sinha
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Chaitali Sen
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - David Castellano
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Shu Yang
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Dvir Blivis
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Daniel F. Bennett
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Giovanni Rosales-Soto
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jason Inman
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Christopher A. LeClair
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Martin Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Erick O. Hernandez-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Michael R. Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Shuibing Chen
- Department of Surgery, Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
- Hypothalamus Consortium
| | - Francis S. Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Hypothalamus Consortium
| | - Carlos A. Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| |
Collapse
|
40
|
M JN, Bharadwaj D. The complex web of obesity: from genetics to precision medicine. Expert Rev Endocrinol Metab 2024; 19:403-418. [PMID: 38869356 DOI: 10.1080/17446651.2024.2365785] [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: 11/06/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION Obesity is a growing public health concern affecting both children and adults. Since it involves both genetic and environmental components, the management of obesity requires both, an understanding of the underlying genetics and changes in lifestyle. The knowledge of obesity genetics will enable the possibility of precision medicine in anti-obesity medications. AREAS COVERED Here, we explore health complications and the prevalence of obesity. We discuss disruptions in energy balance as a symptom of obesity, examining evolutionary theories, its multi-factorial origins, and heritability. Additionally, we discuss monogenic and polygenic obesity, the converging biological pathways, potential pharmacogenomics applications, and existing anti-obesity medications - specifically focussing on the leptin-melanocortin and incretin pathways. Comparisons between childhood and adult obesity genetics are made, along with insights into structural variants, epigenetic changes, and environmental influences on epigenetic signatures. EXPERT OPINION With recent advancements in anti-obesity drugs, genetic studies pinpoint new targets and allow for repurposing existing drugs. This creates opportunities for genotype-informed treatment options. Also, lifestyle interventions can help in the prevention and treatment of obesity by altering the epigenetic signatures. The comparison of genetic architecture in adults and children revealed a significant overlap. However, more robust studies with diverse ethnic representation is required in childhood obesity.
Collapse
Affiliation(s)
- Janaki Nair M
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Dwaipayan Bharadwaj
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
41
|
Xu Y, He C, Fan J, Zhou Y, Cheng C, Meng R, Cui Y, Li W, Gamazon ER, Zhou D. A multi-modal framework improves prediction of tissue-specific gene expression from a surrogate tissue. EBioMedicine 2024; 107:105305. [PMID: 39180788 PMCID: PMC11388271 DOI: 10.1016/j.ebiom.2024.105305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Tissue-specific analysis of the transcriptome is critical to elucidating the molecular basis of complex traits, but central tissues are often not accessible. We propose a methodology, Multi-mOdal-based framework to bridge the Transcriptome between PEripheral and Central tissues (MOTPEC). METHODS Multi-modal regulatory elements in peripheral blood are incorporated as features for gene expression prediction in 48 central tissues. To demonstrate the utility, we apply it to the identification of BMI-associated genes and compare the tissue-specific results with those derived directly from surrogate blood. FINDINGS MOTPEC models demonstrate superior performance compared with both baseline models in blood and existing models across the 48 central tissues. We identify a set of BMI-associated genes using the central tissue MOTPEC-predicted transcriptome data. The MOTPEC-based differential gene expression (DGE) analysis of BMI in the central tissues (including brain caudate basal ganglia and visceral omentum adipose tissue) identifies 378 genes overlapping the results from a TWAS of BMI, while only 162 overlapping genes are identified using gene expression in blood. Cellular perturbation analysis further supports the utility of MOTPEC for identifying trait-associated gene sets and narrowing the effect size divergence between peripheral blood and central tissues. INTERPRETATION The MOTPEC framework improves the gene expression prediction accuracy for central tissues and enhances the identification of tissue-specific trait-associated genes. FUNDING This research is supported by the National Natural Science Foundation of China 82204118 (D.Z.), the seed funding of the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province (2020E10004), the National Institutes of Health (NIH) Genomic Innovator Award R35HG010718 (E.R.G.), NIH/NHGRI R01HG011138 (E.R.G.), NIH/NIA R56AG068026 (E.R.G.), NIH Office of the Director U24OD035523 (E.R.G.), and NIH/NIGMS R01GM140287 (E.R.G.).
