151
|
Chen Y, Liu S, Gong W, Guo P, Xue F, Zhou X, Wang S, Yuan Z. Protein-centric omics integration analysis identifies candidate plasma proteins for multiple autoimmune diseases. Hum Genet 2024; 143:1035-1048. [PMID: 38143258 PMCID: PMC11485194 DOI: 10.1007/s00439-023-02627-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023]
Abstract
It remains challenging to translate the findings from genome-wide association studies (GWAS) of autoimmune diseases (AIDs) into interventional targets, presumably due to the lack of knowledge on how the GWAS risk variants contribute to AIDs. In addition, current immunomodulatory drugs for AIDs are broad in action rather than disease-specific. We performed a comprehensive protein-centric omics integration analysis to identify AIDs-associated plasma proteins through integrating protein quantitative trait loci datasets of plasma protein (1348 proteins and 7213 individuals) and totally ten large-scale GWAS summary statistics of AIDs under a cutting-edge systematic analytic framework. Specifically, we initially screened out the protein-AID associations using proteome-wide association study (PWAS), followed by enrichment analysis to reveal the underlying biological processes and pathways. Then, we performed both Mendelian randomization (MR) and colocalization analyses to further identify protein-AID pairs with putatively causal relationships. We finally prioritized the potential drug targets for AIDs. A total of 174 protein-AID associations were identified by PWAS. AIDs-associated plasma proteins were significantly enriched in immune-related biological process and pathways, such as inflammatory response (P = 3.96 × 10-10). MR analysis further identified 97 protein-AID pairs with potential causal relationships, among which 21 pairs were highly supported by colocalization analysis (PP.H4 > 0.75), 10 of 21 were the newly discovered pairs and not reported in previous GWAS analyses. Further explorations showed that four proteins (TLR3, FCGR2A, IL23R, TCN1) have corresponding drugs, and 17 proteins have druggability. These findings will help us to further understand the biological mechanism of AIDs and highlight the potential of these proteins to develop as therapeutic targets for AIDs.
Collapse
Affiliation(s)
- Yingxuan Chen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China
| | - Shuai Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China
| | - Weiming Gong
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China
| | - Ping Guo
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shukang Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China.
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan, 250003, Shandong, China.
| |
Collapse
|
152
|
Friligkou E, Løkhammer S, Cabrera-Mendoza B, Shen J, He J, Deiana G, Zanoaga MD, Asgel Z, Pilcher A, Di Lascio L, Makharashvili A, Koller D, Tylee DS, Pathak GA, Polimanti R. Gene discovery and biological insights into anxiety disorders from a large-scale multi-ancestry genome-wide association study. Nat Genet 2024; 56:2036-2045. [PMID: 39294497 DOI: 10.1038/s41588-024-01908-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/13/2024] [Indexed: 09/20/2024]
Abstract
We leveraged information from more than 1.2 million participants, including 97,383 cases, to investigate the genetics of anxiety disorders across five continental groups. Through ancestry-specific and cross-ancestry genome-wide association studies, we identified 51 anxiety-associated loci, 39 of which were novel. In addition, polygenic risk scores derived from individuals of European descent were associated with anxiety in African, admixed American and East Asian groups. The heritability of anxiety was enriched for genes expressed in the limbic system, cerebral cortex, cerebellum, metencephalon, entorhinal cortex and brain stem. Transcriptome-wide and proteome-wide analyses highlighted 115 genes associated with anxiety through brain-specific and cross-tissue regulation. Anxiety also showed global and local genetic correlations with depression, schizophrenia and bipolar disorder and widespread pleiotropy with several physical health domains. Overall, this study expands our knowledge regarding the genetic risk and pathogenesis of anxiety disorders, highlighting the importance of investigating diverse populations and integrating multi-omics information.
Collapse
Affiliation(s)
- Eleni Friligkou
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Solveig Løkhammer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Brenda Cabrera-Mendoza
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jie Shen
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Jun He
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Giovanni Deiana
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience, Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Mihaela Diana Zanoaga
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Zeynep Asgel
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Child and Adolescent Psychiatry, NYU Langone Health, New York Metropolitan Area, New York, NY, USA
| | - Abigail Pilcher
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Luciana Di Lascio
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- IRCCS Istituto Clinico Humanitas, Rozzano, Milan, Italy; Humanitas University, Pieve Emanuele, Milan, Italy
| | - Ana Makharashvili
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Dora Koller
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Daniel S Tylee
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Gita A Pathak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Renato Polimanti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA.
- Department of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
153
|
Bahrami S, Nordengen K, Rokicki J, Shadrin AA, Rahman Z, Smeland OB, Jaholkowski PP, Parker N, Parekh P, O'Connell KS, Elvsåshagen T, Toft M, Djurovic S, Dale AM, Westlye LT, Kaufmann T, Andreassen OA. The genetic landscape of basal ganglia and implications for common brain disorders. Nat Commun 2024; 15:8476. [PMID: 39353893 PMCID: PMC11445552 DOI: 10.1038/s41467-024-52583-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
The basal ganglia are subcortical brain structures involved in motor control, cognition, and emotion regulation. We conducted univariate and multivariate genome-wide association analyses (GWAS) to explore the genetic architecture of basal ganglia volumes using brain scans obtained from 34,794 Europeans with replication in 4,808 white and generalization in 5,220 non-white Europeans. Our multivariate GWAS identified 72 genetic loci associated with basal ganglia volumes with a replication rate of 55.6% at P < 0.05 and 87.5% showed the same direction, revealing a distributed genetic architecture across basal ganglia structures. Of these, 50 loci were novel, including exonic regions of APOE, NBR1 and HLAA. We examined the genetic overlap between basal ganglia volumes and several neurological and psychiatric disorders. The strongest genetic overlap was between basal ganglia and Parkinson's disease, as supported by robust LD-score regression-based genetic correlations. Mendelian randomization indicated genetic liability to larger striatal volume as potentially causal for Parkinson's disease, in addition to a suggestive causal effect of greater genetic liability to Alzheimer's disease on smaller accumbens. Functional analyses implicated neurogenesis, neuron differentiation and development in basal ganglia volumes. These results enhance our understanding of the genetic architecture and molecular associations of basal ganglia structure and their role in brain disorders.
Collapse
Grants
- R01 MH129742 NIMH NIH HHS
- Stiftelsen Kristian Gerhard Jebsen (Kristian Gerhard Jebsen Foundation)
- Norwegian Health Association (22731, 25598), the South-Eastern Norway Regional Health Authority (2013-123, 2017-112, 2019-108, 2014-097, 2015-073, 2016-083), the Research Council of Norway (276082, 323961. 213837, 223273, 248778, 273291, 262656, 229129, 283798, 311993, 324499. 204966, 249795, 273345).
Collapse
Affiliation(s)
- Shahram Bahrami
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway.
| | - Kaja Nordengen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Jaroslav Rokicki
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Alexey A Shadrin
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Zillur Rahman
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Olav B Smeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Nadine Parker
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pravesh Parekh
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Torbjørn Elvsåshagen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Department of Behavioral Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mathias Toft
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Lars T Westlye
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, Faculty of Social Sciences, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Ole A Andreassen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway.
- Department of Psychiatry, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
154
|
Feng Y, Shan L, Gong Y, Hang W, Sang Z, Sun Y, Tang K, Wang Y, Hu B, Xi X. Identification of ubiquitin markers for survival and prognosis of ovarian cancer. Heliyon 2024; 10:e37288. [PMID: 39315145 PMCID: PMC11417246 DOI: 10.1016/j.heliyon.2024.e37288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 08/11/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
Ovarian cancer (OC) is one of the most common malignancies and a leading cause of death among women worldwide. The ubiquitin pathway plays an important role in OC development. Using the single nucleotide polymorphism data obtained using the prevalence and dominance strategies, four ubiquitin marker genes were identified according to their expression levels: BARD1, BRCA2, FANCA, and BRCA1. Based on these four genes, a consensus clustering of OC expression data was performed. The significant differences in the survival analysis, ESTIMATE, and CIBERSORT results among the clusters indicated the pivotal role of these four genes in OC development. Of the ubiquitin-representative genes in each cluster, two ubiquitin genes, TOP2A and MYLIP, were identified in the survival risk model after univariate survival, Least Absolute Shrinkage and Selection Operator regression, and multivariate survival analyses. The reliability and robustness of both the training and validation data were confirmed by comparing the significant survival difference between high- and low-risk patients. We further explored the association between our risk model and clinical outcomes as well as predicted potentially interacting drugs. The co-expression network showed clear interactions among the four marker genes and two model genes and between high- and low-risk differentially expressed genes. Significantly enriched genes were found in pathways associated with ion channels, channel activity, and neuroactive ligand-receptor interactions. Therefore, suggesting the involvement of ubiquitin genes in the survival and development of OC through neurohumoral regulation. Our results will provide valuable reference or supplementary information for studies investigating OC diagnosis and therapies.
Collapse
Affiliation(s)
- Yiwen Feng
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Liyun Shan
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Yanping Gong
- Departments of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Wenzhao Hang
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Zhenyu Sang
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Yunyan Sun
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Kefu Tang
- Center for Reproductive Medicne, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200135, PR China
| | - Yulan Wang
- Departments of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Binjie Hu
- Departments of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Xiaowei Xi
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| |
Collapse
|
155
|
Kalaei Z, Shekarchi AA, Hojjat-Farsangi M, Jalali P, Jadidi-Niaragh F. The prognostic and therapeutic potential of vimentin in colorectal cancer. Mol Biol Rep 2024; 51:1027. [PMID: 39347868 DOI: 10.1007/s11033-024-09965-w] [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: 06/15/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
Several cells and molecules in the tumor microenvironment have been introduced as effective factors in the prognosis and progression of colorectal cancer. As a key element of the intermediate filament family, vimentin is expressed by mesenchymal cells in a ubiquitous manner and contributes significantly to cellular integrity and stress resistance in colorectal cancer. Recent studies have shown that alterations in the expression patterns of intermediate filaments are significantly related to cancer progression, especially in phenotypes associated with cellular migration and invasion. In addition to its multiple biological roles, vimentin also has a substantial function in mediating the epithelial-mesenchymal transition. Therefore, evaluating vimentin as an effective factor involved in the prognosis of colorectal cancer and targeting it as a novel approach to cancer therapy have become one of the main goals of many researchers worldwide. In this article, we will review the various biological functions of vimentin, as well as its relationship with colorectal cancer with the aim of providing novel insights into its clinical importance in the prognosis and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Zahra Kalaei
- Department of Biology, Faculty of Natural Sciences, Tabriz University, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
156
|
Mo Q, Liu X, Gong W, Wang Y, Yuan Z, Sun X, Wang S. Pinpointing Novel Plasma and Brain Proteins for Common Ocular Diseases: A Comprehensive Cross-Omics Integration Analysis. Int J Mol Sci 2024; 25:10236. [PMID: 39408566 PMCID: PMC11476976 DOI: 10.3390/ijms251910236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The pathogenesis of ocular diseases (ODs) remains unclear, although genome-wide association studies (GWAS) have identified numerous associated genetic risk loci. We integrated protein quantitative trait loci (pQTL) datasets and five large-scale GWAS summary statistics of ODs under a cutting-edge systematic analytic framework. Proteome-wide association studies (PWAS) identified plasma and brain proteins associated with ODs, and 11 plasma proteins were identified by Mendelian randomization (MR) and colocalization (COLOC) analyses as being potentially causally associated with ODs. Five of these proteins (protein-coding genes ECI1, LCT, and NPTXR for glaucoma, WARS1 for age-related macular degeneration (AMD), and SIGLEC14 for diabetic retinopathy (DR)) are newly reported. Twenty brain-protein-OD pairs were identified by COLOC analysis. Eight pairs (protein-coding genes TOM1L2, MXRA7, RHPN2, and HINT1 for senile cataract, WARS1 and TDRD7 for AMD, STAT6 for myopia, and TPPP3 for DR) are newly reported in this study. Phenotype-disease mapping analysis revealed 10 genes related to the eye/vision phenotype or ODs. Combined with a drug exploration analysis, we found that the drugs related to C3 and TXN have been used for the treatment of ODs, and another eight genes (GSTM3 for senile cataract, IGFBP7 and CFHR1 for AMD, PTPMT1 for glaucoma, EFEMP1 and ACP1 for myopia, SIRPG and CTSH for DR) are promising targets for pharmacological interventions. Our study highlights the role played by proteins in ODs, in which brain proteins were taken into account due to the deepening of eye-brain connection studies. The potential pathogenic proteins finally identified provide a more reliable reference range for subsequent medical studies.
