1
|
Burgess S, Cronjé HT, deGoma E, Chyung Y, Gill D. Human Genetic Evidence to Inform Clinical Development of IL-6 Signaling Inhibition for Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2025; 45:323-331. [PMID: 39633572 DOI: 10.1161/atvbaha.124.321988] [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: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) represents a significant cause of mortality, yet no medical therapies have proven efficacious. The aim of the current study was to leverage human genetic evidence to inform clinical development of IL-6 (interleukin-6) signaling inhibition for the treatment of AAA. METHODS Associations of rs2228145, a missense variant in the IL6R gene region, are expressed per additional copy of the C allele, corresponding to the genetically predicted effect of IL-6 signaling inhibition. We consider genetic associations with AAA risk in the AAAgen consortium (39 221 cases and 1 086 107 controls) and UK Biobank (1963 cases and 365 680 controls). To validate against known effects of IL-6 signaling inhibition, we present associations with rheumatoid arthritis, polymyalgia rheumatica, and severe COVID-19. To explore mechanism specificity, we present associations with thoracic aortic aneurysm, intracranial aneurysm, and coronary artery disease. We further explored genetic associations in clinically relevant subgroups of the population. RESULTS We observed strong genetic associations with AAA risk in the AAAgen consortium, UK Biobank, and FinnGen (odds ratios: 0.91 [95% CI, 0.90-0.92], P=4×10-30; 0.90 [95% CI, 0.84-0.96], P=0.001; and 0.86 [95% CI, 0.82-0.91], P=7×10-9, respectively). The association was similar for fatal AAA but with greater uncertainty due to the lower number of events. The association with AAA was of greater magnitude than associations with coronary artery disease and even rheumatological disorders for which IL-6 inhibitors have been approved. No strong associations were observed with thoracic aortic aneurysm or intracranial aneurysm. Associations attenuated toward the null in populations with concomitant rheumatological or connective tissue disease. CONCLUSIONS Inhibition of IL-6 signaling is a promising strategy for treating AAA but not other types of aneurysmal disease. These findings serve to help inform clinical development of IL-6 signaling inhibition for AAA treatment.
Collapse
Affiliation(s)
- Stephen Burgess
- Medical Research Council Biostatistics Unit (S.B., H.T.C.), University of Cambridge, United Kingdom
- Cardiovascular Epidemiology Unit (S.B.), University of Cambridge, United Kingdom
- Sequoia Genetics, London, United Kingdom (S.B., H.T.C., D.G.)
| | - Héléne T Cronjé
- Medical Research Council Biostatistics Unit (S.B., H.T.C.), University of Cambridge, United Kingdom
- Sequoia Genetics, London, United Kingdom (S.B., H.T.C., D.G.)
| | | | | | - Dipender Gill
- Sequoia Genetics, London, United Kingdom (S.B., H.T.C., D.G.)
| |
Collapse
|
2
|
Yoshiji S, Lu T, Butler-Laporte G, Carrasco-Zanini-Sanchez J, Su CY, Chen Y, Liang K, Willett JDS, Wang S, Adra D, Ilboudo Y, Sasako T, Koyama S, Nakao T, Forgetta V, Farjoun Y, Zeberg H, Zhou S, Marks-Hultström M, Machiela MJ, Kaalia R, Dashti H, Claussnitzer M, Flannick J, Wareham NJ, Mooser V, Timpson NJ, Langenberg C, Richards JB. Integrative proteogenomic analysis identifies COL6A3-derived endotrophin as a mediator of the effect of obesity on coronary artery disease. Nat Genet 2025:10.1038/s41588-024-02052-7. [PMID: 39856218 DOI: 10.1038/s41588-024-02052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
Obesity strongly increases the risk of cardiometabolic diseases, yet the underlying mediators of this relationship are not fully understood. Given that obesity strongly influences circulating protein levels, we investigated proteins mediating the effects of obesity on coronary artery disease, stroke and type 2 diabetes. By integrating two-step proteome-wide Mendelian randomization, colocalization, epigenomics and single-cell RNA sequencing, we identified five mediators and prioritized collagen type VI α3 (COL6A3). COL6A3 levels were strongly increased by body mass index and increased coronary artery disease risk. Notably, the carboxyl terminus product of COL6A3, endotrophin, drove this effect. COL6A3 was highly expressed in disease-relevant cell types and tissues. Finally, we found that body fat reduction could reduce plasma levels of COL6A3-derived endotrophin, indicating a tractable way to modify endotrophin levels. In summary, we provide actionable insights into how circulating proteins mediate the effects of obesity on cardiometabolic diseases and prioritize endotrophin as a potential therapeutic target.
Collapse
Affiliation(s)
- Satoshi Yoshiji
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada.
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada.
- Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Tianyuan Lu
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Guillaume Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Division of Infectious Diseases, McGill University Health Centre, Montréal, Québec, Canada
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julia Carrasco-Zanini-Sanchez
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Chen-Yang Su
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
| | - Yiheng Chen
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- 5 Prime Sciences, Montréal, Québec, Canada
| | - Kevin Liang
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
| | - Julian Daniel Sunday Willett
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
- Department of Anatomic Pathology and Laboratory Medicine, New York Presbyterian - Weill Cornell Medical Center, New York, NY, USA
| | | | - Darin Adra
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Yann Ilboudo
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Takayoshi Sasako
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Satoshi Koyama
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tetsushi Nakao
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Yossi Farjoun
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Fulcrum Genomics, Somerville, MA, USA
| | - Hugo Zeberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Sirui Zhou
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
| | - Michael Marks-Hultström
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rama Kaalia
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hesam Dashti
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, MA, USA
| | - Melina Claussnitzer
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jason Flannick
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Vincent Mooser
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
| | - Nicholas J Timpson
- Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - J Brent Richards
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada.
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada.
- Department of Twin Research, King's College London, London, UK.
| |
Collapse
|
3
|
Haglund A, Zuber V, Abouzeid M, Yang Y, Ko JH, Wiemann L, Otero-Jimenez M, Muhammed L, Feleke R, Nott A, Mills JD, Laaniste L, Gveric DO, Clode D, Babtie AC, Pagni S, Bellampalli R, Somani A, McDade K, Anink JJ, Mesarosova L, Fancy N, Willumsen N, Smith A, Jackson J, Alegre-Abarrategui J, Aronica E, Matthews PM, Thom M, Sisodiya SM, Srivastava PK, Malhotra D, Bryois J, Bottolo L, Johnson MR. Cell state-dependent allelic effects and contextual Mendelian randomization analysis for human brain phenotypes. Nat Genet 2025:10.1038/s41588-024-02050-9. [PMID: 39794547 DOI: 10.1038/s41588-024-02050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/04/2024] [Indexed: 01/13/2025]
Abstract
Gene expression quantitative trait loci are widely used to infer relationships between genes and central nervous system (CNS) phenotypes; however, the effect of brain disease on these inferences is unclear. Using 2,348,438 single-nuclei profiles from 391 disease-case and control brains, we report 13,939 genes whose expression correlated with genetic variation, of which 16.7-40.8% (depending on cell type) showed disease-dependent allelic effects. Across 501 colocalizations for 30 CNS traits, 23.6% had a disease dependency, even after adjusting for disease status. To estimate the unconfounded effect of genes on outcomes, we repeated the analysis using nondiseased brains (n = 183) and reported an additional 91 colocalizations not present in the larger mixed disease and control dataset, demonstrating enhanced interpretation of disease-associated variants. Principled implementation of single-cell Mendelian randomization in control-only brains identified 140 putatively causal gene-trait associations, of which 11 were replicated in the UK Biobank, prioritizing candidate peripheral biomarkers predictive of CNS outcomes.
Collapse
Affiliation(s)
- Alexander Haglund
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Verena Zuber
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Maya Abouzeid
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Yifei Yang
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Jeong Hun Ko
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Liv Wiemann
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Maria Otero-Jimenez
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Louwai Muhammed
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rahel Feleke
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Alexi Nott
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - James D Mills
- Departments of Neuropathology and Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Liisi Laaniste
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Djordje O Gveric
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Daniel Clode
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Ann C Babtie
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Susanna Pagni
- Departments of Neuropathology and Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Ravishankara Bellampalli
- Departments of Neuropathology and Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Alyma Somani
- Departments of Neuropathology and Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Karina McDade
- Department of Neuropathology, University of Edinburgh, Edinburgh, UK
| | - Jasper J Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Lucia Mesarosova
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Nurun Fancy
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Nanet Willumsen
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Amy Smith
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Johanna Jackson
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | | | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Paul M Matthews
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Maria Thom
- Departments of Neuropathology and Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Sanjay M Sisodiya
- Departments of Neuropathology and Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | | | - Dheeraj Malhotra
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Research, Roche Innovation Center, Basel, Switzerland
- MS Research Unit, Biogen, Cambridge, MA, USA
| | - Julien Bryois
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Research, Roche Innovation Center, Basel, Switzerland
| | - Leonardo Bottolo
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Alan Turing Institute, London, UK.
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| | - Michael R Johnson
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
4
|
Yu Z, Yang Y, Yu Z, Yan Z, Gao R. Causal relationship between 14 micronutrients and chronic periodontitis: a Mendelian randomization study. Aust Dent J 2025. [PMID: 39749852 DOI: 10.1111/adj.13056] [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] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Chronic periodontitis is one of the most common inflammatory diseases worldwide. Micronutrients play a significant impact on health and periodontal disease progression. However, there is still a lack of conclusive studies confirming the causal association of micronutrients with chronic periodontitis. MATERIALS AND METHODS Genome-wide association study pooled data on chronic periodontitis were collected from the FinnGen consortium, and 14 micronutrients (calcium, copper, zinc, potassium, magnesium, selenium, iron, carotenoids, vitamin B6, B12, C, D, E and folate) were obtained from the OpenGWAS and UK Biobank Study. Instrumental variables were screened by strict criteria. Five Mendelian randomization (MR) methods were used. Finally, the robustness of the findings was verified by sensitivity analyses. RESULTS We found that vitamin D reduced the risk of chronic periodontitis (OR: 0.605, 95%CI: 0.398-0.921, P = 0.019). However, we had no evidence of a significant association between other micronutrients and chronic periodontitis. Multiple sensitivity analyses confirmed the robustness of the findings. A variety of sensitivity analysis methods confirmed the reliability of the results. CONCLUSION In this study, we found a potential causal relationship between vitamin D and chronic periodontitis by MR analysis, whereas we did not find an association between 13 other micronutrients and chronic periodontitis. This result might provide new ideas for the prevention and treatment of chronic periodontitis to some extent. However, despite the rigorous analytical methods used in this study, this finding needs to be interpreted with caution. Further randomized controlled studies might provide more in-depth references to the findings of this study.
Collapse
Affiliation(s)
- Z Yu
- Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Y Yang
- Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Z Yu
- Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Z Yan
- Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - R Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Rosoff DB, Wagner J, Jung J, Pacher P, Christodoulides C, Davey Smith G, Ray D, Lohoff FW. Multiomic Mendelian Randomization Study Investigating the Impact of PCSK9 and HMGCR Inhibition on Type 2 Diabetes Across Five Populations. Diabetes 2025; 74:120-130. [PMID: 39418486 DOI: 10.2337/db24-0451] [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: 05/29/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
The prevalence of type 2 diabetes (T2D) varies among populations of different races/ethnicities. The influence of genetically proxied LDL cholesterol lowering through proprotein convertase subtilisin/kexin 9 (PCSK9) and HMG-CoA reductase (HMGCR) on T2D in non-European populations is not well established. A drug target Mendelian randomization approach was used to assess the effects of PCSK9 and HMGCR inhibition on T2D risk and glycemic traits in five populations: East Asian (EAS), South Asian (SAS), Hispanic (HISP), African (AFR), and Europe (EUR). Our study did not find relationships between genetically proxied PCSK9 inhibition and T2D risk in the EAS (odds ratio [OR] 1.02; 95% CI 0.95-1.10), SAS (1.05; 0.97-1.14), HISP (1.03; 0.94-1.12), or EUR population (1.04; 0.98-1.11). However, in the AFR population, primary analyses suggested an increased risk of T2D resulting from PCSK9 inhibition (OR 1.53; 95% CI 1.058-2.22; P = 0.024), although this was not supported in sensitivity analyses. Genetically proxied HMGCR inhibition was associated with an increased risk of T2D in SAS (OR 1.44; 95% CI 1.30-1.61; P = 9.8 × 10-12), EAS (1.36; 1.22-1.51; P = 4.2 × 10-10), and EUR populations (1.52; 1.21-1.90; P = 3.3 × 10-4). These results were consistent across various sensitivity analyses, including colocalization, indicating a robust finding. The findings indicate a neutral impact of long-term PCSK9 inhibition on T2D and glycemic markers in most non-EUR populations, with a potential increased risk in AFR cohorts. By contrast, HMGCR inhibition increased the risk of T2D in SAS, EAS, and EUR cohorts, underscoring the need to consider diversity in genetic research on metabolic diseases. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Daniel B Rosoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | - Josephin Wagner
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
| | - Jeesun Jung
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
| | - Constantinos Christodoulides
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | - David Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- National Institute for Health and Care Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
| | - Falk W Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD
| |
Collapse
|
6
|
Huang K, Zeng T, Koc S, Pettet A, Zhou J, Jain M, Sun D, Ruiz C, Ren H, Howe L, Richardson TG, Cortes A, Aiello K, Branson K, Pfenning A, Engreitz JM, Zhang MJ, Leskovec J. Small-cohort GWAS discovery with AI over massive functional genomics knowledge graph. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24318375. [PMID: 39677475 PMCID: PMC11643201 DOI: 10.1101/2024.12.03.24318375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Genome-wide association studies (GWASs) have identified tens of thousands of disease associated variants and provided critical insights into developing effective treatments. However, limited sample sizes have hindered the discovery of variants for uncommon and rare diseases. Here, we introduce KGWAS, a novel geometric deep learning method that leverages a massive functional knowledge graph across variants and genes to improve detection power in small-cohort GWASs significantly. KGWAS assesses the strength of a variant's association to disease based on the aggregate GWAS evidence across molecular elements interacting with the variant within the knowledge graph. Comprehensive simulations and replication experiments showed that, for small sample sizes ( N =1-10K), KGWAS identified up to 100% more statistically significant associations than state-of-the-art GWAS methods and achieved the same statistical power with up to 2.67× fewer samples. We applied KGWAS to 554 uncommon UK Biobank diseases ( N case <5K) and identified 183 more associations (46.9% improvement) than the original GWAS, where the gain further increases to 79.8% for 141 rare diseases (N case <300). The KGWAS-only discoveries are supported by abundant functional evidence, such as rs2155219 (on 11q13) associated with ulcerative colitis potentially via regulating LRRC32 expression in CD4+ regulatory T cells, and rs7312765 (on 12q12) associated with the rare disease myasthenia gravis potentially via regulating PPHLN1 expression in neuron-related cell types. Furthermore, KGWAS consistently improves downstream analyses such as identifying disease-specific network links for interpreting GWAS variants, identifying disease-associated genes, and identifying disease-relevant cell populations. Overall, KGWAS is a flexible and powerful AI model that integrates growing functional genomics data to discover novel variants, genes, cells, and networks, especially valuable for small cohort diseases.