Collapse
Affiliation(s)
- Yue Xu
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chunfeng He
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiayao Fan
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yuan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chunxiao Cheng
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ran Meng
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Cui
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Eric R Gamazon
- Vanderbit Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Data Science Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
42
|
Fitch AK, Malhotra S, Conroy R. Differentiating monogenic and syndromic obesities from polygenic obesity: Assessment, diagnosis, and management. OBESITY PILLARS 2024; 11:100110. [PMID: 38766314 PMCID: PMC11101890 DOI: 10.1016/j.obpill.2024.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Background Obesity is a multifactorial neurohormonal disease that results from dysfunction within energy regulation pathways and is associated with increased morbidity, mortality, and reduced quality of life. The most common form is polygenic obesity, which results from interactions between multiple gene variants and environmental factors. Highly penetrant monogenic and syndromic obesities result from rare genetic variants with minimal environmental influence and can be differentiated from polygenic obesity depending on key symptoms, including hyperphagia; early-onset, severe obesity; and suboptimal responses to nontargeted therapies. Timely diagnosis of monogenic or syndromic obesity is critical to inform management strategies and reduce disease burden. We outline the physiology of weight regulation, role of genetics in obesity, and differentiating characteristics between polygenic and rare genetic obesity to facilitate diagnosis and transition toward targeted therapies. Methods In this narrative review, we focused on case reports, case studies, and natural history studies of patients with monogenic and syndromic obesities and clinical trials examining the efficacy, safety, and quality of life impact of nontargeted and targeted therapies in these populations. We also provide comprehensive algorithms for diagnosis of patients with suspected rare genetic causes of obesity. Results Patients with monogenic and syndromic obesities commonly present with hyperphagia (ie, pathologic, insatiable hunger) and early-onset, severe obesity, and the presence of hallmark characteristics can inform genetic testing and diagnostic approach. Following diagnosis, specialized care teams can address complex symptoms, and hyperphagia is managed behaviorally. Various pharmacotherapies show promise in these patient populations, including setmelanotide and glucagon-like peptide-1 receptor agonists. Conclusion Understanding the pathophysiology and differentiating characteristics of monogenic and syndromic obesities can facilitate diagnosis and management and has led to development of targeted pharmacotherapies with demonstrated efficacy for reducing body weight and hunger in the affected populations.
Collapse
Affiliation(s)
| | - Sonali Malhotra
- Harvard Medical School, Boston, MA, USA
- Rhythm Pharmaceuticals, Inc., Boston, MA, USA
- Massachussetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
43
|
Taylor R. Understanding the cause of type 2 diabetes. Lancet Diabetes Endocrinol 2024; 12:664-673. [PMID: 39038473 DOI: 10.1016/s2213-8587(24)00157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024]
Abstract
Type 2 diabetes has long been thought to have heterogenous causes, even though epidemiological studies uniformly show a tight relationship with overnutrition. The twin cycle hypothesis postulated that interaction of self-reinforcing cycles of fat accumulation inside the liver and pancreas, driven by modest but chronic positive calorie balance, could explain the development of type 2 diabetes. This hypothesis predicted that substantial weight loss would bring about a return to the non-diabetic state, permitting observation of the pathophysiology determining the transition. These changes were postulated to reflect the basic mechanisms of causation in reverse. A series of studies over the past 15 years has elucidated these underlying mechanisms. Together with other research, the interaction of environmental and genetic factors has been clarified. This knowledge has led to successful implementation of a national programme for remission of type 2 diabetes. This Review discusses the paucity of evidence for heterogeneity in causes of type 2 diabetes and summarises the in vivo pathophysiological changes, which cause this disease of overnutrition. Type 2 diabetes has a homogenous cause expressed in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Newcastle Magnetic Resonance Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