Collapse
Affiliation(s)
- Qinyou Mo
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Xinyu Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Weiming Gong
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Yunzhuang Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Xiubin Sun
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Shukang Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Road, Jinan 250012, China; (Q.M.); (X.L.); (W.G.); (Y.W.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| |
Collapse
|
157
|
Mazein I, Rougny A, Mazein A, Henkel R, Gütebier L, Michaelis L, Ostaszewski M, Schneider R, Satagopam V, Jensen LJ, Waltemath D, Wodke JAH, Balaur I. Graph databases in systems biology: a systematic review. Brief Bioinform 2024; 25:bbae561. [PMID: 39565895 PMCID: PMC11578065 DOI: 10.1093/bib/bbae561] [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: 04/18/2024] [Revised: 09/28/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Graph databases are becoming increasingly popular across scientific disciplines, being highly suitable for storing and connecting complex heterogeneous data. In systems biology, they are used as a backend solution for biological data repositories, ontologies, networks, pathways, and knowledge graph databases. In this review, we analyse all publications using or mentioning graph databases retrieved from PubMed and PubMed Central full-text search, focusing on the top 16 available graph databases, Publications are categorized according to their domain and application, focusing on pathway and network biology and relevant ontologies and tools. We detail different approaches and highlight the advantages of outstanding resources, such as UniProtKB, Disease Ontology, and Reactome, which provide graph-based solutions. We discuss ongoing efforts of the systems biology community to standardize and harmonize knowledge graph creation and the maintenance of integrated resources. Outlining prospects, including the use of graph databases as a way of communication between biological data repositories, we conclude that efficient design, querying, and maintenance of graph databases will be key for knowledge generation in systems biology and other research fields with heterogeneous data.
Collapse
Affiliation(s)
- Ilya Mazein
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Adrien Rougny
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Alexander Mazein
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Ron Henkel
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Lea Gütebier
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Lea Michaelis
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Venkata Satagopam
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Lars Juhl Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870 Frederiksberg C, Denmark
| | - Dagmar Waltemath
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Judith A H Wodke
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Irina Balaur
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| |
Collapse
|
158
|
Zhang Y, Li L, Liu Y, Zhang W, Peng W, Zhang S, Qu R, Ma Y, Liu Z, Ge Z, Zhou Y, Tian W, Shen Y, Liu L, Duan J, Chen Z, Zhu L. Identification of CCL20 as a Prognostic Predictor for Severe Fever With Thrombocytopenia Syndrome Based on Plasma Proteomics. J Infect Dis 2024; 230:741-753. [PMID: 38271258 DOI: 10.1093/infdis/jiae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Severe fever with thrombocytopenia syndrome (SFTS), a lethal tick-borne hemorrhagic fever, prompted our investigation into prognostic predictors and potential drug targets using plasma Olink Proteomics. METHODS Employing the Olink assay, we analyzed 184 plasma proteins in 30 survivors and 8 nonsurvivors of SFTS. Validation was performed in a cohort of 154 patients with SFTS via enzyme-linked immunosorbent assay. We utilized the Drug-Gene Interaction Database to identify protein-drug interactions. RESULTS Nonsurvivors exhibited 110 differentially expressed proteins as compared with survivors, with functional enrichment in the cell chemotaxis-related pathway. Thirteen differentially expressed proteins-including C-C motif chemokine 20 (CCL20), calcitonin gene-related peptide alpha, and pleiotrophin-were associated with multiple-organ dysfunction syndrome. CCL20 emerged as the top predictor of death, demonstrating an area under the curve of 1 (P = .0004) and 0.9033 (P < .0001) in the discovery and validation cohorts, respectively. Patients with CCL20 levels exceeding 45.74 pg/mL exhibited a fatality rate of 45.65%, while no deaths occurred in those with lower CCL20 levels. Furthermore, we identified 202 Food and Drug Administration-approved drugs targeting 37 death-related plasma proteins. CONCLUSIONS Distinct plasma proteomic profiles characterize SFTS cases with different outcomes, with CCL20 emerging as a novel, sensitive, accurate, and specific biomarker for predicting SFTS prognosis.
Collapse
Affiliation(s)
- Yue Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Lan Li
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuanni Liu
- Department of Infectious Diseases, Yantai City Hospital for Infectious Disease, Yantai, China
| | - Wei Zhang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wenjuan Peng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shuai Zhang
- Department of Clinical Laboratory, Yantai City Hospital for Infectious Disease, Yantai, China
| | - Renliang Qu
- Department of Clinical Laboratory, Yantai City Hospital for Infectious Disease, Yantai, China
| | - Yuan Ma
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zishuai Liu
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziruo Ge
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanxi Zhou
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wen Tian
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yi Shen
- Department of Infectious Diseases, Dandong Infectious Disease Hospital, Dandong, China
| | - Li Liu
- Department of Infectious Diseases, Taian City Central Hospital, Taian, China
| | - Jianping Duan
- Department of Hepatology, Qing Dao No. 6 People's Hospital, Qingdao, China
| | - Zhihai Chen
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liuluan Zhu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
159
|
Engesser J, Khatri R, Schaub DP, Zhao Y, Paust HJ, Sultana Z, Asada N, Riedel JH, Sivayoganathan V, Peters A, Kaffke A, Jauch-Speer SL, Goldbeck-Strieder T, Puelles VG, Wenzel UO, Steinmetz OM, Hoxha E, Turner JE, Mittrücker HW, Wiech T, Huber TB, Bonn S, Krebs CF, Panzer U. Immune profiling-based targeting of pathogenic T cells with ustekinumab in ANCA-associated glomerulonephritis. Nat Commun 2024; 15:8220. [PMID: 39300109 DOI: 10.1038/s41467-024-52525-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis is a life-threatening autoimmune disease that often results in kidney failure caused by crescentic glomerulonephritis (GN). To date, treatment of most patients with ANCA-GN relies on non-specific immunosuppressive agents, which may have serious adverse effects and be only partially effective. Here, using spatial and single-cell transcriptome analysis, we characterize inflammatory niches in kidney samples from 34 patients with ANCA-GN and identify proinflammatory, cytokine-producing CD4+ and CD8+ T cells as a pathogenic signature. We then utilize these transcriptomic profiles for digital pharmacology and identify ustekinumab, a monoclonal antibody targeting IL-12 and IL-23, as the strongest therapeutic drug to use. Moreover, four patients with relapsing ANCA-GN are treated with ustekinumab in combination with low-dose cyclophosphamide and steroids, with ustekinumab given subcutaneously (90 mg) at weeks 0, 4, 12, and 24. Patients are followed up for 26 weeks to find this treatment well-tolerated and inducing clinical responses, including improved kidney function and Birmingham Vasculitis Activity Score, in all ANCA-GN patients. Our findings thus suggest that targeting of pathogenic T cells in ANCA-GN patients with ustekinumab might represent a potential approach and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Jonas Engesser
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robin Khatri
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Darius P Schaub
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Yu Zhao
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Hans-Joachim Paust
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zeba Sultana
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Nariaki Asada
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Hendrik Riedel
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Varshi Sivayoganathan
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anett Peters
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Kaffke
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Victor G Puelles
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich O Wenzel
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver M Steinmetz
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elion Hoxha
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Eric Turner
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology, Division of Nephropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Bonn
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Christian F Krebs
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Ulf Panzer
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
160
|
Kamble P, Nagar PR, Bhakhar KA, Garg P, Sobhia ME, Naidu S, Bharatam PV. Cancer pharmacoinformatics: Databases and analytical tools. Funct Integr Genomics 2024; 24:166. [PMID: 39294509 DOI: 10.1007/s10142-024-01445-5] [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: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cancer is a subject of extensive investigation, and the utilization of omics technology has resulted in the generation of substantial volumes of big data in cancer research. Numerous databases are being developed to manage and organize this data effectively. These databases encompass various domains such as genomics, transcriptomics, proteomics, metabolomics, immunology, and drug discovery. The application of computational tools into various core components of pharmaceutical sciences constitutes "Pharmacoinformatics", an emerging paradigm in rational drug discovery. The three major features of pharmacoinformatics include (i) Structure modelling of putative drugs and targets, (ii) Compilation of databases and analysis using statistical approaches, and (iii) Employing artificial intelligence/machine learning algorithms for the discovery of novel therapeutic molecules. The development, updating, and analysis of databases using statistical approaches play a pivotal role in pharmacoinformatics. Multiple software tools are associated with oncoinformatics research. This review catalogs the databases and computational tools related to cancer drug discovery and highlights their potential implications in the pharmacoinformatics of cancer.
Collapse
Affiliation(s)
- Pradnya Kamble
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prinsa R Nagar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Kaushikkumar A Bhakhar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Srivatsava Naidu
- Center of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Prasad V Bharatam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
161
|
Gu Y, Jiang L, Shui M, Luo H, Zhou X, Zhang S, Jiang C, Huang J, Chen H, Tang J, Fu Y, Luo H, Yang G, Xu K, Chi H, Liu J, Huang S. Revealing the association between East Asian oral microbiome and colorectal cancer through Mendelian randomization and multi-omics analysis. Front Cell Infect Microbiol 2024; 14:1452392. [PMID: 39355266 PMCID: PMC11443854 DOI: 10.3389/fcimb.2024.1452392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/14/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) poses a global health threat, with the oral microbiome increasingly implicated in its pathogenesis. This study leverages Mendelian Randomization (MR) to explore causal links between oral microbiota and CRC using data from the China National GeneBank and Biobank Japan. By integrating multi-omics approaches, we aim to uncover mechanisms by which the microbiome influences cellular metabolism and cancer development. METHODS We analyzed microbiome profiles from 2017 tongue and 1915 saliva samples, and GWAS data for 6692 CRC cases and 27178 controls. Significant bacterial taxa were identified via MR analysis. Single-cell RNA sequencing and enrichment analyses elucidated underlying pathways, and drug predictions identified potential therapeutics. RESULTS MR identified 19 bacterial taxa significantly associated with CRC. Protective effects were observed in taxa like RUG343 and Streptococcus_umgs_2425, while HOT-345_umgs_976 and W5053_sp000467935_mgs_712 increased CRC risk. Single-cell RNA sequencing revealed key pathways, including JAK-STAT signaling and tyrosine metabolism. Drug prediction highlighted potential therapeutics like Menadione Sodium Bisulfite and Raloxifene. CONCLUSION This study establishes the critical role of the oral microbiome in colorectal cancer development, identifying specific microbial taxa linked to CRC risk. Single-cell RNA sequencing and drug prediction analyses further elucidate key pathways and potential therapeutics, providing novel insights and personalized treatment strategies for CRC.