Collapse
|
7
|
Woolf B, Cronjé HT, Zagkos L, Larsson SC, Gill D, Burgess S. Comparison of caffeine consumption behavior with plasma caffeine levels as exposure measures in drug-target mendelian randomization. Am J Epidemiol 2024; 193:1776-1784. [PMID: 38904434 PMCID: PMC7616520 DOI: 10.1093/aje/kwae143] [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: 05/30/2023] [Revised: 05/09/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
Mendelian randomization is an epidemiologic technique that can explore the potential effect of perturbing a pharmacological target. Plasma caffeine levels can be used as a biomarker to measure the pharmacological effects of caffeine. Alternatively, this can be assessed using a behavioral proxy, such as average number of caffeinated drinks consumed per day. Either variable can be used as the exposure in a Mendelian randomization investigation, and to select which genetic variants to use as instrumental variables. Another possibility is to choose variants in gene regions with known biological relevance to caffeine level regulation. These choices affect the causal question that is being addressed by the analysis, and the validity of the analysis assumptions. Further, even when using the same genetic variants, the sign of Mendelian randomization estimates (positive or negative) can change depending on the choice of exposure. Some genetic variants that decrease caffeine metabolism associate with higher levels of plasma caffeine, but lower levels of caffeine consumption, as individuals with these variants require less caffeine consumption for the same physiological effect. We explore Mendelian randomization estimates for the effect of caffeine on body mass index, and discuss implications for variant and exposure choice in drug target Mendelian randomization investigations.
Collapse
Affiliation(s)
- Benjamin Woolf
- School of Psychological Science, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- MRC Biostatistics Unit at the University of Cambridge, Cambridge, UK
| | - Héléne T Cronjé
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, United Kingdom
| | - Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine Karolinska Institutet, Stockholm, Sweden
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit at the University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Masana L, Ibarretxe D. New drugs for treating dyslipidemias. From small molecules to small interfering RNAs. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36 Suppl 1:S15-S23. [PMID: 39645293 DOI: 10.1016/j.arteri.2024.07.004] [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: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 12/09/2024]
Abstract
Despite the various therapeutic tools available, many patients do not achieve therapeutic goals, and cardiovascular diseases remain a significant cause of death in our setting. Furthermore, even in patients who manage to reduce their LDL-C levels to the recommended targets, cardiovascular events continue to occur. The therapeutic challenge and the persistent risk have led to active research into new drugs targeting novel therapeutic pathways in the field of lipoprotein metabolism disorders. The therapeutic approach involves new pharmacological mechanisms, ranging from small molecules and monoclonal antibodies to RNA interference, with inclisiran being the first drug approved for clinical use in the cardiovascular domain. In this review, we aim to provide a comprehensive overview of the new therapeutic targets and pharmacological mechanisms under development, as well as their potential clinical impact.
Collapse
Affiliation(s)
- Lluís Masana
- Unitat de Recerca en Lípids i Arteriosclerosi, Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España.
| | - Daiana Ibarretxe
- Unitat de Recerca en Lípids i Arteriosclerosi, Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| |
Collapse
|
9
|
Clarke SLN, Maghsoudlou P, Guly CM, Dick AD, Ramanan AV. The management of adult and paediatric uveitis for rheumatologists. Nat Rev Rheumatol 2024; 20:795-808. [PMID: 39506056 DOI: 10.1038/s41584-024-01181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Uveitis encompasses multiple different conditions that are all characterized by intra-ocular inflammation. Uveitis occurs in the context of many different rheumatological conditions and carries a substantial risk to vision. Uveitis can develop both at the early stages of rheumatic diseases, sometimes even preceding other clinical features, and at later stages of disease. Uveitis can also occur as either a direct or an indirect complication of therapies used to treat patients with rheumatic disease. Conversely, patients with uveitis of non-rheumatic aetiology sometimes require immunosuppression, a treatment option that is not readily accessible to ophthalmologists. Thus, collaborative working between rheumatologists and ophthalmologists is critical for optimal management of patients with uveitis. This Review is written with rheumatologists in mind, to assist in the care of patients with uveitis. We collate and summarize the latest evidence and best practice in the diagnosis, management and prognostication of uveitis, including future trends and research priorities.
Collapse
Affiliation(s)
- Sarah L N Clarke
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK
- Department of Paediatrics, Royal United Hospital Bath, Bath, UK
- MRC Integrative Epidemiology Unit and School of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Panagiotis Maghsoudlou
- Regional Ocular Inflammatory Service, Bristol Eye Hospital, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Catherine M Guly
- Regional Ocular Inflammatory Service, Bristol Eye Hospital, Bristol, UK
| | - Andrew D Dick
- Regional Ocular Inflammatory Service, Bristol Eye Hospital, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Institute of Ophthalmology, University College London, London, UK
- NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - Athimalaipet V Ramanan
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK.
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
10
|
Tiwari R, Dev D, Thalla M, Aher VD, Mundada AB, Mundada PA, Vaghela K. Nano-enabled pharmacogenomics: revolutionizing personalized drug therapy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-26. [PMID: 39589779 DOI: 10.1080/09205063.2024.2431426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024]
Abstract
The combination of pharmacogenomics and nanotechnology science of pharmacogenomics into a highly advanced single entity has given birth to personalized medicine known as nano-enabled pharmacogenomics. This review article covers all aspects starting from pharmacogenomics to gene editing tools, how these have evolved or are likely to be evolved for pharmacogenomic application, and how these can be delivered using nanoparticle delivery systems. In this prior work, we explore the evolution of pharmacogenomics over the years, as well as new achievements in the field of genomic sciences, the challenges in drug creation, and application of the strategy of personalized medicine. Particular attention is paid to how nanotechnology helps avoid the problems that accompanied the development of pharmacogenomics earlier, for example, the question of drug resistance and targeted delivery. We also review the latest developments in nano-enabled pharmacogenomics, such as the coupling with other nanobio-technologies, artificial intelligence, and machine learning in pharmacogenomics, and the ethical and regulatory aspects of these developing technologies. The possible uses of nanotechnology in improving the chances of pated and treating drug-resistant cancers are exemplified by case studies together with the current clinical uses of nanotechnology. In the last section, we discuss the future trends and research prospects in this dynamically growing area, stressing the importance of further advancements and collaborations which will advance the nano-enabled pharmacogenomics to their maximum potential.
Collapse
Affiliation(s)
- Ruchi Tiwari
- Psit-Pranveer Singh Institute of Technology (Pharmacy), Kanpur-Agra-Delhi National, Kanpur, India
| | - Dhruv Dev
- Department of Pharmacy, Shivalik College of Pharmacy Nangal, Rupnagar, India
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX, USA
| | - Vaibhav Dagaji Aher
- Department of Pharmaceutical Medicine, Maharashtra University of Health Sciences, Nashik, India
| | - Anand Badrivishal Mundada
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | | | - Krishna Vaghela
- Department of Pharmacy, Saraswati Institute of Pharmaceutical Sciences, National Forensic Sciences University, Gandhinagar, India
| |
Collapse
|
11
|
Pires LVL. Is This a Causal Relationship? Mendelian Randomization as a Statistical Method for Unraveling Connections. Arq Bras Cardiol 2024; 121:e20240606. [PMID: 39570163 PMCID: PMC11634222 DOI: 10.36660/abc.20240606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/22/2024] Open
Affiliation(s)
- Lucas Vieira Lacerda Pires
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São PauloInstituto do CoraçãoSão PauloSPBrasilInstituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP – Brasil
| |
Collapse
|
12
|
Yan R, Liu L, Tzoulaki I, Fan J, Targher G, Yuan Z, Zhao J. Genetic Evidence for GLP-1 and GIP Receptors as Targets for Treatment and Prevention of MASLD/MASH. Liver Int 2024. [PMID: 39487684 DOI: 10.1111/liv.16150] [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: 09/17/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND AND AIMS Glucagon-like peptide-1 receptor (GLP1R) agonists and glucose-dependent insulinotropic polypeptide receptor (GIPR) agonists may help treat metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). However, their definitive effects are still unclear. Our study aims to clarify this uncertainty. METHODS We utilised conventional Mendelian randomisation (MR) analysis to explore potential causal links between plasma GLP-1/GIP concentrations and MASLD and its related traits. Next, we conducted drug-target MR analysis using highly expressed tissue data to assess the effects of corresponding drug perturbation on these traits. Finally, mediation analysis was performed to ascertain whether the potential causal effect is direct or mediated by other MASLD-related traits. RESULTS Circulating 2-h GLP-1 and GIP concentrations measured during an oral glucose tolerance test showed hepatoprotective effects on MASLD risk (ORGLP-1 = 0.168 [95% CI 0.033-0.839], p = 0.030; ORGIP = 0.331 [95% CI 0.222-0.494], p = 6.31 × 10-8). GLP1R expression in the blood had a minimal causal effect on MASLD risk, whereas GIPR expression significantly affected MASLD risk (OR = 0.671 [95% CI 0.531-0.849], p = 9.07 × 10-4). Expression levels of GLP1R or GIPR in the blood significantly influenced MASLD-related clinical traits. Mediation analysis revealed that GIPR expression protected against MASLD, even after adjusting for type 2 diabetes or body mass index. CONCLUSIONS GLP-1/GIP receptor agonists offer promise in lowering MASLD/MASH risk. GIP receptor agonists can exert direct and indirect effects on MASLD mediated by weight reduction or glycemic control improvement.
Collapse
Affiliation(s)
- Ran Yan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Institute for Medical Dataology, Shandong University, Jinan, Shandong, China
| | - Lu Liu
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Ioanna Tzoulaki
- Centre for Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Jiangao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria, Negrar di Valpolicella, Verona, Italy
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Institute for Medical Dataology, Shandong University, Jinan, Shandong, China
| | - Jian Zhao
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, Guangdong, China
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| |
Collapse
|
13
|
Webb RJ, Al-Asmakh M, Banach M, Mazidi M. Application of proteomics for novel drug discovery and risk prediction optimisation in stroke and myocardial infarction: a review of in-human studies. Drug Discov Today 2024; 29:104186. [PMID: 39306234 DOI: 10.1016/j.drudis.2024.104186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
The use of proteomics in human studies investigating stroke and myocardial infarction (MI) has been increasing, prompting a review of the literature. This revealed proteinaceous biomarkers of stroke from thrombi, brain tissue, cells, and particles, some of which cross the blood-brain barrier (BBB). Several proteins were also implicated in coronary artery disease (CAD), which often underlies MI, cholesterol transportation, and inflammation. Furthermore, the platelet proteome revealed itself as a potential therapeutic target, along with differentially expressed proteins associated with MI progression. Moreover, proteomic data enhanced the performance of conventional risk scores and causal protein discovery has improved interventions and drug development for patients with MI and other conditions. These findings suggest that proteomics holds much promise for future stroke and MI research.