44
|
Xue P, Lin J, Tang J, Chen Y, Yu T, Chen C, Kong H, Lin C, Liu S. Association of obesity and menarche SNPs and interaction with environmental factors on precocious puberty. Pediatr Res 2024; 96:1076-1083. [PMID: 38649724 DOI: 10.1038/s41390-024-03168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 02/21/2024] [Accepted: 03/10/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Obesity is an important cause for the precocious or early puberty. However, the association between obesity-related loci and the risk of precocious puberty as well as the effect of gene-environment interaction are unclear, especially in the Chinese children population. METHODS This was a case-control study using baseline data from two cohorts and hospital cases in China. 15 SNPs loci and several environmental factors were included in the analysis of 1201 participants. Chi-square test and logistic regression were used to analyze the association between SNPs and precocious puberty. Additionally, exploratory factor analysis was conducted on 13 environmental variables, and then to explore their interaction with genes on precocious puberty. RESULTS The effect allele C of rs571312, and G of rs12970134 MC4R were associated with precocious puberty in girls with obesity. Regarding the gene-environment interaction, we found that when girls were in the high socioeconomic status, the rs571312 (OR: 3.996; 95% CI: 1.694-9.423) and rs12970134 (OR: 3.529; 95% CI: 1.452-8.573) risk genotypes had a greater effect on precocious puberty. CONCLUSIONS The obesity risk gene polymorphisms MC4R rs571312 and rs12970134 were associated with precocious puberty in Chinese girls with obesity, and girls with risk genotypes and high socioeconomic status should be given extra attention. IMPACT This is the first study that identified the association between rs571312 and rs12970134 of MC4R gene and precocious puberty in Chinese children. We found that when girls were in the high socioeconomic status, the risk genotypes of rs571312 and rs12970134 had a greater effect on precocious puberty. The results of this study have great public health implications. It is recommended that girls who are in high socioeconomic status and have a high genetic risk for early sexual maturity should closely monitor their pubertal development and consider early intervention strategies.
Collapse
Affiliation(s)
- Peng Xue
- School of Public Health, Shanghai Jiao Tong University, Shanghai, China
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianfei Lin
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingyi Tang
- School of Public Health, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Chen
- Department of Endocrinology and Genetic Metabolism, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Yu
- School of Public Health, Shanghai Jiao Tong University, Shanghai, China
- Office of Hospital Infection Management, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chang Chen
- School of Public Health, Shanghai Jiao Tong University, Shanghai, China
| | - Huijun Kong
- Department of Pediatrics, Qufu People's Hospital, Qufu, Shandong, China
| | - Cuilan Lin
- Boai Hospital of Zhongshan, Southern Medical University, Zhongshan, Guangdong, China
| | - Shijian Liu
- School of Public Health, Shanghai Jiao Tong University, Shanghai, China.
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
45
|
Takahashi I, Ohseto H, Ueno F, Oonuma T, Narita A, Obara T, Ishikuro M, Murakami K, Noda A, Hozawa A, Sugawara J, Tamiya G, Kuriyama S. Genome-wide association study based on clustering by obesity-related variables uncovers a genetic architecture of obesity in the Japanese and the UK populations. Heliyon 2024; 10:e36023. [PMID: 39247266 PMCID: PMC11379603 DOI: 10.1016/j.heliyon.2024.e36023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Whether all obesity-related variants contribute to the onset of obesity or one or a few variants cause obesity in genetically heterogeneous populations remains obscure. Here, we investigated the genetic architecture of obesity by clustering the Japanese and British populations with obesity using obesity-related factors. In Step-1, we conducted a genome-wide association study (GWAS) with body mass index (BMI) as the outcome for eligible participants. In Step-2, we assigned participants with obesity (BMI ≥25 kg/m2) to five clusters based on obesity-related factors. Subsequently, participants from each cluster and those with a BMI <25 kg/m2 were combined. A GWAS was conducted for each cluster. Several previously identified obesity-related genes were verified in Step-1. Of the genes detected in Step-1, unique obesity-related genes were detected separately for each cluster in Step-2. Our novel findings suggest that a smaller sample size with increased homogeneity may provide insights into the genetic architecture of obesity.