Collapse
Affiliation(s)
- Yuheng Gu
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Min Shui
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Honghao Luo
- Department of Radiology, Xichong People’s Hospital, Nanchong, China
| | - Xuancheng Zhou
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Shengke Zhang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chenglu Jiang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jinbang Huang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Haiqing Chen
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jingyi Tang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yiping Fu
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Hao Chi
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jie Liu
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
- Department of general surgery, Dazhou Central Hospital, Dazhou, China
| | - Shangke Huang
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
162
|
Koromina M, Ravi A, Panagiotaropoulou G, Schilder BM, Humphrey J, Braun A, Bidgeli T, Chatzinakos C, Coombes B, Kim J, Liu X, Terao C, O.’Connell KS, Adams M, Rolf A, Alda M, Alfredsson L, Andlauer TFM, Andreassen OA, Antoniou A, Baune BT, Bengesser S, Biernacka J, Boehnke M, Bosch R, Cairns MJ, Carr VJ, Casas M, Catts S, Cichon S, Corvin A, Craddock N, Dafnas K, Dalkner N, Dannlowski U, Degenhardt F, Florio AD, Dikeos D, Fellendorf FT, Ferentinos P, Forstner AJ, Forty L, Frye M, Fullerton JM, Gawlik M, Gizer IR, Gordon-Smith K, Green MJ, Grigoroiu-Serbanescu M, Guzman-Parra J, Hahn T, Henskens F, Hillert J, Jablensky AV, Jones L, Jones I, Jonsson L, Kelsoe JR, Kircher T, Kirov G, Kittel-Schneider S, Kogevinas M, Landén M, Leboyer M, Lenger M, Lissowska J, Lochner C, Loughland C, MacIntyre D, Martin NG, Maratou E, Mathews CA, Mayoral F, McElroy SL, McGregor NW, McIntosh A, McQuillin A, Michie P, Mitchell PB, Moutsatsou P, Mowry B, Müller-Myhsok B, Myers RM, Nenadić I, Nievergelt C, Nöthen MM, Nurnberger J, O.’Donovan M, O’Donovan C, Ophoff RA, Owen MJ, Pantelis C, Pato C, Pato MT, Patrinos GP, Pawlak JM, Perlis RH, Porichi E, Posthuma D, Ramos-Quiroga JA, et alKoromina M, Ravi A, Panagiotaropoulou G, Schilder BM, Humphrey J, Braun A, Bidgeli T, Chatzinakos C, Coombes B, Kim J, Liu X, Terao C, O.’Connell KS, Adams M, Rolf A, Alda M, Alfredsson L, Andlauer TFM, Andreassen OA, Antoniou A, Baune BT, Bengesser S, Biernacka J, Boehnke M, Bosch R, Cairns MJ, Carr VJ, Casas M, Catts S, Cichon S, Corvin A, Craddock N, Dafnas K, Dalkner N, Dannlowski U, Degenhardt F, Florio AD, Dikeos D, Fellendorf FT, Ferentinos P, Forstner AJ, Forty L, Frye M, Fullerton JM, Gawlik M, Gizer IR, Gordon-Smith K, Green MJ, Grigoroiu-Serbanescu M, Guzman-Parra J, Hahn T, Henskens F, Hillert J, Jablensky AV, Jones L, Jones I, Jonsson L, Kelsoe JR, Kircher T, Kirov G, Kittel-Schneider S, Kogevinas M, Landén M, Leboyer M, Lenger M, Lissowska J, Lochner C, Loughland C, MacIntyre D, Martin NG, Maratou E, Mathews CA, Mayoral F, McElroy SL, McGregor NW, McIntosh A, McQuillin A, Michie P, Mitchell PB, Moutsatsou P, Mowry B, Müller-Myhsok B, Myers RM, Nenadić I, Nievergelt C, Nöthen MM, Nurnberger J, O.’Donovan M, O’Donovan C, Ophoff RA, Owen MJ, Pantelis C, Pato C, Pato MT, Patrinos GP, Pawlak JM, Perlis RH, Porichi E, Posthuma D, Ramos-Quiroga JA, Reif A, Reininghaus EZ, Ribasés M, Rietschel M, Schall U, Schofield PR, Schulze TG, Scott L, Scott RJ, Serretti A, Weickert CS, Smoller JW, Artigas MS, Stein DJ, Streit F, Toma C, Tooney P, Vawter MP, Vieta E, Vincent JB, Waldman ID, Weickert T, Witt SH, Hong KS, Ikeda M, Iwata N, Świątkowska B, Won HH, Edenberg HJ, Ripke S, Raj T, Coleman JRI, Mullins N. Fine-mapping genomic loci refines bipolar disorder risk genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.12.24302716. [PMID: 38405768 PMCID: PMC10889003 DOI: 10.1101/2024.02.12.24302716] [Show More Authors] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Bipolar disorder (BD) is a heritable mental illness with complex etiology. While the largest published genome-wide association study identified 64 BD risk loci, the causal SNPs and genes within these loci remain unknown. We applied a suite of statistical and functional fine-mapping methods to these loci, and prioritized 17 likely causal SNPs for BD. We mapped these SNPs to genes, and investigated their likely functional consequences by integrating variant annotations, brain cell-type epigenomic annotations, brain quantitative trait loci, and results from rare variant exome sequencing in BD. Convergent lines of evidence supported the roles of genes involved in neurotransmission and neurodevelopment including SCN2A, TRANK1, DCLK3, INSYN2B, SYNE1, THSD7A, CACNA1B, TUBBP5, PLCB3, PRDX5, KCNK4, CRTC3, AP001453.3, TRPT1, FKBP2, DNAJC4, RASGRP1, FURIN, FES, DPH1, GSDMB, MED24 and THRA in BD. These represent promising candidates for functional experiments to understand biological mechanisms and therapeutic potential. Additionally, we demonstrated that fine-mapping effect sizes can improve performance of BD polygenic risk scores across diverse populations, and present a high-throughput fine-mapping pipeline (https://github.com/mkoromina/SAFFARI).
Collapse
Affiliation(s)
- Maria Koromina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashvin Ravi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Brian M. Schilder
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Humphrey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | | | | | - Brandon Coombes
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jaeyoung Kim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kevin S. O.’Connell
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- NORMENT, University of Oslo, Oslo, Norway
| | - Mark Adams
- Division of Psychiatry, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Adolfsson Rolf
- Department of Clinical Sciences, Psychiatry, Umeå, University Medical Faculty, Umeå, Sweden
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- National Institute of Mental Health, Klecany, Czech Republic
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Till F. M. Andlauer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Ole A. Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- NORMENT, University of Oslo, Oslo, Norway
| | - Anastasia Antoniou
- National Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | - Bernhard T. Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Susanne Bengesser
- Medical University of Graz, Division of Psychiatry and Psychotherapeutic Medicine, Graz, Austria
| | - Joanna Biernacka
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Michael Boehnke
- Center for Statistical Genetics and Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Rosa Bosch
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Programa SJD MIND Escoles, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | | | - Vaughan J. Carr
- School of Clinical Medicine, Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, NSW, Australia
| | - Miquel Casas
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Programa SJD MIND Escoles, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | | | - Sven Cichon
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Dept of Psychiatry and Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Nicholas Craddock
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Konstantinos Dafnas
- National Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | - Nina Dalkner
- Medical University of Graz, Division of Psychiatry and Psychotherapeutic Medicine, Graz, Austria
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Arianna Di Florio
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Department of Psychiatry, University of North Caroli at Chapel Hill, Chapel Hill, NC, USA
| | - Dimitris Dikeos
- National Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | | | - Panagiotis Ferentinos
- National Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
- Social, Genetic and Developmental Psychiatry Centre, King’s College London, London, UK
| | - Andreas J. Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
| | - Liz Forty
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Mark Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Janice M. Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Micha Gawlik
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| | - Ian R. Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | | | - Melissa J. Green
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Maria Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania
| | - José Guzman-Parra
- Mental Health Department, University Regional Hospital, Biomedicine Institute (IBIMA), Málaga, Spain
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | | | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Lisa Jones
- Psychological Medicine, University of Worcester, Worcester, UK
| | - Ian Jones
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Lina Jonsson
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - John R. Kelsoe
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Germany
| | - George Kirov
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | | | - Mikael Landén
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Marion Leboyer
- University of Paris Est Créteil, INSERM, IMRB, Translatiol Neuropsychiatry, Créteil, France
- Department of Psychiatry and Addiction Medicine, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Melanie Lenger
- Medical University of Graz, Division of Psychiatry and Psychotherapeutic Medicine, Graz, Austria
| | - Jolanta Lissowska
- Cancer Epidemiology and Prevention, M. Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Christine Lochner
- SA MRC Unit on Risk and Resilience in Mental Disorders, Dept of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | | | - Donald MacIntyre
- Division of Psychiatry, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Nicholas G. Martin
- Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| | - Eirini Maratou
- National and Kapodistrian University of Athens, Medical School, Clinical Biochemistry Laboratory, Attikon General Hospital, Athens, Greece
| | - Carol A. Mathews
- Department of Psychiatry and Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Fermin Mayoral
- Mental Health Department, University Regional Hospital, Biomedicine Institute (IBIMA), Málaga, Spain
| | | | - Nathaniel W. McGregor
- Systems Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Andrew McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | | | - Philip B. Mitchell
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Paraskevi Moutsatsou
- National Kapodistrian University of Athens, Medical School, Clinical Biochemistry Laboratory, Attikon General Hospital, Athens, Greece
| | - Bryan Mowry
- University of Queensland, Brisbane, QLD, Australia
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Caroline Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Research/Psychiatry, Veterans Affairs San, Diego Healthcare System, San Diego, CA, USA
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - John Nurnberger
- Stark Neurosciences Research Institute, Indiana University School of Medicine
- Departments of Psychiatry and Medical and Molecular Genetics, Indiana University School of Medicine
- Indiana University School of Medicine
| | - Michael O.’Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Claire O’Donovan
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Roel A. Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Science, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael J. Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | | - Carlos Pato
- Institute for Genomic Health, SUNY Downstate Medical Center College of Medicine, Brooklyn, NY, USA
| | - Michele T. Pato
- Institute for Genomic Health, SUNY Downstate Medical Center College of Medicine, Brooklyn, NY, USA
| | - George P. Patrinos
- University of Patras, School of Health Sciences, Department of Pharmacy, Laboratory of Pharmacogenomics and Individualized Therapy, Patras, Greece
- United Arab Emirates University, College of Medicine and Health Sciences, Department of Genetics and Genomics, Al-Ain, United Arab Emirates
- United Arab Emirates University, Zayed Center for Health Sciences, Al-Ain, United Arab Emirates
- Erasmus University Medical Center, Faculty of Medicina and Health Sciences, Department of Pathology, Clinical Bioinformatics Unit, Rotterdam, The Netherlands
| | - Joanna M. Pawlak
- Department of Psychiatry, Departmet of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Roy H. Perlis
- Psychiatry, Harvard Medical School, Boston, MA, USA
- Division of Clinical Research, Massachusetts General Hospital, Boston, MA, USA
| | - Evgenia Porichi
- tiol and Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Josep Antoni Ramos-Quiroga
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d´Hebron, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelo, Barcelo, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d´Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Eva Z. Reininghaus
- Medical University of Graz, Division of Psychiatry and Psychotherapeutic Medicine, Graz, Austria
| | - Marta Ribasés
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d´Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d´Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain. Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Thomas G. Schulze
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Laura Scott
- Center for Statistical Genetics and Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | | | - Alessandro Serretti
- Department of Medicine and Surgery, Kore University of Enna, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| | - Cynthia Shannon Weickert
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jordan W. Smoller
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital, Boston, MA, USA
| | - Maria Soler Artigas
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d´Hebron, Barcelo, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d´Hebron Research Institut (VHIR), Universitat Autònoma de Barcelo, Barcelo, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelo, Barcelo, Spain. Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelo, Barcelo, Catalonia, Spain
| | - Dan J. Stein
- SAMRC Unit on Risk and Resilience in Mental Disorders, Dept of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Claudio Toma
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid and CSIC, Madrid, Spain
| | - Paul Tooney
- University of Newcastle, Newcastle, NSW, Australia
| | - Marquis P. Vawter
- Functional Genomics Laboratory, School of Medicine, University of California, Irvine Canada
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine Canada
| | - Eduard Vieta
- Clinical Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Spain
| | - John B. Vincent
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | - Thomas Weickert
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kyung Sue Hong
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Beata Świątkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Howard J. Edenberg
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan R. I. Coleman
- Social, Genetic and Developmental Psychiatry Centre, King’s College London, London, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
| | - Niamh Mullins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
163
|
Song J, Yang H. Identifying new biomarkers and potential therapeutic targets for breast cancer through the integration of human plasma proteomics: a Mendelian randomization study and colocalization analysis. Front Endocrinol (Lausanne) 2024; 15:1449668. [PMID: 39351539 PMCID: PMC11439655 DOI: 10.3389/fendo.2024.1449668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Background The proteome is a crucial reservoir of targets for cancer treatment. While some targeted therapies have been developed, there are still significant challenges in early diagnosis and treatment, highlighting the need to identify new biomarkers and therapeutic targets for breast cancer. Therefore, we conducted a comprehensive proteome-wide Mendelian randomization (MR) study to identify novel biomarkers and potential therapeutic targets for breast cancer. Methods Protein quantitative trait locus (pQTL) data were extracted from two published plasma proteome-wide association studies. Genetic variants associated with breast cancer were obtained from the Breast Cancer Association Consortium, which included 133,384 cases and 113,789 controls, and the Finnish cohort study, comprising 18,786 cases and 182,927 controls. We employed summary-based MR and colocalization methods to identify potential drug targets for breast cancer, which were subsequently validated using a two-sample MR approach. Finally, a protein-protein interaction (PPI) network was constructed to detect interactions between the identified proteins and existing cancer drug targets. Results Gene-predicted levels of ten proteins were associated with breast cancer risk. Decreased levels of CASP8, DDX58, CPNE1, ULK3, PARK7, and BTN2A1, as well as increased levels of TNFRSF9, TNXB, DNPH1, and TLR1, were linked to an elevated risk of breast cancer. Among these, CASP8 and DDX58 were supported by tier-one evidence, while CPNE1, ULK3, PARK7, and TNFRSF9 received tier-two evidence support. The remaining proteins, TNXB, BTN2A1, DNPH1, and TLR1, were supported by tier-three evidence. CASP8, DDX58, CPNE1, ULK3, PARK7, and TNFRSF9 have already been identified as targets in drug development and potential therapeutic targets for breast cancer treatment. Additionally, ULK3 showed promise as a prognostic biomarker for breast cancer. Conclusions The present study identified several novel potential drug targets and biomarkers for breast cancer, providing new insights into its diagnosis and treatment. The integration of PPI and druggability evaluations enhances the prioritization of these therapeutic targets, paving the way for future drug development efforts.