Collapse
Affiliation(s)
- Richard J Webb
- School of Health and Sport Sciences, Hope Park Campus, Liverpool Hope University, Taggart Avenue, Liverpool, UK
| | - Maha Al-Asmakh
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Maciej Banach
- Faculty of Medicine, the John Paul II Catholic University of Lublin, Lublin, Poland; Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), 93-338 Lodz, Poland
| | - Mohsen Mazidi
- Department of Twin Research, King's College London, London, UK; Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
14
|
Gill D, Dib MJ, Cronjé HT, Karhunen V, Woolf B, Gagnon E, Daghlas I, Nyberg M, Drakeman D, Burgess S. Common pitfalls in drug target Mendelian randomization and how to avoid them. BMC Med 2024; 22:473. [PMID: 39407214 PMCID: PMC11481744 DOI: 10.1186/s12916-024-03700-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Drug target Mendelian randomization describes the use of genetic variants as instrumental variables for studying the effects of pharmacological agents. The paradigm can be used to inform on all aspects of drug development and has become increasingly popular over the last decade, particularly given the time- and cost-efficiency with which it can be performed even before commencing clinical studies. MAIN BODY In this review, we describe the recent emergence of drug target Mendelian randomization, its common pitfalls, how best to address them, as well as potential future directions. Throughout, we offer advice based on our experiences on how to approach these types of studies, which we hope will be useful for both practitioners and those translating the findings from such work. CONCLUSIONS Drug target Mendelian randomization is nuanced and requires a combination of biological, statistical, genetic, epidemiological, clinical, and pharmaceutical expertise to be utilized to its full potential. Unfortunately, these skillsets are relatively infrequently combined in any given study.
Collapse
Affiliation(s)
- Dipender Gill
- Sequoia Genetics, London, UK.
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, 90 Wood Lane, London, W12 0BZ, UK.
| | - Marie-Joe Dib
- Cardiovascular Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Héléne T Cronjé
- Sequoia Genetics, London, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Ville Karhunen
- Sequoia Genetics, London, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Benjamin Woolf
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- School of Psychological Science, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Eloi Gagnon
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Canada
| | - Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Michael Nyberg
- Cardiovascular Biology, Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Donald Drakeman
- University of Cambridge Centre for Health Leadership & Enterprise, Judge Business School, Trumpington Street, Cambridge, UK
- Advent Venture Partners, London, UK
| | - Stephen Burgess
- Sequoia Genetics, London, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Tambets R, Kolde A, Kolberg P, Love MI, Alasoo K. Extensive co-regulation of neighboring genes complicates the use of eQTLs in target gene prioritization. HGG ADVANCES 2024; 5:100348. [PMID: 39210598 PMCID: PMC11416642 DOI: 10.1016/j.xhgg.2024.100348] [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: 02/02/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Identifying causal genes underlying genome-wide association studies (GWASs) is a fundamental problem in human genetics. Although colocalization with gene expression quantitative trait loci (eQTLs) is often used to prioritize GWAS target genes, systematic benchmarking has been limited due to unavailability of large ground truth datasets. Here, we re-analyzed plasma protein QTL data from 3,301 individuals of the INTERVAL cohort together with 131 eQTL Catalog datasets. Focusing on variants located within or close to the affected protein identified 793 proteins with at least one cis-pQTL where we could assume that the most likely causal gene was the gene coding for the protein. We then benchmarked the ability of cis-eQTLs to recover these causal genes by comparing three Bayesian colocalization methods (coloc.susie, coloc.abf, and CLPP) and five Mendelian randomization (MR) approaches (three varieties of inverse-variance weighted MR, MR-RAPS, and MRLocus). We found that assigning fine-mapped pQTLs to their closest protein coding genes outperformed all colocalization methods regarding both precision (71.9%) and recall (76.9%). Furthermore, the colocalization method with the highest recall (coloc.susie - 46.3%) also had the lowest precision (45.1%). Combining evidence from multiple conditionally distinct colocalizing QTLs with MR increased precision to 81%, but this was accompanied by a large reduction in recall to 7.1%. Furthermore, the choice of the MR method greatly affected performance, with the standard inverse-variance-weighted MR often producing many false positives. Our results highlight that linking GWAS variants to target genes remains challenging with eQTL evidence alone, and prioritizing novel targets requires triangulation of evidence from multiple sources.
Collapse
Affiliation(s)
- Ralf Tambets
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Anastassia Kolde
- Institute of Genomics, University of Tartu, Tartu, Estonia; Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Peep Kolberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Michael I Love
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kaur Alasoo
- Institute of Computer Science, University of Tartu, Tartu, Estonia.
| |
Collapse
|
16
|
Zhao Z, Wan Y, Fu H, Ying S, Zhang P, Meng H, Song Y, Fu N. Lipid-lowering drugs and risk of rapid renal function decline: a mendelian randomization study. BMC Med Genomics 2024; 17:248. [PMID: 39379957 PMCID: PMC11463126 DOI: 10.1186/s12920-024-02020-4] [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: 10/17/2023] [Accepted: 09/25/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) patients face the risk of rapid kidney function decline leading to adverse outcomes like dialysis and mortality. Lipid metabolism might contribute to acute kidney function decline in CKD patients. Here, we utilized the Mendelian Randomization approach to investigate potential causal relationships between drug target-mediated lipid phenotypes and rapid renal function decline. METHODS In this study, we utilized two methodologies: summarized data-based Mendelian randomization (SMR) and inverse variance-weighted Mendelian randomization (IVW-MR), to approximate exposure to lipid-lowering drugs. This entailed leveraging expression quantitative trait loci (eQTL) for drug target genes and genetic variants proximal to drug target gene regions, which encode proteins associated with low-density lipoprotein (LDL) cholesterol, as identified in genome-wide association studies. The objective was to investigate causal associations with the progression of rapid kidney function decline. RESULTS The SMR analysis revealed a potential association between high expression of PCSK9 and rapid kidney function decline (OR = 1.11, 95% CI= [1.001-1.23]; p = 0.044). Similarly, IVW-MR analysis demonstrated a negative association between LDL cholesterol mediated by HMGCR and kidney function decline (OR = 0.74, 95% CI = 0.60-0.90; p = 0.003). CONCLUSION Genetically predicted inhibition of HMGCR is linked with the progression of kidney function decline, while genetically predicted PCSK9 inhibition is negatively associated with kidney function decline. Future research should incorporate clinical trials to validate the relevance of PCSK9 in preventing kidney function decline.
Collapse
Affiliation(s)
- Zhicheng Zhao
- Graduate school of Tianjin Medical University, Tianjin, 300070, China
- Department of Cardiology, Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Yu Wan
- Graduate school of Tianjin Medical University, Tianjin, 300070, China
| | - Han Fu
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuo Ying
- Department of Cardiology, Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Peng Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Haoyu Meng
- Graduate school of Tianjin Medical University, Tianjin, 300070, China
| | - Yu Song
- Graduate school of Tianjin Medical University, Tianjin, 300070, China
| | - Naikuan Fu
- Department of Cardiology, Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China.
| |
Collapse
|
17
|
Fan B, Zhao JV. Utilizing genetics and proteomics to assess the role of antihypertensive drugs in human longevity and the underlying pathways: a Mendelian randomization study. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2024; 10:537-546. [PMID: 38769606 DOI: 10.1093/ehjcvp/pvae038] [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: 04/02/2024] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Antihypertensive drugs are known to lower cardiovascular mortality, but the role of different types of antihypertensive drugs in lifespan has not been clarified. Moreover, the underlying mechanisms remain unclear. METHODS AND RESULTS To minimize confounding, we used Mendelian randomization to assess the role of different antihypertensive drug classes in longevity and examined the pathways via proteins. Genetic variants associated with systolic blood pressure (SBP) corresponding to drug-target genes were used as genetic instruments. The genetic associations with lifespan were obtained from a large genome-wide association study including 1 million European participants from UK Biobank and LifeGen. For significant antihypertensive drug classes, we performed sex-specific analysis, drug-target analysis, and colocalization. To examine the mediation pathways, we assessed the associations of 2291 plasma proteins with lifespan, and examined the associations of drug classes with the proteins affecting lifespan. After correcting for multiple testing, genetically proxied beta-blockers (BBs), calcium channel blockers (CCBs), and vasodilators were related to longer life years (BBs: 2.03, 95% CI 0.78-3.28 per 5 mmHg reduction in SBP, CCBs: 3.40, 95% CI 1.47-5.33, and vasodilators: 2.92, 95% CI 1.08-4.77). The beneficial effects of BBs and CCBs were more obvious in men. ADRB1, CACNA2D2, CACNB3, CPT1A, CPT2, and EDNRA genes were related to extended lifespan, with CPT2 further supported by colocalization evidence. Eighty-six proteins were related to lifespan, of which four proteins were affected by CCBs. CDH1 may mediate the association between CCBs and lifespan. CONCLUSIONS Beta-blockers, CCBs, and vasodilators may prolong lifespan, with potential sex differences for BBs and CCBs. The role of CCBs in lifespan is partly mediated by CDH1. Prioritizing the potential protein targets can provide new insights into healthy aging.
Collapse
Affiliation(s)
- Bohan Fan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jie V Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Wang T, Ma P, Wang X, Xia Y. Exploration of protein and genetic targets causing atrioventricular block: mendelian-randomization analyses based on eQTL data and pQTL data. BMC Cardiovasc Disord 2024; 24:528. [PMID: 39354406 PMCID: PMC11443760 DOI: 10.1186/s12872-024-04209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Atrioventricular block (AVB) is a heterogeneous group of arrhythmias. AVB can lead to sudden arrest of the heart and subsequent syncope or sudden cardiac death. Few scholars have investigated the underlying molecular mechanisms of AVB. Finding molecular markers can facilitate understanding of AVB and exploration of therapeutic targets. METHODS Two-sample Mendelian randomization (MR) analysis was undertaken with inverse variance weighted (IVW) model and Wald ratio as the primary approach. Reverse MR analysis was undertaken to identify the associated protein targets and gene targets. Expression quantitative trait loci (eQTL) data from the eQTLGen database and protein quantitative trait loci (pQTL) data from three previous large-scale proteomic studies on plasma were retrieved as exposure data. Genome-wide association study (GWAS) summary data (586 cases and 379,215 controls) for AVB were retrieved from the UK Biobank database. Colocalization analyses were undertaken to identify the effect of filtered markers on outcome data. Databases (DrugBank, Therapeutic Target, PubChem) were used to identify drugs that interacted with targets. RESULTS We discovered that 692 genes and 42 proteins showed a significant correlation with the AVB phenotype. Proteins (cadherin-5, sTie-1, Notch 1) and genes (DNAJC30, ABO) were putative molecules to AVB. Drug-interaction analyses revealed anticancer drugs such as tyrosine-kinase inhibitors and TIMD3 inhibitors could cause AVB. Other substances (e.g. toxins, neurological drugs) could also cause AVB. CONCLUSIONS We identified the proteins (cadherin-5, sTie-1, Notch 1) and gene (DNAJC30, ABO) targets associated with AVB pathogenesis. Anticancer drugs (tyrosine-kinase inhibitors, TIMD3 inhibitors), toxins, or neurological drugs could also cause AVB.
Collapse
Affiliation(s)
- Tongyu Wang
- Department of cardiology, First affiliated hospital of Dalian Mediacal University, Liaoning, China
| | - Peipei Ma
- Department of cardiology, First affiliated hospital of Dalian Mediacal University, Liaoning, China
| | - Xiaofang Wang
- Department of Biochemiacal Informatics, School of Basic Mediacal Sciences, Peking University, Beijing, 100191, China
| | - Yunlong Xia
- Department of cardiology, First affiliated hospital of Dalian Mediacal University, Liaoning, China.
| |
Collapse
|
19
|
Triozzi JL, Yu Z, Giri A, Chen HC, Wilson OD, Ferolito B, Ikizler TA, Akwo EA, Robinson-Cohen C, Gaziano JM, Cho K, Phillips LS, Tao R, Pereira AC, Hung AM. GLP1R Gene Expression and Kidney Disease Progression. JAMA Netw Open 2024; 7:e2440286. [PMID: 39453656 PMCID: PMC11581634 DOI: 10.1001/jamanetworkopen.2024.40286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/21/2024] [Indexed: 10/26/2024] Open
Abstract
Importance Glucagon-like peptide 1 receptor agonists (GLP-1RAs) may have nephroprotective properties beyond those related to weight loss and glycemic control. Objective To investigate the association of genetically proxied GLP-1RAs with kidney disease progression. Design, Setting, and Participants This genetic association study assembled a national retrospective cohort of veterans aged 18 years or older from the US Department of Veterans Affairs Million Veteran Program between January 10, 2011, and December 31, 2021. Data were analyzed from November 2023 to February 2024. Exposures Genetic risk score for systemic GLP1R gene expression that was calculated for each study participant based on genetic variants associated with GLP1R mRNA levels across all tissue samples within the Genotype-Tissue Expression project. Main Outcomes and Measures The primary composite outcome was incident end-stage kidney disease or a 40% decline in estimated glomerular filtration rate. Cox proportional hazards regression survival analysis assessed the association between genetically proxied GLP-1RAs and kidney disease progression. Results Among 353 153 individuals (92.5% men), median age was 66 years (IQR, 58.0-72.0 years) and median follow-up was 5.1 years (IQR, 3.1-7.2 years). Overall, 25.7% had diabetes, and 45.0% had obesity. A total of 4.6% experienced kidney disease progression. Overall, higher genetic GLP1R gene expression was associated with a lower risk of kidney disease progression in the unadjusted model (hazard ratio [HR], 0.96; 95% CI, 0.92-0.99; P = .02) and in the fully adjusted model accounting for baseline patient characteristics, body mass index, and the presence or absence of diabetes (HR, 0.96; 95% CI, 0.92-1.00; P = .04). The results were similar in sensitivity analyses stratified by diabetes or obesity status. Conclusions and Relevance In this genetic association study, higher GLP1R gene expression was associated with a small reduction in risk of kidney disease progression. These findings support pleiotropic nephroprotective mechanisms of GLP-1RAs independent of their effects on body weight and glycemic control.