Collapse
Affiliation(s)
- Ippei Takahashi
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hisashi Ohseto
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Fumihiko Ueno
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Tomomi Oonuma
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Taku Obara
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Tohoku University Hospital, Sendai, Japan
| | - Mami Ishikuro
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Keiko Murakami
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Aoi Noda
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Tohoku University Hospital, Sendai, Japan
| | - Atsushi Hozawa
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Junichi Sugawara
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Gen Tamiya
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Shinichi Kuriyama
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| |
Collapse
|
46
|
Martone A, Possidente C, Fanelli G, Fabbri C, Serretti A. Genetic factors and symptom dimensions associated with antidepressant treatment outcomes: clues for new potential therapeutic targets? Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01873-1. [PMID: 39191930 DOI: 10.1007/s00406-024-01873-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
Treatment response and resistance in major depressive disorder (MDD) show a significant genetic component, but previous studies had limited power also due to MDD heterogeneity. This literature review focuses on the genetic factors associated with treatment outcomes in MDD, exploring their overlap with those associated with clinically relevant symptom dimensions. We searched PubMed for: (1) genome-wide association studies (GWASs) or whole exome sequencing studies (WESs) that investigated efficacy outcomes in MDD; (2) studies examining the association between MDD treatment outcomes and specific depressive symptom dimensions; and (3) GWASs of the identified symptom dimensions. We identified 13 GWASs and one WES of treatment outcomes in MDD, reporting several significant loci, genes, and gene sets involved in gene expression, immune system regulation, synaptic transmission and plasticity, neurogenesis and differentiation. Nine symptom dimensions were associated with poor treatment outcomes and studied by previous GWASs (anxiety, neuroticism, anhedonia, cognitive functioning, melancholia, suicide attempt, psychosis, sleep, sociability). Four genes were associated with both treatment outcomes and these symptom dimensions: CGREF1 (anxiety); MCHR1 (neuroticism); FTO and NRXN3 (sleep). Other overlapping signals were found when considering genes suggestively associated with treatment outcomes. Genetic studies of treatment outcomes showed convergence at the level of biological processes, despite no replication at gene or variant level. The genetic signals overlapping with symptom dimensions of interest may point to shared biological mechanisms and potential targets for new treatments tailored to the individual patient's clinical profile.
Collapse
Affiliation(s)
- Alfonso Martone
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Viale Carlo Pepoli 5, 40123, Bologna, Italy
| | - Chiara Possidente
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Viale Carlo Pepoli 5, 40123, Bologna, Italy
| | - Giuseppe Fanelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Viale Carlo Pepoli 5, 40123, Bologna, Italy
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Viale Carlo Pepoli 5, 40123, Bologna, Italy.
| | - Alessandro Serretti
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| |
Collapse
|
47
|
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
|
48
|
Lin CH, Ho CJ, Chen SY, Lu YT, Tsai MH. Review of pharmacogenetics of antiseizure medications: focusing on genetic variants of mechanistic targets. Front Pharmacol 2024; 15:1411487. [PMID: 39228521 PMCID: PMC11368862 DOI: 10.3389/fphar.2024.1411487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024] Open
Abstract
Antiseizure medications (ASMs) play a central role in seizure management, however, unpredictability in the response to treatment persists, even among patients with similar seizure manifestations and clinical backgrounds. An objective biomarker capable of reliably predicting the response to ASMs would profoundly impact epilepsy treatment. Presently, clinicians rely on a trial-and-error approach when selecting ASMs, a time-consuming process that can result in delays in receiving alternative non-pharmacological therapies such as a ketogenetic diet, epilepsy surgery, and neuromodulation therapies. Pharmacogenetic studies investigating the correlation between ASMs and genetic variants regarding their mechanistic targets offer promise in predicting the response to treatment. Sodium channel subunit genes have been extensively studied along with other ion channels and receptors as targets, however, the results have been conflicting, possibly due to methodological disparities including inconsistent definitions of drug response, variations in ASM combinations, and diversity of genetic variants/genes studied. Nonetheless, these studies underscore the potential effect of genetic variants on the mechanism of ASMs and consequently the prediction of treatment response. Recent advances in sequencing technology have led to the generation of large genetic datasets, which may be able to enhance the predictive accuracy of the response to ASMs.