Collapse
Affiliation(s)
- Jingshuang Song
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Huawei Yang
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University, Nanning, China
| |
Collapse
|
164
|
Słyk Ż, Stachowiak N, Małecki M. Gene Therapy in the Light of Lifestyle Diseases: Budesonide, Acetaminophen and Simvastatin Modulates rAAV Transduction Efficiency. Pharmaceuticals (Basel) 2024; 17:1213. [PMID: 39338375 PMCID: PMC11434873 DOI: 10.3390/ph17091213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Recombinant AAV (rAAV) vectors are increasingly favored for gene therapy due to their useful features of vectorology, such as transfection of dividing and nondividing cells, the presence of tissue-specific serotypes, and biosafety considerations. This study investigates the impact of commonly used therapeutic drugs-acetaminophen, budesonide, and simvastatin-on rAAV transduction efficiency in HEK-293 cells. Cells were transduced with an AAV mosaic vector under the control of a cytomegalovirus (CMV) promoter encoding green fluorescent protein (GFP). Transduction efficiency was assessed by qPCR and fluorescent microscopy. Analysis of functional interactions between genes potentially involved in rAAV transduction in drug-exposed cells was also performed. This study showed a clear effect of drugs on rAAV transmission. Notably, acetaminophen enhanced transduction efficiency by 9-fold, while budesonide and simvastatin showed 2-fold and 3-fold increases, respectively. The gene analysis illustrates the possible involvement of genes related to cell membranes in the potentiation of rAAV transduction induced by the drugs under investigation. Attention should be paid to S100A8, which is a common drug-modified gene for drugs showing anti-inflammatory effects (budesonide and simvastatin), demonstrating an interaction with the gene encoding the receptor for AAV (HGFR). This study underscores the significance of assessing rAAV pharmacokinetics/pharmacodynamics (PKs/PDs) and drug-gene therapy interactions in optimizing gene therapy protocols.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Natalia Stachowiak
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
165
|
Chakraborty C, Bhattacharya M, Lee SS, Wen ZH, Lo YH. The changing scenario of drug discovery using AI to deep learning: Recent advancement, success stories, collaborations, and challenges. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102295. [PMID: 39257717 PMCID: PMC11386122 DOI: 10.1016/j.omtn.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Due to the transformation of artificial intelligence (AI) tools and technologies, AI-driven drug discovery has come to the forefront. It reduces the time and expenditure. Due to these advantages, pharmaceutical industries are concentrating on AI-driven drug discovery. Several drug molecules have been discovered using AI-based techniques and tools, and several newly AI-discovered drug molecules have already entered clinical trials. In this review, we first present the data and their resources in the pharmaceutical sector for AI-driven drug discovery and illustrated some significant algorithms or techniques used for AI and ML which are used in this field. We gave an overview of the deep neural network (NN) models and compared them with artificial NNs. Then, we illustrate the recent advancement of the landscape of drug discovery using AI to deep learning, such as the identification of drug targets, prediction of their structure, estimation of drug-target interaction, estimation of drug-target binding affinity, design of de novo drug, prediction of drug toxicity, estimation of absorption, distribution, metabolism, excretion, toxicity; and estimation of drug-drug interaction. Moreover, we highlighted the success stories of AI-driven drug discovery and discussed several collaboration and the challenges in this area. The discussions in the article will enrich the pharmaceutical industry.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-Do 24252, Republic of Korea
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 813204, Taiwan
- Shu-Zen Junior College of Medicine and Management, Kaohsiung 821004, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| |
Collapse
|
166
|
Gong Y, Wang J, Pan M, Zhao Y, Zhang H, Zhang F, Liu J, Yang J, Hu J. Harmine inhibits pulmonary fibrosis through regulating DNA damage repair-related genes and activation of TP53-Gadd45α pathway. Int Immunopharmacol 2024; 138:112542. [PMID: 38924867 DOI: 10.1016/j.intimp.2024.112542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Harmine has many pharmacological activities and has been found to significantly inhibit the fibrosis of keloid fibroblasts. DNA damage repair (DDR) is essential to prevent fibrosis. This study aimed to investigate the effects of harmine on pulmonary fibrosis and its underlying mechanisms. METHODS Bleomycin and TGF-β1 were used to construct pulmonary fibrosis models in vivo and in vitro, then treated with harmine to explore harmine's effects in treating experimental pulmonary fibrosis and its related mechanisms. Then, RNA sequencing was applied to investigate further the crucial DDR-related genes and drug targets of harmine against pulmonary fibrosis. Finally, the expression levels of DDR-related genes were verified by real-time quantitative PCR (RT-qPCR) and western blot. RESULTS Our in vivo experiments showed that harmine treatment could improve weight loss and lung function and reduce tissue fibrosis in mice with pulmonary fibrosis. The results confirmed that harmine could inhibit the viability and migration of TGF-β1-induced MRC-5 cells, induce their apoptosis, and suppress the F-actin expression, suggesting that harmine could suppress the phenotypic transition from lung fibroblasts to lung myoblasts. In addition, RNA sequencing identified 1692 differential expressed genes (DEGs), and 10 DDR-related genes were screened as critical DDR-related genes. RT-qPCR and western blotting showed that harmine could down-regulate the expression of CHEK1, ERCC1, ERCC4, POLD1, RAD51, RPA1, TOP1, and TP53, while up-regulate FEN1, H2AX and GADD45α expression. CONCLUSIONS Harmine may inhibit pulmonary fibrosis by regulating DDR-related genes and activating the TP53-Gadd45α pathway.
Collapse
Affiliation(s)
- Yuehong Gong
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China; Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, Xinjiang 830011, China
| | - Jie Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China; Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, Xinjiang 830011, China
| | - Meichi Pan
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China
| | - Yicong Zhao
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China
| | - Haibo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China; Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, Xinjiang 830011, China
| | - Fei Zhang
- Department of Medicine, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China
| | - Jiangyun Liu
- Soochow Univ, College of Pharmaceutic Science, Suzhou 215123, China
| | - Jianhua Yang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China; Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, Xinjiang 830011, China.
| | - Junping Hu
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China.
| |
Collapse
|
167
|
Fathi M, Zarei A, Moghimi A, Jalali P, Salehi Z, Gholamin S, Jadidi-Niaragh F. Combined cancer immunotherapy based on targeting adenosine pathway and PD-1/PDL-1 axis. Expert Opin Ther Targets 2024; 28:757-777. [PMID: 39305018 DOI: 10.1080/14728222.2024.2405090] [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: 01/27/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Cancer immunotherapy has revolutionized the field of oncology, offering new hope to patients with advanced malignancies. Tumor-induced immunosuppression limits the effectiveness of current immunotherapeutic strategies, such as PD-1/PDL-1 checkpoint inhibitors. Adenosine, a purine nucleoside molecule, is crucial to this immunosuppression because it stops T cells from activating and helps regulatory T cells grow. Targeting the adenosine pathway and blocking PD-1/PDL-1 is a potential way to boost the immune system's response to tumors. AREAS COVERED This review discusses the current understanding of the adenosine pathway in tumor immunology and the preclinical and clinical data supporting the combination of adenosine pathway inhibitors with PD-1/PDL-1 blockade. We also discuss the challenges and future directions for developing combination immunotherapy targeting the adenosine pathway and the PD-1/PDL-1 axis for cancer treatment. EXPERT OPINION The fact that the adenosine signaling pathway controls many immune system processes suggests that it has a wide range of therapeutic uses. Within the next five years, there will be tremendous progress in this area, and the standard of care for treating malignant tumors will have switched from point-to-point therapy to the integration of immunological networks comprised of multiple signaling pathways, like the adenosine axis.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ata Moghimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sharareh Gholamin
- City of Hope Beckman Research Institute and Medical Center, Duarte, CA, USA
- City of Hope Department of Radiation Oncology, Duarte, CA, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
168
|
Jiang D, Nan H, Chen Z, Zou WQ, Wu L. Genetic insights into drug targets for sporadic Creutzfeldt-Jakob disease: Integrative multi-omics analysis. Neurobiol Dis 2024; 199:106599. [PMID: 38996988 DOI: 10.1016/j.nbd.2024.106599] [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: 04/18/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
OBJECTIVE Sporadic Creutzfeldt-Jakob disease (sCJD) is a fatal rapidly progressive neurodegenerative disorder with no effective therapeutic interventions. We aimed to identify potential genetically-supported drug targets for sCJD by integrating multi-omics data. METHODS Multi-omics-wide association studies, Mendelian randomization, and colocalization analyses were employed to explore potential therapeutic targets using expression, single-cell expression, DNA methylation, and protein quantitative trait locus data from blood and brain tissues. Outcome data was from a case-control genome-wide association study, which included 4110 sCJD patients and 13,569 controls. Further investigations encompassed druggability, potential side effects, and associated biological pathways of the identified targets. RESULTS Integrative multi-omics analysis identified 23 potential therapeutic targets for sCJD, with five targets (STX6, XYLT2, PDIA4, FUCA2, KIAA1614) having higher levels of evidence. One target (XYLT2) shows promise for repurposing, two targets (XYLT2, PDIA4) are druggable, and three (STX6, KIAA1614, and FUCA2) targets represent potential future breakthrough points. The expression level of STX6 and XYLT2 in neurons and oligodendrocytes was closely associated with an increased risk of sCJD. Brain regions with high expression of STX6 or causal links to sCJD were often those areas commonly affected by sCJD. CONCLUSIONS Our study identified five potential therapeutic targets for sCJD. Further investigations are warranted to elucidate the mechanisms underlying the new targets for developing disease therapies or initiate clinical trials.