Collapse
Affiliation(s)
- Jefferson L. Triozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Zhihong Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ayush Giri
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University, Nashville, Tennessee
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Otis D. Wilson
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville
| | - Brian Ferolito
- Million Veteran Program Coordinating Center, VA Boston Healthcare System, Boston, Massachusetts
| | - T. Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elvis A. Akwo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John Michael Gaziano
- Million Veteran Program Coordinating Center, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Brigham and Women’s Hospital and Harvard School of Medicine, Boston, Massachusetts
| | - Kelly Cho
- Million Veteran Program Coordinating Center, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Brigham and Women’s Hospital and Harvard School of Medicine, Boston, Massachusetts
| | - Lawrence S. Phillips
- VA Atlanta Health Care System, Decatur, Georgia
- Division of Endocrinology and Metabolism, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexandre C. Pereira
- Million Veteran Program Coordinating Center, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Brigham and Women’s Hospital and Harvard School of Medicine, Boston, Massachusetts
| | - Adriana M. Hung
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville
| |
Collapse
|
20
|
Khodursky S, Yuan S, Spin JM, Tsao PS, Levin MG, Damrauer SM. Plasma proteomics reveals the potential causal impact of extracellular matrix proteins on abdominal aortic aneurysm. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.20.24314065. [PMID: 39398994 PMCID: PMC11469359 DOI: 10.1101/2024.09.20.24314065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Background Abdominal aortic aneurysm (AAA) is a common and life-threatening vascular disease. Genetic studies have identified numerous associated loci, many potentially encoding plasma proteins. However, the causal effects of plasma proteins on AAA have not been thoroughly studied. We used genetic causal inference approaches to identify plasma proteins that have a potential causal impact on AAA. Methods Causal inference was performed using two-sample Mendelian randomization (MR). For AAA, we utilized recently published summary statistics from a multi-population genome-wide association (GWAS) meta-analysis including 39,221 individuals with, and 1,086,107 individuals without AAA from 14 cohorts. We used protein quantitative trait loci (pQTLs) identified in two large-scale plasma-proteomics studies (deCODE and UKB-PPP) to generate genetic instruments. We tested 2,783 plasma proteins for possible causal effects on AAA using two-sample MR with inverse variance weighting and common sensitivity analyses to evaluate the MR assumptions. Bayesian colocalization and gene ontology (GO) enrichment analyses provided additional insights. Results MR identified 90 plasma proteins associated with AAA at FDR<0.05, with 25 supported by colocalization analysis. Among those supported by both MR and colocalization were previously experimentally validated proteins such as PCSK9 (OR 1.3; 95%CI 1.2-1.4; P<1e-10), LTBP4 (OR 3.4; 95%CI 2.6-4.6; P<1e-10) and COL6A3 (OR 0.6; 95%CI 0.5-0.7; P<1e-6). GO analysis revealed enrichment of proteins found in extracellular matrix (ECM, OR 7.8; P<1e-4), some with maximal mRNA levels in aortic tissue. Bi-directional MR suggested plasma level changes were not caused by liability to AAA itself. We then investigated whether variants responsible for expression changes in the aorta also influenced plasma levels and AAA risk. Colocalization analysis showed that an aortic expression quantitative trait locus (eQTL) for COL6A3, and a splicing quantitative trait locus (sQTL) for LTBP4 colocalized with their respective plasma pQTLs and AAA signals (posterior probabilities 0.84 and 0.89, respectively). Conclusions Our results highlight proteins and pathways with potential causal effects on AAA, providing a foundation for future functional experiments. These findings suggest a possible causal pathway whereby genetic variation affecting ECM proteins expressed in the aortic wall cause their levels to change in blood plasma, influencing development of AAA.
Collapse
Affiliation(s)
- Samuel Khodursky
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shuai Yuan
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joshua M. Spin
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Philip S. Tsao
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael G. Levin
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott M. Damrauer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Chen B, Huang M, Pu B, Dong H. Impact of SGLT2 inhibitors on lower limb complications: a mendelian randomization perspective. Front Pharmacol 2024; 15:1401103. [PMID: 39355774 PMCID: PMC11442421 DOI: 10.3389/fphar.2024.1401103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/06/2024] [Indexed: 10/03/2024] Open
Abstract
Background While Sodium-glucose cotransporter 2 (SGLT2) inhibitors are effective in managing diabetes and reducing cardiovascular risk, concerns about their association with lower limb complications, including, osteomyelitis, ulcers, and peripheral artery disease (PAD), persist. This study employs Mendelian Randomization (MR) to assess the causal relationship between SGLT2 inhibitors and these lower limb safety outcomes. Methods A two-sample drug-target MR approach was used, complemented by a one-sample MR and genetic association analysis. Six SNPs were selected as instrumental variables to proxy the effect of SGLT2 inhibition. Primary outcomes were major limb safety outcomes, including osteomyelitis, lower limb ulcers, PAD, and cellulitis. The primary analytical method was the generalized inverse variance-weighted (IVW) approach, along with several sensitivity analyses. Results The MR analysis indicated no significant causal association between genetically proxied SGLT2 inhibition and most of the studied lower limb safety outcomes. However, a significant association with PAD was observed, necessitating careful interpretation due to discrepancies between IVW and MR-Egger results. Sensitivity analyses supported these findings, showing little evidence of heterogeneity or directional pleiotropy. Conclusion This study suggests that SGLT2 inhibitors may not be significantly associated with an increased risk of most lower limb safety outcomes, including osteomyelitis, lower limb ulcers, and cellulitis, in patients with type 2 diabetes. However, the complex relationship with PAD highlights the need for further research. These findings contribute to the understanding of the safety profile of SGLT2 inhibitors, supporting their continued use in diabetes management while underlining the importance of continuous safety monitoring.
Collapse
Affiliation(s)
- Baixing Chen
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Mingling Huang
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Bin Pu
- Department of Orthopedics, Suining City Traditional Chinese Medicine Hospital, Affiliated with North Sichuan Medical College, Sichuan, China
| | - Hang Dong
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
22
|
Engelbrecht E, Rodriguez OL, Watson CT. Addressing Technical Pitfalls in Pursuit of Molecular Factors That Mediate Immunoglobulin Gene Regulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:651-662. [PMID: 39007649 PMCID: PMC11333172 DOI: 10.4049/jimmunol.2400131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024]
Abstract
The expressed Ab repertoire is a critical determinant of immune-related phenotypes. Ab-encoding transcripts are distinct from other expressed genes because they are transcribed from somatically rearranged gene segments. Human Abs are composed of two identical H and L chain polypeptides derived from genes in IGH locus and one of two L chain loci. The combinatorial diversity that results from Ab gene rearrangement and the pairing of different H and L chains contributes to the immense diversity of the baseline Ab repertoire. During rearrangement, Ab gene selection is mediated by factors that influence chromatin architecture, promoter/enhancer activity, and V(D)J recombination. Interindividual variation in the composition of the Ab repertoire associates with germline variation in IGH, implicating polymorphism in Ab gene regulation. Determining how IGH variants directly mediate gene regulation will require integration of these variants with other functional genomic datasets. In this study, we argue that standard approaches using short reads have limited utility for characterizing regulatory regions in IGH at haplotype resolution. Using simulated and chromatin immunoprecipitation sequencing reads, we define features of IGH that limit use of short reads and a single reference genome, namely 1) the highly duplicated nature of the DNA sequence in IGH and 2) structural polymorphisms that are frequent in the population. We demonstrate that personalized diploid references enhance performance of short-read data for characterizing mappable portions of the locus, while also showing that long-read profiling tools will ultimately be needed to fully resolve functional impacts of IGH germline variation on expressed Ab repertoires.
Collapse
Affiliation(s)
- Eric Engelbrecht
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY
| | - Oscar L Rodriguez
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY
| | - Corey T Watson
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY
| |
Collapse
|
23
|
Li Y, Quan X, Tai Y, Wu YT, Wei B, Wu H. Causal association between 731 immunocyte phenotypes and liver cirrhosis: A bidirectional two-sample mendelian randomization analysis. World J Hepatol 2024; 16:1156-1166. [PMID: 39221101 PMCID: PMC11362904 DOI: 10.4254/wjh.v16.i8.1156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Liver cirrhosis is a progressive hepatic disease whose immunological basis has attracted increasing attention. However, it remains unclear whether a concrete causal association exists between immunocyte phenotypes and liver cirrhosis. AIM To explore the concrete causal relationships between immunocyte phenotypes and liver cirrhosis through a mendelian randomization (MR) study. METHODS Data on 731 immunocyte phenotypes were obtained from genome-wide association studies. Liver cirrhosis data were derived from the Finn Gen dataset, which included 214403 individuals of European ancestry. We used inverse variable weighting as the primary analysis method to assess the causal relationship. Sensitivity analyses were conducted to evaluate heterogeneity and horizontal pleiotropy. RESULTS The MR analysis demonstrated that 11 immune cell phenotypes have a positive association with liver cirrhosis [P < 0.05, odds ratio (OR) > 1] and that 9 immunocyte phenotypes were negatively correlated with liver cirrhosis (P < 0.05, OR < 1). Liver cirrhosis was positively linked to 9 immune cell phenotypes (P < 0.05, OR > 1) and negatively linked to 10 immune cell phenotypes (P < 0.05; OR < 1). None of these associations showed heterogeneity or horizontally pleiotropy (P > 0.05). CONCLUSION This bidirectional two-sample MR study demonstrated a concrete causal association between immunocyte phenotypes and liver cirrhosis. These findings offer new directions for the treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Ying Li
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Quan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yang Tai
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu-Tong Wu
- Department of Clinical Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Bo Wei
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hao Wu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.
| |
Collapse
|
24
|
Gill D, Dib MJ, Gill R, Bornstein SR, Burgess S, Birkenfeld AL. Effects of ACLY Inhibition on Body Weight Distribution: A Drug Target Mendelian Randomization Study. Genes (Basel) 2024; 15:1059. [PMID: 39202419 PMCID: PMC11353272 DOI: 10.3390/genes15081059] [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: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/10/2024] [Indexed: 09/03/2024] Open
Abstract
Background: Adenosine triphosphate-citrate lyase (ACLY) inhibition has proven clinically efficacious for low-density lipoprotein cholesterol (LDL-c) lowering and cardiovascular disease (CVD) risk reduction. Clinical and genetic evidence suggests that some LDL-c lowering strategies, such as 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) inhibition with statin therapy increase body weight and the risk of developing type 2 diabetes mellitus (T2DM). However, whether ACLY inhibition affects metabolic risk factors is currently unknown. We aimed to investigate the effects of ACLY inhibition on glycaemic and anthropometric traits using Mendelian randomization (MR). Methods: As genetic instruments for ACLY inhibition, we selected weakly correlated single-nucleotide polymorphisms at the ACLY gene associated with lower ACLY gene expression in the eQTLGen study (N = 31,684) and lower LDL-c levels in the Global Lipid Genetic Consortium study (N = 1.65 million). Two-sample Mendelian randomization was employed to investigate the effects of ACLY inhibition on T2DM risk, and glycaemic and anthropometric traits using summary data from large consortia, with sample sizes ranging from 151,013 to 806,834 individuals. Findings for genetically predicted ACLY inhibition were compared to those obtained for genetically predicted HMGCR inhibition using the same instrument selection strategy and outcome data. Results: Primary MR analyses showed that genetically predicted ACLY inhibition was associated with lower waist-to-hip ratio (β per 1 standard deviation lower LDL-c: -1.17; 95% confidence interval (CI): -1.61 to -0.73; p < 0.001) but not with risk of T2DM (odds ratio (OR) per standard deviation lower LDL-c: 0.74, 95% CI = 0.25 to 2.19, p = 0.59). In contrast, genetically predicted HMGCR inhibition was associated with higher waist-to-hip ratio (β = 0.15; 95%CI = 0.04 to 0.26; p = 0.008) and T2DM risk (OR = 1.73, 95% CI = 1.27 to 2.36, p < 0.001). The MR analyses considering secondary outcomes showed that genetically predicted ACLY inhibition was associated with a lower waist-to-hip ratio adjusted for body mass index (BMI) (β = -1.41; 95%CI = -1.81 to -1.02; p < 0.001). In contrast, genetically predicted HMGCR inhibition was associated with higher HbA1c (β = 0.19; 95%CI = 0.23 to 0.49; p < 0.001) and BMI (β = 0.36; 95%CI = 0.23 to 0.49; p < 0.001). Conclusions: Human genetic evidence supports the metabolically favourable effects of ACLY inhibition on body weight distribution, in contrast to HMGCR inhibition. These findings should be used to guide and prioritize ongoing clinical development efforts.