Collapse
Affiliation(s)
- Chih-Hsiang Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chen-Jui Ho
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Ying Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yan-Ting Lu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung, Taiwan
| |
Collapse
|
49
|
Li Y, Kim M, Jiang L, Baron L, Faulkner LD, Olson DP, Li X, Gannot N, Li P, Rui L. SH2B1 Defends Against Energy Imbalance, Obesity, and Metabolic Disease via a Paraventricular Hypothalamus→Dorsal Raphe Nucleus Neurocircuit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400437. [PMID: 38885417 PMCID: PMC11336965 DOI: 10.1002/advs.202400437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/01/2024] [Indexed: 06/20/2024]
Abstract
SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Min‐Hyun Kim
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- College of Health SolutionsArizona State UniversityPhoenixAZ85004USA
| | - Lin Jiang
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Lorelei Baron
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Latrice D. Faulkner
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - David P. Olson
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Department of PediatricsUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Xingyu Li
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Noam Gannot
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Peng Li
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Life Sciences InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Biologic and Materials SciencesSchool of DentistryUniversity of MichiganAnn ArborMI48109USA
| | - Liangyou Rui
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| |
Collapse
|
50
|
Chen W, Zhou H, Zhang M, Shi Y, Li T, Qian D, Yang J, Yu F, Li G. Novel progressive deep learning algorithm for uncovering multiple single nucleotide polymorphism interactions to predict paclitaxel clearance in patients with nonsmall cell lung cancer. CANCER INNOVATION 2024; 3:e110. [PMID: 38948246 PMCID: PMC11212318 DOI: 10.1002/cai2.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/24/2023] [Accepted: 11/21/2023] [Indexed: 07/02/2024]
Abstract
Background The rate at which the anticancer drug paclitaxel is cleared from the body markedly impacts its dosage and chemotherapy effectiveness. Importantly, paclitaxel clearance varies among individuals, primarily because of genetic polymorphisms. This metabolic variability arises from a nonlinear process that is influenced by multiple single nucleotide polymorphisms (SNPs). Conventional bioinformatics methods struggle to accurately analyze this complex process and, currently, there is no established efficient algorithm for investigating SNP interactions. Methods We developed a novel machine-learning approach called GEP-CSIs data mining algorithm. This algorithm, an advanced version of GEP, uses linear algebra computations to handle discrete variables. The GEP-CSI algorithm calculates a fitness function score based on paclitaxel clearance data and genetic polymorphisms in patients with nonsmall cell lung cancer. The data were divided into a primary set and a validation set for the analysis. Results We identified and validated 1184 three-SNP combinations that had the highest fitness function values. Notably, SERPINA1, ATF3 and EGF were found to indirectly influence paclitaxel clearance by coordinating the activity of genes previously reported to be significant in paclitaxel clearance. Particularly intriguing was the discovery of a combination of three SNPs in genes FLT1, EGF and MUC16. These SNPs-related proteins were confirmed to interact with each other in the protein-protein interaction network, which formed the basis for further exploration of their functional roles and mechanisms. Conclusion We successfully developed an effective deep-learning algorithm tailored for the nuanced mining of SNP interactions, leveraging data on paclitaxel clearance and individual genetic polymorphisms.
Collapse
Affiliation(s)
- Wei Chen
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haiyan Zhou
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mingyu Zhang
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yafei Shi
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Taifeng Li
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Di Qian
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jun Yang
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Feng Yu
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Guohui Li
- Pharmacy Department, National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|