Collapse
Affiliation(s)
- Deming Jiang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haitian Nan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhongyun Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wen-Quan Zou
- Institute of Neurology, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Liyong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
169
|
Chen H, Lu J, Wang Z, Wu S, Zhang S, Geng J, Hou C, He P, Lu X. Unlocking reproducible transcriptomic signatures for acute myeloid leukaemia: Integration, classification and drug repurposing. J Cell Mol Med 2024; 28:e70085. [PMID: 39267259 PMCID: PMC11392829 DOI: 10.1111/jcmm.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/25/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a highly heterogeneous disease, which lead to various findings in transcriptomic research. This study addresses these challenges by integrating 34 datasets, including 26 control groups, 6 prognostic datasets and 2 single-cell RNA sequencing (scRNA-seq) datasets to identify 10,000 AML-related genes (ARGs). We focused on genes with low variability and high consistency and successfully discovered 191 AML signatures (ASs). Leveraging machine learning techniques, specifically the XGBoost model and our custom framework, we classified AML subtypes with both scRNA-seq and bulk RNA-seq data, complementing the ELN2022 classification approach. Our research also identified promising treatments for AML through drug repurposing, with solasonine showing potential efficacy for high-risk AML patients, supported by molecular docking and transcriptomic analyses. To enhance reproducibility and customizability, we developed CSAMLdb, a user-friendly database platform. It facilitates the reuse and personalized analysis of nearly all results obtained in this research, including single-gene prognostics, multi-gene scoring, enrichment analysis, machine learning risk assessment, drug repositioning analysis and literature abstract named entity recognition. CSAMLdb is available at http://www.csamldb.com.
Collapse
Affiliation(s)
- Haoran Chen
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- School of Management, Shanxi Medical University, Taiyuan, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Jinqi Lu
- Department of Computer Science, Boston University, Boston, Massachusetts, USA
| | - Zining Wang
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Shengnan Wu
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Shengxiao Zhang
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Jie Geng
- Basic Medicine College, Shanxi Medical University, Taiyuan, China
| | - Chuandong Hou
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Peifeng He
- School of Management, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Xuechun Lu
- School of Management, Shanxi Medical University, Taiyuan, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing, China
| |
Collapse
|
170
|
Feng Y, Li C, Cheng B, Chen Y, Chen P, Wang Z, Zheng X, He J, Zhu F, Wang W, Liang W. Identifying genetically-supported drug repurposing targets for non-small cell lung cancer through mendelian randomization of the druggable genome. Transl Lung Cancer Res 2024; 13:1780-1793. [PMID: 39263038 PMCID: PMC11384480 DOI: 10.21037/tlcr-24-65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/04/2024] [Indexed: 09/13/2024]
Abstract
Background Lung cancer is responsible for most cancer-related deaths, and non-small cell lung cancer (NSCLC) accounts for the majority of cases. Targeted therapy has made promising advancements in systemic treatment for NSCLC over the last two decades, but inadequate drug targets with clinically proven survival benefits limit its universal application in clinical practice compared to chemotherapy and immunotherapy. There is an urgent need to explore new drug targets to expand the beneficiary group. This study aims to identify druggable genes and to predict the efficacy and prognostic value of the corresponding targeted drugs in NSCLC. Methods Two-sample mendelian randomization (MR) of druggable genes was performed to predict the efficacy of their corresponding targeted therapy for NSCLC. Subsequent sensitivity analyses were performed to assess potential confounders. Accessible RNA sequencing data were incorporated for subsequent verifications, and Kaplan-Meier survival curves of different gene expressions were used to explore the prognostic value of candidate druggable genes. Results MR screening encompassing 4,863 expression quantitative trait loci (eQTL) and 1,072 protein quantitative trait loci (pQTL, with 453 proteins overlapping) were performed. Seven candidate druggable genes were identified, including CD33, ENG, ICOSLG and IL18R1 for lung adenocarcinoma, and VSIR, FSTL1 and TIMP2 for lung squamous cell carcinoma. The results were validated by further transcriptomic investigations. Conclusions Drugs targeting genetically supported genomes are considerably more likely to yield promising efficacy and succeed in clinical trials. We provide compelling genetic evidence to prioritize drug development for NSCLC.
Collapse
Affiliation(s)
- Yi Feng
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Caichen Li
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Bo Cheng
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Ying Chen
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Peiling Chen
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Zixun Wang
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Xiangyuan Zheng
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Juan He
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Feng Zhu
- Internal Medicine Department, Detroit Medical Center Sinai-Grace Hospital, Detroit, MI, USA
| | - Wei Wang
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Wenhua Liang
- Dpartment of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
- Department of Oncology Medical Center, The First People's Hospital of Zhaoqing, Zhaoqing, China
| |
Collapse
|
171
|
Rabby MG, Suzauddula M, Hasan MS, Dewan MA, Islam MN. In-silico identification and functional characterization of common genes associated with type 2 diabetes and hypertension. Heliyon 2024; 10:e36546. [PMID: 39262940 PMCID: PMC11388505 DOI: 10.1016/j.heliyon.2024.e36546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Type 2 diabetes (T2D) and hypertension are global public health concerns and major metabolic disorders in humans. Experimental evidence indicates considerable hereditary influences on the etiology of T2D and hypertension, but the molecular basis of these diseases is still limited. Thus, the current study analyzed 185 (132 T2D and 53 hypertension) GWAS catalog datasets and identified 83 common genes linked to T2D and hypertension pathogenesis. These genes were further examined using various bioinformatics approaches to elucidate their molecular mechanisms underlying the pathophysiology of T2D and hypertension. Gene ontology (GO) analysis revealed the biological, cellular, and molecular functions of these genes, which were also linked to different T2D and hypertension pathways. Specifically, seven genes were found to be crucial for T2D, and nine were directly associated with hypertension. Protein-protein interaction (PPI) analysis identified 28 candidate genes and seven hub genes through 11 topological methods. Among 231 miRNAs, seven were significant in interacting with the hub genes, and nine transcription factors (TFs) out of 36 were linked to these hub genes. Additionally, two of the seven hub genes were downregulated by 43 FDA-approved drugs. These findings elucidate the molecular processes underlying T2D and hypertension, suggesting that targeting these genes could lead to future drug development and therapeutic strategies to treat T2D and hypertension.
Collapse
Affiliation(s)
- Md Golam Rabby
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Suzauddula
- College of Agriculture and Natural Resources, National Chung Hsing University, Taichung City, 40227, Taiwan
| | - Md Sakib Hasan
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mahbubur Alam Dewan
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Numan Islam
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
- Department of Food Science and Technology, University of Nebraska Lincoln, USA
| |
Collapse
|
172
|
Song M, Chen F, Li X, Chen L. Exploring causal correlations between plasma proteins and peripheral neuropathy: a Mendelian randomization. Front Neurol 2024; 15:1431669. [PMID: 39268072 PMCID: PMC11390399 DOI: 10.3389/fneur.2024.1431669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background Peripheral neuropathy (PN) is a common neurological disorder, and circulating plasma proteins with causal genetic evidence are a major source of therapeutic targets. This study identifies several potential plasma proteins that are causally related to PN risk, providing new insights into protein-mediated pathogenesis of PN and potential targets for novel therapies. Methods To identify potential therapeutic targets for PN, we employed two-sample Mendelian randomization (MR) to identify plasma proteins associated with six common PN. First, we screened for proteins related to PN using genome-wide association studies (GWAS), obtaining genetic data on plasma proteomes from 35,559 Icelanders. Summary data for six common PN, including Carpal Tunnel Syndrome (CTS), Trigeminal Neuralgia (TN), Alcoholic Neuropathy (AIP), Drug-induced Neuropathy (DIP), Diabetic Neuropathy (DP), and Guillain-Barré syndrome (GBS), were obtained from the FinnGen database. Two-sample MR and colocalization analyses were then conducted to further identify protein-PN pairs with presumed causal relationships. Enrichment analysis of positive proteins revealed potential biological processes and pathways. Based on drug-gene interaction analysis, we ultimately identified causal proteins associated with PN that could serve as potential drug targets for treating PN. Results Through MR analysis, we identified eight proteins (UBC12, SEM4C, IL23R, Prothrombin, CBS, Microglobulin, MATN4, COLEC12) with causal relationships to PN. We found that UBC12 is a protective factor for DP and CTS, while the remaining proteins are risk factors. Further colocalization analysis showed a posterior probability of hypothesis 4 (PPH4) less than 0.75, indicating no positive colocalization results were found. From the pathway enrichment analysis, we discovered that the proteins were mainly concentrated in pathways related to defense response to bacterium, receptor signaling pathway via STAT, cell killing, negative regulation of cytokine production, and leukocyte mediated immunity. Finally, in Drug-Gene Interaction database (DGIdb), we identified three protein-coding genes (IL23R, F2, CBS) as potential drug targets for PN. Conclusion Mendelian randomization studies confirm the causal relationship between genetically predicted PN-related risk and genetically predicted plasma protein abundance. Plasma proteins, as biomarkers associated with PN, can provide potential drug targets for etiological intervention research in PN.
Collapse
Affiliation(s)
- Man Song
- Department of Intensive Care Unit, Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| | - Fang Chen
- Department of Intensive Care Unit, Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| | - Xiaocong Li
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lu Chen
- Department of Intensive Care Unit, Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| |
Collapse
|
173
|
Boulay G, Broye LC, Dong R, Iyer S, Sanalkumar R, Xing YH, Buisson R, Rengarajan S, Naigles B, Duc B, Volorio A, Awad ME, Renella R, Chebib I, Nielsen GP, Choy E, Cote GM, Zou L, Letovanec I, Stamenkovic I, Rivera MN, Riggi N. EWS-WT1 fusion isoforms establish oncogenic programs and therapeutic vulnerabilities in desmoplastic small round cell tumors. Nat Commun 2024; 15:7460. [PMID: 39198430 PMCID: PMC11358472 DOI: 10.1038/s41467-024-51851-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
EWS fusion oncoproteins underlie several human malignancies including Desmoplastic Small Round Cell Tumor (DSRCT), an aggressive cancer driven by EWS-WT1 fusion proteins. Here we combine chromatin occupancy and 3D profiles to identify EWS-WT1-dependent gene regulation networks and target genes. We show that EWS-WT1 is a powerful chromatin activator controlling an oncogenic gene expression program that characterizes primary tumors. Similar to wild type WT1, EWS-WT1 has two isoforms that differ in their DNA binding domain and we find that they have distinct DNA binding profiles and are both required to generate viable tumors that resemble primary DSRCT. Finally, we identify candidate EWS-WT1 target genes with potential therapeutic implications, including CCND1, whose inhibition by the clinically-approved drug Palbociclib leads to marked tumor burden decrease in DSRCT PDXs in vivo. Taken together, our studies identify gene regulation programs and therapeutic vulnerabilities in DSRCT and provide a mechanistic understanding of the complex oncogenic activity of EWS-WT1.