Collapse
Affiliation(s)
- Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
- Primula Group Ltd., London N8 0RL, UK;
| | - Marie-Joe Dib
- Division of Cardiovascular Medicine, Perelman School of Advanced Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | | | - Stefan R. Bornstein
- Department of Internal Medicine III, University Clinic, Technical University Dresden, D-01062 Dresden, Germany;
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany;
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London WC2R 2LS, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK;
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
| | - Andreas L. Birkenfeld
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany;
- Department of Internal Medicine IV, Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, D-72074 Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University Tübingen, D-72074Tübingen, Germany
| |
Collapse
|
25
|
Liu Y, Wang H, Yu M, Cai L, Zhao Y, Cheng Y, Deng Y, Zhao Y, Lu H, Wu X, Zhao G, Xue C, Liu H, Surakka I, Schwendeman A, Lu HS, Daugherty A, Chang L, Zhang J, Temel RE, Chen YE, Guo Y. Hypertriglyceridemia as a Key Contributor to Abdominal Aortic Aneurysm Development and Rupture: Insights from Genetic and Experimental Models. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311621. [PMID: 39211871 PMCID: PMC11361217 DOI: 10.1101/2024.08.07.24311621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease without effective medications. This study integrated genetic, proteomic, and metabolomic data to identify causation between increased triglyceride (TG)-rich lipoproteins and AAA risk. Three hypertriglyceridemia mouse models were employed to test the hypothesis that increased plasma TG concentrations accelerate AAA development and rupture. In the angiotensin II-infusion AAA model, most Lpl -deficient mice with severely high plasma TG concentrations died of aortic rupture. Consistently, Apoa5 -deficient mice with moderately increased TG concentrations had accelerated AAA development, while human APOC3 transgenic mice with dramatically increased TG concentrations exhibited aortic dissection and rupture. Increased TG concentrations and palmitate inhibited lysyl oxidase maturation. Administration of antisense oligonucleotide targeting Angptl3 profoundly inhibited AAA progression in human APOC3 transgenic mice and Apoe -deficient mice. These results indicate that hypertriglyceridemia is a key contributor to AAA pathogenesis, highlighting the importance of triglyceride-rich lipoprotein management in treating AAA.
Collapse
|
26
|
Hu X, Cai M, Xiao J, Wan X, Wang Z, Zhao H, Yang C. Benchmarking Mendelian randomization methods for causal inference using genome-wide association study summary statistics. Am J Hum Genet 2024; 111:1717-1735. [PMID: 39059387 PMCID: PMC11339627 DOI: 10.1016/j.ajhg.2024.06.016] [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: 01/31/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Mendelian randomization (MR), which utilizes genetic variants as instrumental variables (IVs), has gained popularity as a method for causal inference between phenotypes using genetic data. While efforts have been made to relax IV assumptions and develop new methods for causal inference in the presence of invalid IVs due to confounding, the reliability of MR methods in real-world applications remains uncertain. Instead of using simulated datasets, we conducted a benchmark study evaluating 16 two-sample summary-level MR methods using real-world genetic datasets to provide guidelines for the best practices. Our study focused on the following crucial aspects: type I error control in the presence of various confounding scenarios (e.g., population stratification, pleiotropy, and family-level confounders like assortative mating), the accuracy of causal effect estimates, replicability, and power. By comprehensively evaluating the performance of compared methods over one thousand exposure-outcome trait pairs, our study not only provides valuable insights into the performance and limitations of the compared methods but also offers practical guidance for researchers to choose appropriate MR methods for causal inference.
Collapse
Affiliation(s)
- Xianghong Hu
- School of Mathematical Sciences, Institute of Statistical Sciences, Shenzhen University, Shenzhen 518060, China; Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong, China; Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou 511458, China
| | - Mingxuan Cai
- Department of Biostatistics, City University of Hong Kong, Hong Kong, China
| | - Jiashun Xiao
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China
| | - Xiaomeng Wan
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong, China; Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou 511458, China
| | - Zhiwei Wang
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong, China; Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou 511458, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, USA.
| | - Can Yang
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong, China; Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou 511458, China; Big Data Bio-Intelligence Lab, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| |
Collapse
|
27
|
Xie Y, Zhu S, Wu S, Liu C, Shen J, Jin C, Ma H, Xiang M. Hypnotic Use and the Risk of Cardiovascular Diseases in Insomnia Patients. Eur J Prev Cardiol 2024:zwae263. [PMID: 39110833 DOI: 10.1093/eurjpc/zwae263] [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: 04/01/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 11/30/2024]
Abstract
AIMS We aimed to examine the association between hypnotic agents and cardiovascular outcomes in general individuals with insomnia. METHODS In a propensity score matched cohort of UK Biobank (UKB) participants with insomnia, Cox proportional hazard model was used to estimate the association between regular use of hypnotic agents and predetermined cardiovascular outcomes including incident coronary heart diseases (CHD), heart failure (HF), stroke, and cardiovascular death. Inverse probability of treatment weighting, competing risk models, and shared frailty models were further performed during sensitivity analysis. Drug-target Mendelian randomization (MR) analyses were employed for further evaluation of the association between therapeutic targets of hypnotics and cardiovascular diseases. RESULTS During a median follow-up of 14.3 years, the matched cohort documented a total of 929 CHD cases, 360 HF cases, 262 stroke cases, and 180 cardiovascular deaths. No significant association was detected between Z-meds and CHD, stroke, and cardiovascular mortality. Benzodiazepine use was significantly associated with the increased risk of CHD, HF, and cardiovascular mortality. The inverse probability of treatment weighting, competing risk models, and shared frailty models didn't alter the above associations. Moreover, drug-target MR analyses corroborated the safety of Z-meds in the general population regarding cardiovascular health. CONCLUSIONS Our findings suggested the heterogeneous associations between different categories of hypnotics and incident cardiovascular events in individuals with insomnia. Both observational and genetic evidence raised safety concerns regarding the cardiovascular impact of benzodiazepines. No cardiovascular hazard of Z-meds was discovered in the UKB population with insomnia.
Collapse
Affiliation(s)
- Yao Xie
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Shiyu Zhu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Shuang Wu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Chang Liu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jian Shen
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Chunna Jin
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Hong Ma
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
28
|
Zisman AL. Thiazides for Nephrolithiasis Prevention: Written in Stone? Am J Kidney Dis 2024; 84:241-243. [PMID: 38461949 DOI: 10.1053/j.ajkd.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/28/2024] [Accepted: 02/02/2024] [Indexed: 03/12/2024]
Affiliation(s)
- Anna L Zisman
- University of Chicago Pritzker School of Medicine, Chicago, Illinois.
| |
Collapse
|
29
|
Pu J, Zhao Z, Duan Y, Lu J, Yao Y, Wen Y, Li Y, Zhang Y, Ye F. Causal role of immune cells in uveitis: a Mendelian randomization study. Front Med (Lausanne) 2024; 11:1445775. [PMID: 39144656 PMCID: PMC11322614 DOI: 10.3389/fmed.2024.1445775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Background Uveitis refers to a group inflammation affecting the uvea, retina, retinal blood vessels as well as vitreous body, which is one of the common causes of blindness. There is growing evidence linking different types of immune cells to uveitis, although it remains uncertain if these associations imply causal relationships. Recent advancements in high-density genetic markers like SNPs or CNVs for genotyping, along with the progress in genome-wide association studies (GWAS) technologies, have improved our understanding of the immunological mechanisms involved in ocular diseases. Therefore, our objective was to investigate the potential causal link between immune cells and uveitis using a Mendelian randomization study. Methods The exposure and outcome GWAS data for this study were sourced from an open-access database (https://gwas.mrcieu.ac.uk/). Two-sample MR analysis was utilized to evaluate the causal relationship between 731 immune cell features and uveitis. Various MR methods were employed to reduce bias and obtain dependable estimates of the causal link between the immune cell variables and the outcomes. Instrumental variable selection criteria were carefully chosen to enhance the accuracy and efficacy of the causal relationship between different immune cell types and the risk of uveitis. Results Using two-sample MR, IVW modeling showed that GAD had significant effect on immunophenotypes. CD3 levels on CD45RA- CD4+ T cells (OR = 1.087, 95%CI = 1.029 ~ 1.147, p = 0.003) and CD3 levels on CM CD4+ T cells (OR = 1.086, 95%CI = 1.033 ~ 1.141, p = 0.001) were found to be elevated in cases of uveitis. HLA DR levels in CD14- CD16+ monocyte cells (OR = 0.735, 95% CI = 0.635 ~ 0.850, p < 0.001) and HLA DR levels in NK cells (OR = 0.910, 95% CI = 0.851 ~ 0.972, p = 0.005) were observed to be reduced in individuals with uveitis. Furthermore, Two cells were identified to be significantly associated with uveitis risk: HLA DR on in NK cells (OR = 0.938, 95%CI = 0.899 ~ 0.979, p = 0.003), HLA DR on CD14- CD16+ monocytes (OR = 0.924, 95%CI = 0.878 ~ 0.972, p = 0.002). Conclusion This study highlights the intricate relationship between immune cells and generalized anxiety disorder using genetic methods, offering valuable insights for future clinical investigations.
Collapse
Affiliation(s)
- Jianping Pu
- Department of Ophthalmology, Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zhuanghong Zhao
- Department of Ophthalmology, Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yanping Duan
- Department of Ophthalmology, Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jun Lu
- Department of Pathology, Kunming Maternal and Children Hospital, Kunming, Yunnan, China
| | - Yuchen Yao
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Yuxin Wen
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Yanxun Li
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Yu Zhang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fengyu Ye
- Department of Ophthalmology, Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
30
|
Chen Y, Wang G, Chen J, Wang C, Dong X, Chang HM, Yuan S, Zhao Y, Mu L. Genetic and Epigenetic Landscape for Drug Development in Polycystic Ovary Syndrome. Endocr Rev 2024; 45:437-459. [PMID: 38298137 DOI: 10.1210/endrev/bnae002] [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: 11/13/2023] [Revised: 12/26/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
The treatment of polycystic ovary syndrome (PCOS) faces challenges as all known treatments are merely symptomatic. The US Food and Drug Administration has not approved any drug specifically for treating PCOS. As the significance of genetics and epigenetics rises in drug development, their pivotal insights have greatly enhanced the efficacy and success of drug target discovery and validation, offering promise for guiding the advancement of PCOS treatments. In this context, we outline the genetic and epigenetic advancement in PCOS, which provide novel insights into the pathogenesis of this complex disease. We also delve into the prospective method for harnessing genetic and epigenetic strategies to identify potential drug targets and ensure target safety. Additionally, we shed light on the preliminary evidence and distinctive challenges associated with gene and epigenetic therapies in the context of PCOS.
Collapse
Affiliation(s)
- Yi Chen
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Guiquan Wang
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Reproduction and Genetics, Xiamen University, Xiamen 361023, China
| | - Jingqiao Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Congying Wang
- The Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang 322000, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 40400, Taiwan
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm 171 65, Sweden
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing 100007, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University, Beijing 100191, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
31
|
Yang Q, He B, Borges MC, Lawlor DA. Accuracy in drug target Mendelian randomization of maternal and foetal health. J Hypertens 2024; 42:1283-1284. [PMID: 38818841 DOI: 10.1097/hjh.0000000000003707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Affiliation(s)
- Qian Yang
- MRC Integrative Epidemiology Unit at the University of Bristol
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Baoting He
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, PR China
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit at the University of Bristol
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
32
|
Omidiran O, Patel A, Usman S, Mhatre I, Abdelhalim H, DeGroat W, Narayanan R, Singh K, Mendhe D, Ahmed Z. GWAS advancements to investigate disease associations and biological mechanisms. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4:e296. [PMID: 38737752 PMCID: PMC11086745 DOI: 10.1002/ctd2.296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024]
Abstract
Genome-wide association studies (GWAS) have been instrumental in elucidating the genetic architecture of various traits and diseases. Despite the success of GWAS, inherent limitations such as identifying rare and ultra-rare variants, the potential for spurious associations, and in pinpointing causative agents can undermine diagnostic capabilities. This review provides an overview of GWAS and highlights recent advances in genetics that employ a range of methodologies, including Whole Genome Sequencing (WGS), Mendelian Randomization (MR), the Pangenome's high-quality T2T-CHM13 panel, and the Human BioMolecular Atlas Program (HuBMAP), as potential enablers of current and future GWAS research. State of the literature demonstrate the capabilities of these techniques in enhancing the statistical power of GWAS. WGS, with its comprehensive approach, captures the entire genome, surpassing the capabilities of the traditional GWAS technique focused on predefined Single Nucleotide Polymorphism (SNP) sites. The Pangenome's T2T-CHM13 panel, with its holistic approach, aids in the analysis of regions with high sequence identity, such as segmental duplications (SDs). Mendelian Randomization has advanced causative inference, improving clinical diagnostics and facilitating definitive conclusions. Furthermore, spatial biology techniques like HuBMAP, enable 3D molecular mapping of tissues at single-cell resolution, offering insights into pathology of complex traits. This study aims to elucidate and advocate for the increased application of these technologies, highlighting their potential to shape the future of GWAS research.
Collapse
Affiliation(s)
- Oluwaferanmi Omidiran
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Aashna Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Sarah Usman
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Ishani Mhatre
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - William DeGroat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Rishabh Narayanan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Kritika Singh
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Dinesh Mendhe
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Zeeshan Ahmed
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA
| |
Collapse
|
33
|
Hu X, Zhang P, Gao Y, Ding WW, Cheng XE, Shi QQ, Li S, Zhu YY, Pan HF, Wang P. Identification of lipid-modifying drug targets for autoimmune diseases: insights from drug target mendelian randomization. Lipids Health Dis 2024; 23:193. [PMID: 38909219 PMCID: PMC11193261 DOI: 10.1186/s12944-024-02181-2] [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: 02/12/2024] [Accepted: 06/10/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUNDS A growing body of evidence has highlighted the interactions of lipids metabolism and immune regulation. Nevertheless, there is still a lack of evidence regarding the causality between lipids and autoimmune diseases (ADs), as well as their possibility as drug targets for ADs. OBJECTIVES This study was conducted to comprehensively understand the casual associations between lipid traits and ADs, and evaluate the therapeutic possibility of lipid-lowering drug targets on ADs. METHODS Genetic variants for lipid traits and variants encoding targets of various lipid-lowering drugs were derived from Global Lipid Genetics Consortium (GLGC) and verified in Drug Bank. Summary data of ADs were obtained from MRC Integrative Epidemiology Unit (MER-IEU) database and FinnGen consortium, respectively. The causal inferences between lipid traits/genetic agents of lipid-lowering targets and ADs were evaluated by Mendelian randomization (MR), summary data-based MR (SMR), and multivariable MR (MVMR) analyses. Enrichment analysis and protein interaction network were employed to reveal the functional characteristics and biological relevance of potential therapeutic lipid-lowering targets. RESULTS There was no evidence of causal effects regarding 5 lipid traits and 9 lipid-lowering drug targets on ADs. Genetically proxied 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibition was associated with a reduced risk of rheumatoid arthritis (RA) in both discovery (OR [odds ratio] = 0.45, 95%CI: 0.32, 0.63, P = 6.79 × 10- 06) and replicate datasets (OR = 0.37, 95%CI: 0.23, 0.61, P = 7.81 × 10- 05). SMR analyses supported that genetically proxied HMGCR inhibition had causal effects on RA in whole blood (OR = 0.48, 95%CI: 0.29, 0.82, P = 6.86 × 10- 03) and skeletal muscle sites (OR = 0.75, 95%CI: 0.56, 0.99, P = 4.48 × 10- 02). After controlling for blood pressure, body mass index (BMI), smoking and drinking alchohol, HMGCR suppression showed a direct causal effect on a lower risk of RA (OR = 0.33, 95%CI: 0.40, 0.96, P = 0.042). CONCLUSIONS Our study reveals causal links of genetically proxied HMGCR inhibition (lipid-lowering drug targets) and HMGCR expression inhibition with a decreased risk of RA, suggesting that HMGCR may serve as candidate drug targets for the treatment and prevention of RA.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Health Promotion and Behavioral Sciences, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China
| | - Peng Zhang
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Yuan Gao
- Health Services and Management, School of Health Management, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Wen-Wen Ding
- The Second Clinical School of Medicine, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Xue-Er Cheng
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Qian-Qian Shi
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Sheng Li
- Department of Health Promotion and Behavioral Sciences, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yan-Yu Zhu
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Hai-Feng Pan
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Peng Wang
- Department of Health Promotion and Behavioral Sciences, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
34
|
Gong J, Williams DM, Scholes S, Assaad S, Bu F, Hayat S, Zaninotto P, Steptoe A. Unraveling the role of plasma proteins in dementia: insights from two cohort studies in the UK, with causal evidence from Mendelian randomization. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.04.24308415. [PMID: 38883777 PMCID: PMC11177911 DOI: 10.1101/2024.06.04.24308415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Population-based proteomics offer a groundbreaking avenue to predict dementia onset. This study employed a proteome-wide, data-driven approach to investigate protein-dementia associations in 229 incident all-cause dementia (ACD) among 3,249 participants from the English Longitudinal Study of Ageing (ELSA) over a median 9.8-year follow-up, then validated in 1,506 incident ACD among 52,745 individuals from the UK Biobank (UKB) over median 13.7 years. NEFL and RPS6KB1 were robustly associated with incident ACD; MMP12 was associated with vascular dementia in ELSA. Additional markers EDA2R and KIM1 (HAVCR1) were identified from sensitivity analyses. Combining NEFL and RPS6KB1 with other factors yielded high predictive accuracy (area under the curve (AUC)=0.871) for incident ACD. Replication in the UKB confirmed associations between identified proteins with various dementia subtypes. Results from reverse Mendelian Randomization also supported the role of several proteins as early dementia biomarkers. These findings underscore proteomics' potential in identifying novel risk screening targets for dementia.
Collapse
|
35
|
Patel A, Gill D, Shungin D, Mantzoros CS, Knudsen LB, Bowden J, Burgess S. Robust use of phenotypic heterogeneity at drug target genes for mechanistic insights: Application of cis-multivariable Mendelian randomization to GLP1R gene region. Genet Epidemiol 2024; 48:151-163. [PMID: 38379245 PMCID: PMC7616158 DOI: 10.1002/gepi.22551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/08/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Phenotypic heterogeneity at genomic loci encoding drug targets can be exploited by multivariable Mendelian randomization to provide insight into the pathways by which pharmacological interventions may affect disease risk. However, statistical inference in such investigations may be poor if overdispersion heterogeneity in measured genetic associations is unaccounted for. In this work, we first develop conditional F statistics for dimension-reduced genetic associations that enable more accurate measurement of phenotypic heterogeneity. We then develop a novel extension for two-sample multivariable Mendelian randomization that accounts for overdispersion heterogeneity in dimension-reduced genetic associations. Our empirical focus is to use genetic variants in the GLP1R gene region to understand the mechanism by which GLP1R agonism affects coronary artery disease (CAD) risk. Colocalization analyses indicate that distinct variants in the GLP1R gene region are associated with body mass index and type 2 diabetes (T2D). Multivariable Mendelian randomization analyses that were corrected for overdispersion heterogeneity suggest that bodyweight lowering rather than T2D liability lowering effects of GLP1R agonism are more likely contributing to reduced CAD risk. Tissue-specific analyses prioritized brain tissue as the most likely to be relevant for CAD risk, of the tissues considered. We hope the multivariable Mendelian randomization approach illustrated here is widely applicable to better understand mechanisms linking drug targets to diseases outcomes, and hence to guide drug development efforts.
Collapse
Affiliation(s)
- Ashish Patel
- MRC Biostatistics Unit, University of Cambridge, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Dmitry Shungin
- Human Genetics Centre of Excellence, AI and Digital Research, Novo Nordisk, Denmark
| | - Christos S. Mantzoros
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
- Department of Internal Medicine, Boston VA Healthcare System, Harvard Medical School, USA
| | - Lotte Bjerre Knudsen
- Chief Scientific Advisor Office, Research and Early Development, Novo Nordisk, Denmark
| | - Jack Bowden
- Department of Clinical and Biomedical Sciences, University of Exeter, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, U.K
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, UK
- Cardiovascular Epidemiology Unit, University of Cambridge, UK
| |
Collapse
|
36
|
Chen H, Zhu G, Liu Y, Huang D, Zhang X, She L. Allergic Diseases and Chronic Adenotonsillar Diseases: A Mendelian Randomization Study. Laryngoscope 2024; 134:2653-2658. [PMID: 38193619 DOI: 10.1002/lary.31275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVE The existing epidemiological evidence regarding the intricate relationship between allergic diseases and chronic adenotonsillar diseases (CATD) remains inconclusive. Herein, the objective of our study is to explore the causal association using Mendelian randomization (MR). METHODS Employing data from large genome-wide association studies, a comprehensive two-sample bidirectional MR study was conducted. The studied traits encompassed allergic rhinitis (cases n = 9707, controls n = 331173), allergic asthma (cases n = 8525, controls n = 193857), allergic conjunctivitis (cases n = 18321, controls n = 324178), atopic dermatitis (cases n = 11964, controls n = 306909), and CATD (cases n = 38983, controls n = 258553). All the patients were of European descent and participants in cohort studies. The primary analysis was executed using inverse-variance-weighted MR. Furthermore, six additional MR methods (MR-Egger, weighted median, simple mode, weighted mode, MR pleiotropy residual sum and outlier, MR robust adjusted profile score) were employed to ensure the reliability and detect potential horizontal pleiotropy within the results. The estimates obtained from the MR analysis were factored into the overall effect calculation. RESULTS Genetically anticipated outcomes demonstrated a significant association between CATD risk and allergic rhinitis (OR = 1.141, p = 6.30E-06), allergic asthma (OR = 1.115, p = 8.31E-05), allergic conjunctivitis (OR = 1.197, p = 8.69E-07), and a suggestive association with atopic dermatitis (OR = 1.053, p = 0.040). However, no substantial correlation was observed in the reverse direction. CONCLUSIONS Findings of our study provide evidence supporting a causal role of allergic diseases in the development of CATD, whereas the converse relationship does not appear to hold true. LEVEL OF EVIDENCE 3 Laryngoscope, 134:2653-2658, 2024.
Collapse
Affiliation(s)
- Huihong Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, People's Republic of China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, People's Republic of China
| | - Gangcai Zhu
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yong Liu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, People's Republic of China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, People's Republic of China
| | - Donghai Huang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, People's Republic of China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, People's Republic of China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, People's Republic of China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, People's Republic of China
| | - Li She
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, People's Republic of China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, People's Republic of China
| |
Collapse
|
37
|
Minikel EV, Painter JL, Dong CC, Nelson MR. Refining the impact of genetic evidence on clinical success. Nature 2024; 629:624-629. [PMID: 38632401 PMCID: PMC11096124 DOI: 10.1038/s41586-024-07316-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024]
Abstract
The cost of drug discovery and development is driven primarily by failure1, with only about 10% of clinical programmes eventually receiving approval2-4. We previously estimated that human genetic evidence doubles the success rate from clinical development to approval5. In this study we leverage the growth in genetic evidence over the past decade to better understand the characteristics that distinguish clinical success and failure. We estimate the probability of success for drug mechanisms with genetic support is 2.6 times greater than those without. This relative success varies among therapy areas and development phases, and improves with increasing confidence in the causal gene, but is largely unaffected by genetic effect size, minor allele frequency or year of discovery. These results indicate we are far from reaching peak genetic insights to aid the discovery of targets for more effective drugs.
Collapse
Affiliation(s)
| | - Jeffery L Painter
- JiveCast, Raleigh, NC, USA
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | | | - Matthew R Nelson
- Deerfield Management Company LP, New York, NY, USA.
- Genscience LLC, New York, NY, USA.
| |
Collapse
|
38
|
Sharma P, Klarin D, Voight BF, Tsao PS, Levin MG, Damrauer SM. Evaluation of Plasma Biomarkers for Causal Association With Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:1114-1123. [PMID: 38545784 PMCID: PMC11043009 DOI: 10.1161/atvbaha.124.320674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Hundreds of biomarkers for peripheral artery disease (PAD) have been reported in the literature; however, the observational nature of these studies limits causal inference due to the potential of reverse causality and residual confounding. We sought to evaluate the potential causal impact of putative PAD biomarkers identified in human observational studies through genetic causal inference methods. METHODS Putative circulating PAD biomarkers were identified from human observational studies through a comprehensive literature search based on terms related to PAD using PubMed, Cochrane, and Embase. Genetic instruments were generated from publicly available genome-wide association studies of circulating biomarkers. Two-sample Mendelian randomization was used to test the association of genetically determined biomarker levels with PAD using summary statistics from a genome-wide association study of 31 307 individuals with and 211 753 individuals without PAD in the Veterans Affairs Million Veteran Program and replicated in data from FinnGen comprised of 11 924 individuals with and 288 638 individuals without PAD. RESULTS We identified 204 unique circulating biomarkers for PAD from the observational literature, of which 173 were genetically instrumented using genome-wide association study results. After accounting for multiple testing (false discovery rate, <0.05), 10 of 173 (5.8%) biomarkers had significant associations with PAD. These 10 biomarkers represented categories including plasma lipoprotein regulation, lipid homeostasis, and protein-lipid complex remodeling. Observational literature highlighted different pathways including inflammatory response, negative regulation of multicellular organismal processes, and regulation of response to external stimuli. CONCLUSIONS Integrating human observational studies and genetic causal inference highlights several key pathways in PAD pathophysiology. This work demonstrates that a substantial portion of biomarkers identified in observational studies are not well supported by human genetic evidence and emphasizes the importance of triangulating evidence to understand PAD pathophysiology. Although the identified biomarkers offer insights into atherosclerotic development in the lower limb, their specificity to PAD compared with more widespread atherosclerosis requires further study.