Collapse
Affiliation(s)
- Gaylor Boulay
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Liliane C Broye
- Experimental Pathology Service, Lausanne University Hospital & University of Lausanne, Lausanne, Switzerland
| | - Rui Dong
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sowmya Iyer
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rajendran Sanalkumar
- Experimental Pathology Service, Lausanne University Hospital & University of Lausanne, Lausanne, Switzerland
| | - Yu-Hang Xing
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rémi Buisson
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shruthi Rengarajan
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Beverly Naigles
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benoît Duc
- Experimental Pathology Service, Lausanne University Hospital & University of Lausanne, Lausanne, Switzerland
| | - Angela Volorio
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mary E Awad
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raffaele Renella
- Department Woman-Mother-Child, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ivan Chebib
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - G Petur Nielsen
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Edwin Choy
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Gregory M Cote
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Lee Zou
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Igor Letovanec
- Department of Histopathology, Central Institute, Valais Hospital, Sion, Switzerland
- Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ivan Stamenkovic
- Experimental Pathology Service, Lausanne University Hospital & University of Lausanne, Lausanne, Switzerland
| | - Miguel N Rivera
- Department of Pathology & Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Nicolò Riggi
- Experimental Pathology Service, Lausanne University Hospital & University of Lausanne, Lausanne, Switzerland.
- Genentech Inc, Department of Cell and Tissue Genomics (CTG), South San Francisco, CA, USA.
| |
Collapse
|
174
|
Zhang C, Yang Z, Li X, Zhao L, Guo W, Deng W, Wang Q, Hu X, Li M, Sham PC, Xiao X, Li T. Unraveling NEK4 as a Potential Drug Target in Schizophrenia and Bipolar I Disorder: A Proteomic and Genomic Approach. Schizophr Bull 2024; 50:1185-1196. [PMID: 38869147 PMCID: PMC11349004 DOI: 10.1093/schbul/sbae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
BACKGROUND AND HYPOTHESIS Investigating the shared brain protein and genetic components of schizophrenia (SCZ) and bipolar I disorder (BD-I) presents a unique opportunity to understand the underlying pathophysiological processes and pinpoint potential drug targets. STUDY DESIGN To identify overlapping susceptibility brain proteins in SCZ and BD-I, we carried out proteome-wide association studies (PWAS) and Mendelian Randomization (MR) by integrating human brain protein quantitative trait loci with large-scale genome-wide association studies for both disorders. We utilized transcriptome-wide association studies (TWAS) to determine the consistency of mRNA-protein dysregulation in both disorders. We applied pleiotropy-informed conditional false discovery rate (pleioFDR) analysis to identify common risk genetic loci for SCZ and BD-I. Additionally, we performed a cell-type-specific analysis in the human brain to detect risk genes notably enriched in distinct brain cell types. The impact of risk gene overexpression on dendritic arborization and axon length in neurons was also examined. STUDY RESULTS Our PWAS identified 42 proteins associated with SCZ and 14 with BD-I, among which NEK4, HARS2, SUGP1, and DUS2 were common to both conditions. TWAS and MR analysis verified the significant risk gene NEK4 for both SCZ and BD-I. PleioFDR analysis further supported genetic risk loci associated with NEK4 for both conditions. The cell-type specificity analysis revealed that NEK4 is expressed on the surface of glutamatergic neurons, and its overexpression enhances dendritic arborization and axon length in cultured primary neurons. CONCLUSIONS These findings underscore a shared genetic origin for SCZ and BD-I, offering novel insights for potential therapeutic target identification.
Collapse
Affiliation(s)
- Chengcheng Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
| | - ZhiHui Yang
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaojing Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liansheng Zhao
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wanjun Guo
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Deng
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiang Wang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xun Hu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ming Li
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Pak Chung Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Xiao Xiao
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Tao Li
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
175
|
Wang Y, Zhang S, Gong W, Liu X, Mo Q, Shen L, Zhao Y, Wang S, Yuan Z. Multi-Omics Integration Analysis Pinpoint Proteins Influencing Brain Structure and Function: Toward Drug Targets and Neuroimaging Biomarkers for Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:9223. [PMID: 39273172 PMCID: PMC11395524 DOI: 10.3390/ijms25179223] [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: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Integrating protein quantitative trait loci (pQTL) data and summary statistics from genome-wide association studies (GWAS) of brain image-derived phenotypes (IDPs) can benefit in identifying IDP-related proteins. Here, we developed a systematic omics-integration analytic framework by sequentially using proteome-wide association study (PWAS), Mendelian randomization (MR), and colocalization (COLOC) analyses to identify the potentially causal brain and plasma proteins for IDPs, followed by pleiotropy analysis, mediation analysis, and drug exploration analysis to investigate potential mediation pathways of pleiotropic proteins to neuropsychiatric disorders (NDs) as well as candidate drug targets. A total of 201 plasma proteins and 398 brain proteins were significantly associated with IDPs from PWAS analysis. Subsequent MR and COLOC analyses further identified 313 potentially causal IDP-related proteins, which were significantly enriched in neural-related phenotypes, among which 91 were further identified as pleiotropic proteins associated with both IDPs and NDs, including EGFR, TMEM106B, GPT, and HLA-B. Drug prioritization analysis showed that 6.33% of unique pleiotropic proteins had drug targets or interactions with medications for NDs. Nine potential mediation pathways were identified to illustrate the mediating roles of the IDPs in the causal effect of the pleiotropic proteins on NDs, including the indirect effect of TMEM106B on Alzheimer's disease (AD) risk via radial diffusivity (RD) of the posterior limb of the internal capsule (PLIC), with the mediation proportion being 11.18%, and the indirect effect of EGFR on AD through RD of PLIC, RD of splenium of corpus callosum (SCC), and fractional anisotropy (FA) of SCC, with the mediation proportion being 18.99%, 22.79%, and 19.91%, respectively. These findings provide novel insights into pathogenesis, drug targets, and neuroimaging biomarkers of NDs.
Collapse
Affiliation(s)
- Yunzhuang Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Sunjie Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Weiming Gong
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Xinyu Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Qinyou Mo
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Lujia Shen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Yansong Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Shukang Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhua West Road, Jinan 250012, China
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| |
Collapse
|
176
|
Yang S, Guo J, Xiong Y, Han G, Luo T, Peng S, Liu J, Hu T, Zha Y, Lin X, Tan Y, Zhang J. Unraveling the genetic and molecular landscape of sepsis and acute kidney injury: A comprehensive GWAS and machine learning approach. Int Immunopharmacol 2024; 137:112420. [PMID: 38851159 DOI: 10.1016/j.intimp.2024.112420] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVES This study aimed to explore the underlying mechanisms of sepsis and acute kidney injury (AKI), including sepsis-associated AKI (SA-AKI), a frequent complication in critically ill sepsis patients. METHODS GWAS data was analyzed for genetic association between AKI and sepsis. Then, we systematically applied three distinct machine learning algorithms (LASSO, SVM-RFE, RF) to rigorously identify and validate signature genes of SA-AKI, assessing their diagnostic and prognostic value through ROC curves and survival analysis. The study also examined the functional and immunological aspects of these genes, potential drug targets, and ceRNA networks. A mouse model of sepsis was created to test the reliability of these signature genes. RESULTS LDSC confirmed a positive genetic correlation between AKI and sepsis, although no significant shared loci were found. Bidirectional MR analysis indicated mutual increased risks of AKI and sepsis. Then, 311 key genes common to sepsis and AKI were identified, with 42 significantly linked to sepsis prognosis. Six genes, selected through LASSO, SVM-RFE, and RF algorithms, showed excellent predictive performance for sepsis, AKI, and SA-AKI. The models demonstrated near-perfect AUCs in both training and testing datasets, and a perfect AUC in a sepsis mouse model. Significant differences in immune cells, immune-related pathways, HLA, and checkpoint genes were found between high- and low-risk groups. The study identified 62 potential drug treatments for sepsis and AKI and constructed a ceRNA network. CONCLUSIONS The identified signature genes hold potential clinical applications, including prognostic evaluation and targeted therapeutic strategies for sepsis and AKI. However, further research is needed to confirm these findings.
Collapse
Affiliation(s)
- Sha Yang
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China
| | - Jing Guo
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China
| | - Yunbiao Xiong
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Tao Luo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shuo Peng
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jian Liu
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Tieyi Hu
- Department of Neurology, the Affiliated Dazu Hospital of Chongqing Medical University , China
| | - Yan Zha
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xin Lin
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China.
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China.
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.
| |
Collapse
|
177
|
Yuan W, Li J, Gao S, Sun W, Zhao F. Novel therapeutic targets for primary open-angle glaucoma identified through multicenter proteome-wide mendelian randomization. Front Pharmacol 2024; 15:1428472. [PMID: 39221148 PMCID: PMC11362091 DOI: 10.3389/fphar.2024.1428472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Background This study aimed to identify novel therapeutic targets for primary open-angle glaucoma (POAG). Methods The summary-data-based Mendelian randomization (SMR) method was used to evaluate the genetic association between plasma proteins and POAG. Two sets of plasma protein quantitative trait loci (pQTLs) data considered exposures were obtained from the Icelandic Decoding Genetics Study and UK Biobank Pharma Proteomics Project. The summary-level genome-wide association studies data for POAG were extracted from the latest Round 10 release of the FinnGen consortium (8,530 cases and 391,275 controls) and the UK Biobank (4,737 cases and 458,196 controls). Colocalization analysis was used to screen out pQTLs that share the same variant with POAG as drug targets identified. The two-sample Mendelian randomization, reverse causality testing and phenotype scanning were performed to further validate the main findings. Protein-protein interaction, pathway enrichment analysis and druggability assessment were conducted to determine whether the identified plasma proteins have potential as drug targets. Results After systematic analysis, this study identified eight circulating proteins as potential therapeutic targets for POAG. Three causal proteins with strong evidence of colocalization, ROBO1 (OR = 1.38, p = 1.48 × 10-4, PPH4 = 0.865), FOXO3 (OR = 0.35, p = 4.34 × 10-3, PPH4 = 0.796), ITIH3 (OR = 0.89, p = 2.76 × 10-4, PPH4 = 0.767), were considered tier one targets. Five proteins with medium support evidence of colocalization, NCR1 (OR = 1.25, p = 4.18 × 10-4, PPH4 = 0.682), NID1 (OR = 1.38, p = 1.54 × 10-3, PPH4 = 0.664), TIMP3 (OR = 0.91, p = 4.01 × 10-5, PPH4 = 0.659), SERPINF1 (OR = 0.81, p = 2.77 × 10-4, PPH4 = 0.59), OXT (OR = 1.17, p = 9.51 × 10-4, PPH4 = 0.526), were classified as tier two targets. Additional sensitivity analyses further validated the robustness and directionality of these findings. According to druggability assessment, Pimagedine, Resveratrol, Syringaresinol and Clozapine may potentially be important in the development of new anti-glaucoma agents. Conclusion Our integrated study identified eight potential associated proteins for POAG. These proteins play important roles in neuroprotection, extracellular matrix regulation and oxidative stress. Therefore, they have promising potential as therapeutic targets to combat POAG.
Collapse
Affiliation(s)
- Weichen Yuan
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Jun Li
- Department of Ultrasonography, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shang Gao
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Wei Sun
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Fangkun Zhao
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| |
Collapse
|
178
|
Savage SR, Yi X, Lei JT, Wen B, Zhao H, Liao Y, Jaehnig EJ, Somes LK, Shafer PW, Lee TD, Fu Z, Dou Y, Shi Z, Gao D, Hoyos V, Gao Q, Zhang B. Pan-cancer proteogenomics expands the landscape of therapeutic targets. Cell 2024; 187:4389-4407.e15. [PMID: 38917788 PMCID: PMC12010439 DOI: 10.1016/j.cell.2024.05.039] [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: 02/16/2023] [Revised: 04/03/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
Fewer than 200 proteins are targeted by cancer drugs approved by the Food and Drug Administration (FDA). We integrate Clinical Proteomic Tumor Analysis Consortium (CPTAC) proteogenomics data from 1,043 patients across 10 cancer types with additional public datasets to identify potential therapeutic targets. Pan-cancer analysis of 2,863 druggable proteins reveals a wide abundance range and identifies biological factors that affect mRNA-protein correlation. Integration of proteomic data from tumors and genetic screen data from cell lines identifies protein overexpression- or hyperactivation-driven druggable dependencies, enabling accurate predictions of effective drug targets. Proteogenomic identification of synthetic lethality provides a strategy to target tumor suppressor gene loss. Combining proteogenomic analysis and MHC binding prediction prioritizes mutant KRAS peptides as promising public neoantigens. Computational identification of shared tumor-associated antigens followed by experimental confirmation nominates peptides as immunotherapy targets. These analyses, summarized at https://targets.linkedomics.org, form a comprehensive landscape of protein and peptide targets for companion diagnostics, drug repurposing, and therapy development.
Collapse
Affiliation(s)
- Sara R Savage
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xinpei Yi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bo Wen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongwei Zhao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yuxing Liao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric J Jaehnig
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lauren K Somes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Paul W Shafer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tobie D Lee
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zile Fu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yongchao Dou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhiao Shi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daming Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Valentina Hoyos
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
179
|
Qing L, Wu W. The mechanism of geniposide in patients with COVID-19 and atherosclerosis: A pharmacological and bioinformatics analysis. Medicine (Baltimore) 2024; 103:e39065. [PMID: 39093733 PMCID: PMC11296471 DOI: 10.1097/md.0000000000039065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
In patients with severe acute respiratory syndrome coronavirus 2 (which causes coronavirus disease 2019 [COVID-19]), oxidative stress (OS) is associated with disease severity and death. OS is also involved in the pathogenesis of atherosclerosis (AS). Previous studies have shown that geniposide has anti-inflammatory and anti-viral properties, and can protect cells against OS. However, the potential target(s) of geniposide in patients with COVID-19 and AS, as well as the mechanism it uses, are unclear. We combined pharmacology and bioinformatics analysis to obtain geniposide against COVID-19/AS targets, and build protein-protein interaction network to filter hub genes. The hub genes were performed an enrichment analysis by ClueGO, including Gene Ontology and KEGG. The Enrichr database and the target microRNAs (miRNAs) of hub genes were predicted through the MiRTarBase via Enrichr. The common miRNAs were used to construct the miRNAs-mRNAs regulated network, and the miRNAs' function was evaluated by mirPath v3.0 software. Two hundred forty-seven targets of geniposide were identified in patients with COVID-19/AS comorbidity by observing the overlap between the genes modulated by geniposide, COVID-19, and AS. A protein-protein interaction network of geniposide in patients with COVID-19/AS was constructed, and 27 hub genes were identified. The results of enrichment analysis suggested that geniposide may be involved in regulating the OS via the FoxO signaling pathway. MiRNA-mRNA network revealed that hsa-miR-34a-5p may play an important role in the therapeutic mechanism of geniposide in COVID-19/AS patients. Our study found that geniposide represents a promising therapy for patients with COVID-19 and AS comorbidity. Furthermore, the target genes and miRNAs that we identified may aid the development of new treatment strategies against COVID-19/AS.
Collapse
Affiliation(s)
- Lijin Qing
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| |
Collapse
|
180
|
Jin X, Dong S, Yang Y, Bao G, Ma H. Nominating novel proteins for anxiety via integrating human brain proteomes and genome-wide association study. J Affect Disord 2024; 358:129-137. [PMID: 38697224 DOI: 10.1016/j.jad.2024.04.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND The underlying pathogenesis of anxiety remain elusive, making the pinpointing of potential therapeutic and diagnostic biomarkers for anxiety paramount to its efficient treatment. METHODS We undertook a proteome-wide association study (PWAS), fusing human brain proteomes from both discovery (ROS/MAP; N = 376) and validation cohorts (Banner; N = 152) with anxiety genome-wide association study (GWAS) summary statistics. Complementing this, we executed transcriptome-wide association studies (TWAS) leveraging human brain transcriptomic data from the Common Mind Consortium (CMC) to discern the confluence of genetic influences spanning both proteomic and transcriptomic levels. We further scrutinized significant genes through a suite of methodologies. RESULTS We discerned 14 genes instrumental in the genesis of anxiety through their specific cis-regulated brain protein abundance. Out of these, 6 were corroborated in the confirmatory PWAS, with 4 also showing associations with anxiety via their cis-regulated brain mRNA levels. A heightened confidence level was attributed to 5 genes (RAB27B, CCDC92, BTN2A1, TMEM106B, and DOC2A), taking into account corroborative evidence from both the confirmatory PWAS and TWAS, coupled with insights from mendelian randomization analysis and colocalization evaluations. A majority of the identified genes manifest in brain regions intricately linked to anxiety and predominantly partake in lysosomal metabolic processes. LIMITATIONS The limited scope of the brain proteome reference datasets, stemming from a relatively modest sample size, potentially curtails our grasp on the entire gamut of genetic effects. CONCLUSION The genes pinpointed in our research present a promising groundwork for crafting therapeutic interventions and diagnostic tools for anxiety.
Collapse
Affiliation(s)
- Xing Jin
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shuangshuang Dong
- Department of Neurology, General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Yang Yang
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guangyu Bao
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Haochuan Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Hospital of Chinese Medicine Postdoctoral Research Workstation, Guangzhou, Guangdong, China.
| |
Collapse
|
181
|
Shi F, Li T, Shi H, Wei Y, Wang J, Liu C, Liang R. Identification of potential therapeutic targets for skin cutaneous melanoma on the basic of transcriptomics. Skin Res Technol 2024; 30:e13916. [PMID: 39113615 PMCID: PMC11306917 DOI: 10.1111/srt.13916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/11/2024]
Abstract
BACKGROUND Advanced skin cutaneous melanoma (SKCM) is responsible for the majority of skin cancer-related deaths. Apart from the rare BRAF V600F mutation, which can be targeted with specific drugs, there are currently no other novel effective therapeutic targets. METHODS We used SMR analysis with cis-expressed quantitative trait locus (cis-eQTL) as the exposure variable and SKCM as the outcome variable to identify potential therapeutic targets for SKCM. Colocalization assays and HEIDI tests are used to test whether SKCM risk and gene expression are driven by common SNPs. Replication analysis further validated the findings, and we also constructed protein-protein interaction networks to explore the relationship between the identified genes and known SKCM targets. Drug prediction and molecular docking further validated the medicinal value of drug targets. Transcriptome differential analysis further validated that there were differences between normal tissues and SKCM for the selected targets. RESULTS We identified 13 genes significantly associated with the risk of SKCM, including five protective genes and eight harmful genes. The HEIDI test and co-localization analysis further indicates a causal association between genes (SOX4, MAFF) and SKCM, categorized as Class 1 evidence targets. The remaining 11 genes, except for HELZ2 show a moderately causal association with SKCM, categorized as Class 2 evidence targets. Target druggability predictions from DGIdb suggest that SOX4, MAFF, ACSF3, CDK10, SPG7, and TCF25 are likely to be future drug targets. CONCLUSION The study provides genetic evidence for targeting available drug genes for the treatment of SKCM.
Collapse
Affiliation(s)
- Fengling Shi
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouChina
| | - Tao Li
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouChina
| | - Huiling Shi
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouChina
| | - Yushan Wei
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouChina
| | - Juan Wang
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouChina
| | - Canyu Liu
- Department of Radiation OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
| | - Ruirong Liang
- Department of OncologyThe Fourth Affiliated Hospital of Soochow UniversitySuzhou Dushu Lake HospitalMedical Center of Soochow UniversitySuzhouChina
- Division of Clinical OncologyMedical Center of Soochow UniversitySuzhouChina
- Department of OncologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
182
|
Yuan C, Lin X, Liao R. Decoding the genetic landscape of allergic rhinitis: a comprehensive network analysis revealing key genes and potential therapeutic targets. J Asthma 2024; 61:823-834. [PMID: 38266128 DOI: 10.1080/02770903.2024.2306619] [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: 11/07/2023] [Accepted: 01/13/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Allergic Rhinitis (AR), an inflammatory affliction impacting the upper respiratory tract, has been registering a substantial surge in incidence across the globe. METHODS We embarked on examination of differentially expressed genes (DEGs) and the Weighted Gene Co-Expression Network Analysis (WGCNA). With this armory of genes identified, we engaged the tools of Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Our study continued with the establishment of a protein-protein interaction (PPI) network and the application of LASSO regression. Finally, we leveraged a docking model to elucidate potential drug-gene interactions involving these key genes. RESULTS Through WGCNA and different express genes screening, PPI network was performed, identifying top 20 key genes, including CD44, CD69, CD274. LASSO regression identified three independent factors, STARD5, CST1, and CHAC1, that were significantly associated with AR. A predictive model was developed with an AUC value over 0.75. Also, 105 potential therapeutic agents were discovered, including Fluorouracil, Cyclophosphamide, Doxorubicin, and Hydrocortisone, offering promising therapeutic strategies for AR. CONCLUSION By fuzing DEGs with key genes derived from WGCNA, this study has illuminated a comprehensive network of gene interactions involved in the pathogenesis of AR, paving the way for future biomarker and therapeutic target discovery in AR.
Collapse
Affiliation(s)
- Chile Yuan
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaohong Lin
- WEN Ziyuan Pediatric Academic School Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ruosha Liao
- Department of Pediatrics, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
183
|
Qi T, Song L, Guo Y, Chen C, Yang J. From genetic associations to genes: methods, applications, and challenges. Trends Genet 2024; 40:642-667. [PMID: 38734482 DOI: 10.1016/j.tig.2024.04.008] [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: 11/08/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024]
Abstract
Genome-wide association studies (GWASs) have identified numerous genetic loci associated with human traits and diseases. However, pinpointing the causal genes remains a challenge, which impedes the translation of GWAS findings into biological insights and medical applications. In this review, we provide an in-depth overview of the methods and technologies used for prioritizing genes from GWAS loci, including gene-based association tests, integrative analysis of GWAS and molecular quantitative trait loci (xQTL) data, linking GWAS variants to target genes through enhancer-gene connection maps, and network-based prioritization. We also outline strategies for generating context-dependent xQTL data and their applications in gene prioritization. We further highlight the potential of gene prioritization in drug repurposing. Lastly, we discuss future challenges and opportunities in this field.