Collapse
Affiliation(s)
- Pranav Sharma
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Derek Klarin
- Veterans Affairs Palo Alto Healthcare System, CA
- Division of Vascular Surgery, Stanford University School of Medicine, CA
| | - Benjamin F. Voight
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United State
| | - Philip S. Tsao
- Veterans Affairs Palo Alto Healthcare System, CA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA
| | - Michael G. Levin
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Scott M. Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
39
|
Dixon P, Martin RM, Harrison S. Causal Estimation of Long-term Intervention Cost-effectiveness Using Genetic Instrumental Variables: An Application to Cancer. Med Decis Making 2024; 44:283-295. [PMID: 38426435 PMCID: PMC10988994 DOI: 10.1177/0272989x241232607] [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: 07/29/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND This article demonstrates a means of assessing long-term intervention cost-effectiveness in the absence of data from randomized controlled trials and without recourse to Markov simulation or similar types of cohort simulation. METHODS Using a Mendelian randomization study design, we developed causal estimates of the genetically predicted effect of bladder, breast, colorectal, lung, multiple myeloma, ovarian, prostate, and thyroid cancers on health care costs and quality-adjusted life-years (QALYs) using outcome data drawn from the UK Biobank cohort. We then used these estimates in a simulation model to estimate the cost-effectiveness of a hypothetical population-wide preventative intervention based on a repurposed class of antidiabetic drugs known as sodium-glucose cotransporter-2 (SGLT2) inhibitors very recently shown to reduce the odds of incident prostate cancer. RESULTS Genetic liability to prostate cancer and breast cancer had material causal impacts on either or both health care costs and QALYs. Mendelian randomization results for the less common cancers were associated with considerable uncertainty. SGLT2 inhibition was unlikely to be a cost-effective preventative intervention for prostate cancer, although this conclusion depended on the price at which these drugs would be offered for a novel anticancer indication. IMPLICATIONS Our new causal estimates of cancer exposures on health economic outcomes may be used as inputs into decision-analytic models of cancer interventions such as screening programs or simulations of longer-term outcomes associated with therapies investigated in randomized controlled trials with short follow-ups. Our method allowed us to rapidly and efficiently estimate the cost-effectiveness of a hypothetical population-scale anticancer intervention to inform and complement other means of assessing long-term intervention value. HIGHLIGHTS The article demonstrates a novel method of assessing long-term intervention cost-effectiveness without relying on randomized controlled trials or cohort simulations.Mendelian randomization was used to estimate the causal effects of certain cancers on health care costs and quality-adjusted life-years (QALYs) using data from the UK Biobank cohort.Given causal data on the association of different cancer exposures on costs and QALYs, it was possible to simulate the cost-effectiveness of an anticancer intervention.Genetic liability to prostate cancer and breast cancer significantly affected health care costs and QALYs, but the hypothetical intervention using SGLT2 inhibitors for prostate cancer may not be cost-effective, depending on the drug's price for the new anticancer indication. The methods we propose and implement can be used to efficiently estimate intervention cost-effectiveness and to inform decision making in all manner of preventative and therapeutic contexts.
Collapse
Affiliation(s)
- Padraig Dixon
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Richard M. Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, UK
| | - Sean Harrison
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- UK Health Security Agency
| |
Collapse
|
40
|
Chen L, Zhao Y, Li M, Lv G. Proteome-wide Mendelian randomization highlights AIF1 and HLA-DQA2 as targets for primary sclerosing cholangitis. Hepatol Int 2024; 18:517-528. [PMID: 37950809 DOI: 10.1007/s12072-023-10608-8] [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: 07/18/2023] [Accepted: 10/09/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND Primary sclerosing cholangitis (PSC) is a kind of cholestatic liver disease without effective therapies and its pathogenesis is largely unknown. METHODS We performed the proteome-wide Mendelian randomization (MR) design to estimate the causal associations of protein levels with PSC risk. Therein, genetic associations with 4,907 plasma protein levels were extracted from a proteome-wide genome-wide association study (GWAS) with 35,559 individuals and those with PSC were obtained from the International PSC Study Group (2,871 cases and 12,019 controls) and the FinnGen study (1,491 cases and 301,383 controls). The colocalization analysis was performed to detect causal variants shared by proteins and PSC. The identified proteins were further enriched in pathways and diseases. A phenome-wide association screening was performed and potential drugs were assessed as well. RESULTS The results indicated that genetically predicted plasma levels of 14 proteins were positively associated with an increased risk of PSC and 8 proteins were inversely associated with PSC risk in both PSC GWAS data sets, and they all survived in sensitivity analyses. The colocalization indicated that AIF1 (allograft inflammatory factor 1) and HLA-DQA2 (major histocompatibility complex, class II, DQ alpha 2) were shared proteins with PSC, and they should be direct targets for PSC. The phenome-wide screening suggested that variants located at AIF1 or HLA-DQA2 region were closely associated with several autoimmune diseases, such as rheumatoid arthritis, implicating the shared pathogenesis among them. CONCLUSIONS Our study highly pinpointed two candidate targets (AIF1 and HLA-DQA2) for PSC.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yuexuan Zhao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Mingyue Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
41
|
Zhao HH, Ma Z, Guan DS. Causal role of immune cells in obstructive sleep apnea hypopnea syndrome: Mendelian randomization study. World J Clin Cases 2024; 12:1227-1234. [PMID: 38524502 PMCID: PMC10955532 DOI: 10.12998/wjcc.v12.i7.1227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/02/2024] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Despite being one of the most prevalent sleep disorders, obstructive sleep apnea hypoventilation syndrome (OSAHS) has limited information on its immunologic foundation. The immunological underpinnings of certain major psychiatric diseases have been uncovered in recent years thanks to the extensive use of genome-wide association studies (GWAS) and genotyping techniques using high-density genetic markers (e.g., SNP or CNVs). But this tactic hasn't yet been applied to OSAHS. Using a Mendelian randomization analysis, we analyzed the causal link between immune cells and the illness in order to comprehend the immunological bases of OSAHS. AIM To investigate the immune cells' association with OSAHS via genetic methods, guiding future clinical research. METHODS A comprehensive two-sample mendelian randomization study was conducted to investigate the causal relationship between immune cell characteristics and OSAHS. Summary statistics for each immune cell feature were obtained from the GWAS catalog. Information on 731 immune cell properties, such as morphologic parameters, median fluorescence intensity, absolute cellular, and relative cellular, was compiled using publicly available genetic databases. The results' robustness, heterogeneity, and horizontal pleiotropy were confirmed using extensive sensitivity examination. RESULTS Following false discovery rate (FDR) correction, no statistically significant effect of OSAHS on immunophenotypes was observed. However, two lymphocyte subsets were found to have a significant association with the risk of OSAHS: Basophil %CD33dim HLA DR- CD66b- (OR = 1.03, 95%CI = 1.01-1.03, P < 0.001); CD38 on IgD+ CD24- B cell (OR = 1.04, 95%CI = 1.02-1.04, P = 0.019). CONCLUSION This study shows a strong link between immune cells and OSAHS through a gene approach, thus offering direction for potential future medical research.
Collapse
Affiliation(s)
- Huang-Hong Zhao
- Department of Encephalopathy, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Zhen Ma
- Department of Personnel, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Dong-Sheng Guan
- Department of Neurology, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou 450000, Henan Province, China
| |
Collapse
|
42
|
Xu X, Wang SY, Wang R, Wu LY, Yan M, Sun ZL, Sun QH. Association of antihypertensive drugs with psoriasis: A trans-ancestry and drug-target Mendelian randomization study. Vascul Pharmacol 2024; 154:107284. [PMID: 38360195 DOI: 10.1016/j.vph.2024.107284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/02/2024] [Accepted: 02/11/2024] [Indexed: 02/17/2024]
Affiliation(s)
- Xiao Xu
- Department of Nursing, Nantong Health College of Jiangsu Province, Nantong, China.
| | - Shu-Yun Wang
- Academic Affair Office, Nantong Vocational University, Nantong, China; Department of Postgraduate, St. Paul University Philippines, Tuggegarau, Philippines.
| | - Rongyun Wang
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Lin-Yun Wu
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Min Yan
- Department of Epidemiology, School of Public Health, Changzhou University, Changzhou, China; Faculty of Health and Welfare, Satakunta University of Applied Sciences, Pori, Finland.
| | - Zhi-Ling Sun
- Department of Rheumatology, School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Qiu-Hua Sun
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
43
|
Wang K, Wang S, Chen Y, Lu X, Wang D, Zhang Y, Pan W, Zhou C, Zou D. Causal relationship between gut microbiota and risk of gastroesophageal reflux disease: a genetic correlation and bidirectional Mendelian randomization study. Front Immunol 2024; 15:1327503. [PMID: 38449873 PMCID: PMC10914956 DOI: 10.3389/fimmu.2024.1327503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Background Numerous observational studies have identified a linkage between the gut microbiota and gastroesophageal reflux disease (GERD). However, a clear causative association between the gut microbiota and GERD has yet to be definitively ascertained, given the presence of confounding variables. Methods The genome-wide association study (GWAS) pertaining to the microbiome, conducted by the MiBioGen consortium and comprising 18,340 samples from 24 population-based cohorts, served as the exposure dataset. Summary-level data for GERD were obtained from a recent publicly available genome-wide association involving 78 707 GERD cases and 288 734 controls of European descent. The inverse variance-weighted (IVW) method was performed as a primary analysis, the other four methods were used as supporting analyses. Furthermore, sensitivity analyses encompassing Cochran's Q statistics, MR-Egger intercept, MR-PRESSO global test, and leave-one-out methodology were carried out to identify potential heterogeneity and horizontal pleiotropy. Ultimately, a reverse MR assessment was conducted to investigate the potential for reverse causation. Results The IVW method's findings suggested protective roles against GERD for the Family Clostridiales Vadin BB60 group (P = 0.027), Genus Lachnospiraceae UCG004 (P = 0.026), Genus Methanobrevibacter (P = 0.026), and Phylum Actinobacteria (P = 0.019). In contrast, Class Mollicutes (P = 0.037), Genus Anaerostipes (P = 0.049), and Phylum Tenericutes (P = 0.024) emerged as potential GERD risk factors. In assessing reverse causation with GERD as the exposure and gut microbiota as the outcome, the findings indicate that GERD leads to dysbiosis in 13 distinct gut microbiota classes. The MR results' reliability was confirmed by thorough assessments of heterogeneity and pleiotropy. Conclusions For the first time, the MR analysis indicates a genetic link between gut microbiota abundance changes and GERD risk. This not only substantiates the potential of intestinal microecological therapy for GERD, but also establishes a basis for advanced research into the role of intestinal microbiota in the etiology of GERD.
Collapse
Affiliation(s)
- Kui Wang
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Gastroenterology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Suijian Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Stantou University Medical College, Stantou, China
| | - Yuhua Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xinchen Lu
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Danshu Wang
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Pan
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Chunhua Zhou
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Tian H, Tom BDM, Burgess S. A data-adaptive method for investigating effect heterogeneity with high-dimensional covariates in Mendelian randomization. BMC Med Res Methodol 2024; 24:34. [PMID: 38341532 PMCID: PMC10858611 DOI: 10.1186/s12874-024-02153-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Mendelian randomization is a popular method for causal inference with observational data that uses genetic variants as instrumental variables. Similarly to a randomized trial, a standard Mendelian randomization analysis estimates the population-averaged effect of an exposure on an outcome. Dividing the population into subgroups can reveal effect heterogeneity to inform who would most benefit from intervention on the exposure. However, as covariates are measured post-"randomization", naive stratification typically induces collider bias in stratum-specific estimates. METHOD We extend a previously proposed stratification method (the "doubly-ranked method") to form strata based on a single covariate, and introduce a data-adaptive random forest method to calculate stratum-specific estimates that are robust to collider bias based on a high-dimensional covariate set. We also propose measures based on the Q statistic to assess heterogeneity between stratum-specific estimates (to understand whether estimates are more variable than expected due to chance alone) and variable importance (to identify the key drivers of effect heterogeneity). RESULT We show that the effect of body mass index (BMI) on lung function is heterogeneous, depending most strongly on hip circumference and weight. While for most individuals, the predicted effect of increasing BMI on lung function is negative, it is positive for some individuals and strongly negative for others. CONCLUSION Our data-adaptive approach allows for the exploration of effect heterogeneity in the relationship between an exposure and an outcome within a Mendelian randomization framework. This can yield valuable insights into disease aetiology and help identify specific groups of individuals who would derive the greatest benefit from targeted interventions on the exposure.
Collapse
Affiliation(s)
- Haodong Tian
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| | - Brian D M Tom
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Ma Z, Zhao M, Zhao H, Qu N. Causal role of immune cells in generalized anxiety disorder: Mendelian randomization study. Front Immunol 2024; 14:1338083. [PMID: 38264647 PMCID: PMC10803460 DOI: 10.3389/fimmu.2023.1338083] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Background Generalized anxiety disorder (GAD) is a prevalent emotional disorder that has received relatively little attention regarding its immunological basis. Recent years have seen the widespread use of high-density genetic markers such as SNPs or CNVs for genotyping, as well as the advancement of genome-wide association studies (GWAS) technologies, which have facilitated the understanding of immunological mechanisms underlying several major psychiatric disorders. Despite these advancements, the immunological basis of GAD remains poorly understood. In light of this, we aimed to explore the causal relationship between immune cells and the disease through a Mendelian randomization study. Methods The summary information for GAD (Ncase=4,666, Ncontrol=337,577) was obtained from the FinnGen dataset. Summary statistics for the characterization of 731 immune cells, including morphological parameters (MP=32), median fluorescence intensity (MFI=389), absolute cells (AC=118), and relative cells (RC=192), were derived from the GWAS catalog. The study involved both forward MR analysis, with immune cell traits as the exposure and GAD as the outcome, and reverse MR analysis, with GAD as the exposure and immune cell traits as the outcome. We performed extensive sensitivity analyses to confirm the robustness, heterogeneity, and potential multi-biological effects of the study results. Also, to control for false positive results during multiple hypothesis testing, we adopted a false discovery rate (FDR) to control for statistical bias due to multiple comparisons. Results After FDR correction, GAD had no statistically significant effect on immunophenotypes. Several phenotypes with unadjusted low P-values are worth mentioning, including decreased PB/PC levels on B cells(β=-0.289, 95%CI=0.044~0.194, P=0.002), reduced PB/PC AC in GAD patients (β=-0.270, 95% CI=0.77~0.92, P=0.000), and diminished PB/PC on lymphocytes (β=-0.315, 95% CI=0.77~0.93, P=0.001). GAD also exerted a causal effect on CD27 on IgD-CD38br (β=-0.155,95%CI=0.78~0.94,P=0.002), CD20-%B cell (β= -0.105,95% CI=0.77~0.94, P=0.002), IgD-CD38br%lymphocyte(β=-0.305, 95%CI=0.79~0.95, P=0.002), FSC-A level on granulocytes (β=0.200, 95%CI=0.75~0.91, P=8.35×10-5), and CD4RA on TD CD4+(β=-0.150, 95% CI=0.82~1.02, P=0.099). Furthermore, Two lymphocyte subsets were identified to be significantly associated with GAD risk: CD24+ CD27+ B cell (OR=1.066,95%CI=1.04~1.10,P=1.237×10-5),CD28+CD4+T cell (OR=0.927, 95%CI=0.89~0.96, P=8.085×10-5). Conclusion The study has shown the close association between immune cells and GAD through genetic methods, thereby offering direction for future clinical research.
Collapse
Affiliation(s)
- Zhen Ma
- Department of Neurology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Min Zhao
- Department of Neurology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Huanghong Zhao
- Department of Neurology, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Nan Qu
- Department of Neurology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
46
|
Abstract
Importance Mendelian randomization (MR) is a statistical approach that has become increasingly popular in the field of cardiovascular disease research. It offers a way to infer potentially causal relationships between risk factors and outcomes using observational data, which is particularly important in cases where randomized clinical trials are not feasible or ethical. With the growing availability of large genetic data sets, MR has become a powerful and accessible tool for studying the risk factors for cardiovascular disease. Observations MR uses genetic variation associated with modifiable exposures or risk factors to mitigate biases that affect traditional observational study designs. The approach uses genetic variants that are randomly assigned at conception as proxies for exposure to a risk factor, mimicking a randomized clinical trial. By comparing the outcomes of individuals with different genetic variants, researchers may draw causal inferences about the effects of specific risk factors on cardiovascular disease, provided assumptions are met that address (1) the association between each genetic variant and risk factor and (2) the association of the genetic variants with confounders and (3) that the association between each genetic variant and the outcome only occurs through the risk factor. Like other observational designs, MR has limitations, which include weak instruments that are not strongly associated with the exposure of interest, linkage disequilibrium where genetic instruments influence the outcome via correlated rather than direct effects, overestimated genetic associations, and selection and survival biases. In addition, many genetic databases and MR studies primarily include populations genetically similar to European reference populations; improved diversity of participants in these databases and studies is critically needed. Conclusions and Relevance This review provides an overview of MR methodology, including assumptions, strengths, and limitations. Several important applications of MR in cardiovascular disease research are highlighted, including the identification of drug targets, evaluation of potential cardiovascular risk factors, as well as emerging methodology. Overall, while MR alone can never prove a causal relationship beyond reasonable doubt, MR offers a rigorous approach for investigating possible causal relationships in observational data and has the potential to transform our understanding of the etiology and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
47
|
Liang Y, Luo S, Wong THT, He B, Schooling CM, Au Yeung SL. Association of iron homeostasis biomarkers in type 2 diabetes and glycaemic traits: a bidirectional two-sample Mendelian randomization study. Int J Epidemiol 2023; 52:1914-1925. [PMID: 37400992 DOI: 10.1093/ije/dyad093] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/14/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Mendelian randomization (MR) studies show iron positively associated with type 2 diabetes (T2D) but included potentially biasing hereditary haemochromatosis variants and did not assess reverse causality. METHODS We assessed the relation of iron homeostasis with T2D and glycaemic traits bidirectionally, using genome-wide association studies (GWAS) of iron homeostasis biomarkers [ferritin, serum iron, total iron-binding capacity (TIBC), transferrin saturation (TSAT) (n ≤ 246 139)], T2D (DIAMANTE n = 933 970 and FinnGen n = 300 483), and glycaemic traits [fasting glucose (FG), 2-h glucose, glycated haemoglobin (HbA1c) and fasting insulin (FI) (n ≤ 209 605)]. Inverse variance weighting (IVW) was the main analysis, supplemented with sensitivity analyses and assessment of mediation by hepcidin. RESULTS Iron homeostasis biomarkers were largely unrelated to T2D, although serum iron was potentially associated with higher T2D [odds ratio: 1.07 per standard deviation; 95% confidence interval (CI): 0.99 to 1.16; P-value: 0.078) in DIAMANTE only. Higher ferritin, serum iron, TSAT and lower TIBC likely decreased HbA1c, but were not associated with other glycaemic traits. Liability to T2D likely increased TIBC (0.03 per log odds; 95% CI: 0.01 to 0.05; P-value: 0.005), FI likely increased ferritin (0.29 per log pmol/L; 95% CI: 0.12 to 0.47; P-value: 8.72 x 10-4). FG likely increased serum iron (0.06 per mmol/L; 95% CI: 0.001 to 0.12; P-value: 0.046). Hepcidin did not mediate these associations. CONCLUSION It is unlikely that ferritin, TSAT and TIBC cause T2D although an association for serum iron could not be excluded. Glycaemic traits and liability to T2D may affect iron homeostasis, but mediation by hepcidin is unlikely. Corresponding mechanistic studies are warranted.
Collapse
Affiliation(s)
- Ying Liang
- School of Public Health, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Shan Luo
- School of Public Health, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Tommy Hon Ting Wong
- School of Public Health, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Baoting He
- School of Public Health, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - C Mary Schooling
- School of Public Health, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- School of Public Health and Health Policy, City University of New York, New York, NY, USA
| | - Shiu Lun Au Yeung
- School of Public Health, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
48
|
Gill D, Zagkos L, Gill R, Benzing T, Jordan J, Birkenfeld AL, Burgess S, Zahn G. The citrate transporter SLC13A5 as a therapeutic target for kidney disease: evidence from Mendelian randomization to inform drug development. BMC Med 2023; 21:504. [PMID: 38110950 PMCID: PMC10729503 DOI: 10.1186/s12916-023-03227-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Solute carrier family 13 member 5 (SLC13A5) is a Na+-coupled citrate co-transporter that mediates entry of extracellular citrate into the cytosol. SLC13A5 inhibition has been proposed as a target for reducing progression of kidney disease. The aim of this study was to leverage the Mendelian randomization paradigm to gain insight into the effects of SLC13A5 inhibition in humans, towards prioritizing and informing clinical development efforts. METHODS The primary Mendelian randomization analyses investigated the effect of SLC13A5 inhibition on measures of kidney function, including creatinine and cystatin C-based measures of estimated glomerular filtration rate (creatinine-eGFR and cystatin C-eGFR), blood urea nitrogen (BUN), urine albumin-creatinine ratio (uACR), and risk of chronic kidney disease and microalbuminuria. Secondary analyses included a paired plasma and urine metabolome-wide association study, investigation of secondary traits related to SLC13A5 biology, a phenome-wide association study (PheWAS), and a proteome-wide association study. All analyses were compared to the effect of genetically predicted plasma citrate levels using variants selected from across the genome, and statistical sensitivity analyses robust to the inclusion of pleiotropic variants were also performed. Data were obtained from large-scale genetic consortia and biobanks, with sample sizes ranging from 5023 to 1,320,016 individuals. RESULTS We found evidence of associations between genetically proxied SLC13A5 inhibition and higher creatinine-eGFR (p = 0.002), cystatin C-eGFR (p = 0.005), and lower BUN (p = 3 × 10-4). Statistical sensitivity analyses robust to the inclusion of pleiotropic variants suggested that these effects may be a consequence of higher plasma citrate levels. There was no strong evidence of associations of genetically proxied SLC13A5 inhibition with uACR or risk of CKD or microalbuminuria. Secondary analyses identified evidence of associations with higher plasma calcium levels (p = 6 × 10-13) and lower fasting glucose (p = 0.02). PheWAS did not identify any safety concerns. CONCLUSIONS This Mendelian randomization analysis provides human-centric insight to guide clinical development of an SLC13A5 inhibitor. We identify plasma calcium and citrate as biologically plausible biomarkers of target engagement, and plasma citrate as a potential biomarker of mechanism of action. Our human genetic evidence corroborates evidence from various animal models to support effects of SLC13A5 inhibition on improving kidney function.
Collapse
Affiliation(s)
- Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- Primula Group Ltd, London, UK.
| | - Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | | | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
| | - Andreas L Birkenfeld
- Department of Diabetology Endocrinology and Nephrology, Internal Medicine IV, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Division of Translational Diabetology, Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Diabetes, School of Life Course Science and Medicine, King's College London, London, UK
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit at the University of Cambridge, Cambridge, UK
| | | |
Collapse
|
49
|
Larsson SC, Butterworth AS, Burgess S. Mendelian randomization for cardiovascular diseases: principles and applications. Eur Heart J 2023; 44:4913-4924. [PMID: 37935836 PMCID: PMC10719501 DOI: 10.1093/eurheartj/ehad736] [Citation(s) in RCA: 167] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/13/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023] Open
Abstract
Large-scale genome-wide association studies conducted over the last decade have uncovered numerous genetic variants associated with cardiometabolic traits and risk factors. These discoveries have enabled the Mendelian randomization (MR) design, which uses genetic variation as a natural experiment to improve causal inferences from observational data. By analogy with the random assignment of treatment in randomized controlled trials, the random segregation of genetic alleles when DNA is transmitted from parents to offspring at gamete formation is expected to reduce confounding in genetic associations. Mendelian randomization analyses make a set of assumptions that must hold for valid results. Provided that the assumptions are well justified for the genetic variants that are employed as instrumental variables, MR studies can inform on whether a putative risk factor likely has a causal effect on the disease or not. Mendelian randomization has been increasingly applied over recent years to predict the efficacy and safety of existing and novel drugs targeting cardiovascular risk factors and to explore the repurposing potential of available drugs. This review article describes the principles of the MR design and some applications in cardiovascular epidemiology.
Collapse
Affiliation(s)
- Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Papworth Road, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
50
|
Zahn G, Baukmann HA, Wu J, Jordan J, Birkenfeld AL, Dirckx N, Schmidt MF. Targeting Longevity Gene SLC13A5: A Novel Approach to Prevent Age-Related Bone Fragility and Osteoporosis. Metabolites 2023; 13:1186. [PMID: 38132868 PMCID: PMC10744747 DOI: 10.3390/metabo13121186] [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: 11/09/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Reduced expression of the plasma membrane citrate transporter SLC13A5, also known as INDY, has been linked to increased longevity and mitigated age-related cardiovascular and metabolic diseases. Citrate, a vital component of the tricarboxylic acid cycle, constitutes 1-5% of bone weight, binding to mineral apatite surfaces. Our previous research highlighted osteoblasts' specialized metabolic pathway facilitated by SLC13A5 regulating citrate uptake, production, and deposition within bones. Disrupting this pathway impairs bone mineralization in young mice. New Mendelian randomization analysis using UK Biobank data indicated that SNPs linked to reduced SLC13A5 function lowered osteoporosis risk. Comparative studies of young (10 weeks) and middle-aged (52 weeks) osteocalcin-cre-driven osteoblast-specific Slc13a5 knockout mice (Slc13a5cKO) showed a sexual dimorphism: while middle-aged females exhibited improved elasticity, middle-aged males demonstrated enhanced bone strength due to reduced SLC13A5 function. These findings suggest reduced SLC13A5 function could attenuate age-related bone fragility, advocating for SLC13A5 inhibition as a potential osteoporosis treatment.
Collapse
Affiliation(s)
- Grit Zahn
- Eternygen GmbH, Westhafenstrasse 1, 13353 Berlin, Germany
| | | | - Jasmine Wu
- Department of Orthopaedics, School of Medicine, University of Maryland-Baltimore, Baltimore, MD 21201, USA
| | - Jens Jordan
- German Aerospace Center (DLR), Institute of Aerospace Medicine, 51147 Cologne, Germany;
| | - Andreas L. Birkenfeld
- Department of Diabetology Endocrinology and Nephrology, Internal Medicine IV, University Hospital Tübingen, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
- German Center for Diabetes Research (DZD), Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
- Department of Diabetes, Life Sciences and Medicine, Cardiovascular Medicine and Sciences, Kings College London, London WC2R 2LS, UK
| | - Naomi Dirckx
- Department of Orthopaedics, School of Medicine, University of Maryland-Baltimore, Baltimore, MD 21201, USA
| | - Marco F. Schmidt
- biotx.ai GmbH, Am Mühlenberg 11, 14476 Potsdam, Germany (M.F.S.)
| |
Collapse
|