Collapse
Affiliation(s)
- Ting Qi
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China.
| | - Liyang Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Yazhou Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Chang Chen
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Jian Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China.
| |
Collapse
|
184
|
Abulimiti M, Jia ZY, Wu Y, Yu J, Gong YH, Guan N, Xiong DQ, Ding N, Uddin N, Wang J. Exploring and clinical validation of prognostic significance and therapeutic implications of copper homeostasis-related gene dysregulation in acute myeloid leukemia. Ann Hematol 2024; 103:2797-2826. [PMID: 38879648 DOI: 10.1007/s00277-024-05841-6] [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: 05/09/2024] [Accepted: 06/08/2024] [Indexed: 07/28/2024]
Abstract
The patterns and biological functions of copper homeostasis-related genes (CHRGs) in acute myeloid leukemia (AML) remain unclear. We explored the patterns and biological functions of CHRGs in AML. Using independent cohorts, including TCGA-GTEx, GSE114868, GSE37642, and clinical samples, we identified 826 common differentially expressed genes. Specifically, 12 cuproptosis-related genes (e.g., ATP7A, ATP7B) were upregulated, while 17 cuproplasia-associated genes (e.g., ATOX1, ATP7A) were downregulated in AML. We used LASSO-Cox, Kaplan-Meier, and Nomogram analyses to establish prognostic risk models, effectively stratifying patients with AML into high- and low-risk groups. Subgroup analysis revealed that high-risk patients exhibited poorer overall survival and involvement in fatty acid metabolism, apoptosis, and glycolysis. Immune infiltration analysis indicated differences in immune cell composition, with notable increases in B cells, cytotoxic T cells, and memory T cells in the low-risk group, and increased monocytes and neutrophils in the high-risk group. Single-cell sequencing analysis corroborated the expression characteristics of critical CHRGs, such as MAPK1 and ATOX1, associated with the function of T, B, and NK cells. Drug sensitivity analysis suggested potential therapeutic agents targeting copper homeostasis, including Bicalutamide and Sorafenib. PCR validation confirmed the differential expression of 4 cuproptosis-related genes (LIPT1, SLC31A1, GCSH, and PDHA1) and 9 cuproplasia-associated genes (ATOX1, CCS, CP, MAPK1, SOD1, COA6, PDK1, DBH, and PDE3B) in AML cell line. Importantly, these genes serve as potential biomarkers for patient stratification and treatment. In conclusion, we shed light on the expression patterns and biological functions of CHRGs in AML. The developed risk models provided prognostic implications for patient survival, offering valuable information on the regulatory characteristics of CHRGs and potential avenues for personalized treatment in AML.
Collapse
Affiliation(s)
| | - Zheng-Yi Jia
- School of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Yun Wu
- Department of General Medicine, The First Affiliated Hospital of the Xinjiang Medical University, Urumqi, 830011, China
| | - Jing Yu
- Department of Teaching and Research, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Yue-Hong Gong
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, 830011, China
| | - Na Guan
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Dai-Qin Xiong
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, 830011, China
| | - Nan Ding
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, 830011, China
| | - Nazim Uddin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, 1205, Bangladesh
| | - Jie Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China.
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, 830011, China.
| |
Collapse
|
185
|
Yin K, Chen T, Gu X, Su W, Jiang Z, Lu S, Cao B, Chi L, Gao X, Chen Y. Systematic druggable genome-wide Mendelian randomization identifies therapeutic targets for sarcopenia. J Cachexia Sarcopenia Muscle 2024; 15:1324-1334. [PMID: 38644354 PMCID: PMC11294052 DOI: 10.1002/jcsm.13479] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND There are no effective pharmacological treatments for sarcopenia. We aim to identify potential therapeutic targets for sarcopenia by integrating various publicly available datasets. METHODS We integrated druggable genome data, cis-eQTL/cis-pQTL from human blood and skeletal muscle tissue, and GWAS summary data of sarcopenia-related traits to analyse the potential causal relationships between drug target genes and sarcopenia using the Mendelian Randomization (MR) method. Sensitivity analyses and Bayesian colocalization were employed to validate the causal relationships. We also assessed the side effects or additional indications of the identified drug targets using a phenome-wide MR (Phe-MR) approach and investigated actionable drugs for target genes using available databases. RESULTS MR analysis identified 17 druggable genes with potential causation to sarcopenia in human blood or skeletal muscle tissue. Six of them (HP, HLA-DRA, MAP 3K3, MFGE8, COL15A1, and AURKA) were further confirmed by Bayesian colocalization (PPH4 > 90%). The up-regulation of HP [higher ALM (beta: 0.012, 95% CI: 0.007-0.018, P = 1.2*10-5) and higher grip strength (OR: 0.96, 95% CI: 0.94-0.98, P = 4.2*10-5)], MAP 3K3 [higher ALM (beta: 0.24, 95% CI: 0.21-0.26, P = 1.8*10-94), higher grip strength (OR: 0.82, 95% CI: 0.75-0.90, P = 2.1*10-5), and faster walking pace (beta: 0.03, 95% CI: 0.02-0.05, P = 8.5*10-6)], and MFGE8 [higher ALM (muscle eQTL, beta: 0.09, 95% CI: 0.06-0.11, P = 6.1*10-13; blood pQTL, beta: 0.05, 95% CI: 0.03-0.07, P = 3.8*10-09)], as well as the down-regulation of HLA-DRA [lower ALM (beta: -0.09, 95% CI: -0.11 to -0.08, P = 5.4*10-36) and lower grip strength (OR: 1.13, 95% CI: 1.07-1.20, P = 1.8*10-5)] and COL15A1 [higher ALM (muscle eQTL, beta: -0.07, 95% CI: -0.10 to -0.04, P = 3.4*10-07; blood pQTL, beta: -0.05, 95% CI: -0.06 to -0.03, P = 1.6*10-07)], decreased the risk of sarcopenia. AURKA in blood (beta: -0.16, 95% CI: -0.22 to -0.09, P = 2.1*10-06) and skeletal muscle (beta: 0.03, 95% CI: 0.02 to 0.05, P = 5.3*10-05) tissues showed an inverse relationship with sarcopenia risk. The Phe-MR indicated that the six potential therapeutic targets for sarcopenia had no significant adverse effects. Drug repurposing analysis supported zinc supplementation and collagenase clostridium histolyticum might be potential therapeutics for sarcopenia by activating HP and inhibiting COL15A1, respectively. CONCLUSIONS Our research indicated MAP 3K3, MFGE8, COL15A1, HP, and HLA-DRA may serve as promising targets for sarcopenia, while the effectiveness of zinc supplementation and collagenase clostridium histolyticum for sarcopenia requires further validation.
Collapse
Affiliation(s)
- Kang‐Fu Yin
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology, West China HospitalSichuan UniversityChengduChina
| | - Ting Chen
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology, West China HospitalSichuan UniversityChengduChina
| | - Xiao‐Jing Gu
- Mental Health Center, West China HospitalSichuan UniversityChengduChina
| | - Wei‐Ming Su
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology, West China HospitalSichuan UniversityChengduChina
| | - Zheng Jiang
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology, West China HospitalSichuan UniversityChengduChina
| | - Si‐Jia Lu
- Department of RespiratoryThe Fourth People's Hospital of Chengdu, Mental Health Center of ChengduChengduChina
| | - Bei Cao
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology, West China HospitalSichuan UniversityChengduChina
| | - Li‐Yi Chi
- Department of NeurologyFirst Affiliated Hospital of Air Force Military Medical UniversityXi'anChina
| | - Xia Gao
- Department of GeriatricsDazhou Central HospitalDazhouChina
| | - Yong‐Ping Chen
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
- Institute of Brain Science and Brain‐Inspired Technology, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
186
|
Kouhmareh K, Martin E, Finlay D, Bhadada A, Hernandez-Vargas H, Downey F, Allen JK, Teriete P. Capture of circulating metastatic cancer cell clusters from lung cancer patients can reveal unique genomic profiles and potential anti-metastatic molecular targets: A proof-of-concept study. PLoS One 2024; 19:e0306450. [PMID: 39083508 PMCID: PMC11290651 DOI: 10.1371/journal.pone.0306450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
Metastasis remains the leading cause of cancer deaths worldwide and lung cancer, known for its highly metastatic progression, remains among the most lethal of malignancies. Lung cancer metastasis can selectively spread to multiple different organs, however the genetic and molecular drivers for this process are still poorly understood. Understanding the heterogeneous genomic profile of lung cancer metastases is considered key in identifying therapeutic targets that prevent its spread. Research has identified the key source for metastasis being clusters of cells rather than individual cancer cells. These clusters, known as metastatic cancer cell clusters (MCCCs) have been shown to be 100-fold more tumorigenic than individual cancer cells. Unfortunately, access to these primary drivers of metastases remains difficult and has limited our understanding of their molecular and genomic profiles. Strong evidence in the literature suggests that differentially regulated biological pathways in MCCCs can provide new therapeutic drug targets to help combat cancer metastases. In order to expand research into MCCCs and their role in metastasis, we demonstrate a novel, proof of principle technology, to capture MCCCs directly from patients' whole blood. Our platform can be readily tuned for different solid tumor types by combining a biomimicry-based margination effect coupled with immunoaffinity to isolate MCCCs. Adopting a selective capture approach based on overexpressed CD44 in MCCCs provides a methodology that preferentially isolates them from whole blood. Furthermore, we demonstrate a high capture efficiency of more than 90% when spiking MCCC-like model cell clusters into whole blood. Characterization of the captured MCCCs from lung cancer patients by immunofluorescence staining and genomic analyses, suggests highly differential morphologies and genomic profiles. This study lays the foundation to identify potential drug targets thus unlocking a new area of anti-metastatic therapeutics.
Collapse
Affiliation(s)
| | - Erika Martin
- PhenoVista Biosciences, San Diego, CA, United States of America
| | - Darren Finlay
- National Cancer Institute Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States of America
| | | | | | | | | | - Peter Teriete
- TumorGen Inc., San Diego, CA, United States of America
- IDEAYA Biosciences, South San Francisco, CA, United States of America
| |
Collapse
|
187
|
Gao B, Yan S, Xie W, Shao G. Bioinformatics reveals the potential mechanisms and biomarkers of necroptosis in neuroblastoma. Transl Cancer Res 2024; 13:3599-3619. [PMID: 39145050 PMCID: PMC11319990 DOI: 10.21037/tcr-24-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/29/2024] [Indexed: 08/16/2024]
Abstract
Background Neuroblastoma (NB) is a malignant tumor primarily found in children, presenting significant challenges in its development and prognosis. The role of necroptosis in the pathogenesis of NB has been acknowledged as crucial for treatment. This study aimed to investigate the key genes and functional pathways associated with necroptosis, as well as immune infiltration analysis, in NB. Furthermore, we aimed to evaluate the diagnostic significance of these genes for prognostic assessment and explore their potential immunological characteristics. Methods The NB dataset (GSE19274, GSE73517, and GSE85047) was obtained from the Gene Expression Omnibus (GEO) database, and genes associated with necroptosis were collected from GeneCards and previous literature. First, we conducted differential expression analysis and performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We employed gene set enrichment analysis (GSEA) to identify overlapping enriched functional pathways from the NB dataset. In addition, we constructed a protein-protein interaction (PPI) network, predicting relevant microRNAs (miRNAs) and transcription factors (TFs), as well as their corresponding drug predictions. Furthermore, the diagnostic value was assessed using receiver operating characteristic (ROC) curves. Finally, an immune infiltration analysis was performed. Results We identified six necroptosis-related differentially expressed genes (NRDEGs) closely associated with necroptosis in NB. They were enriched in Tuberculosis, Apoptosis-multiple species, Salmonella infection, legionellosis, and platinum drug resistance. GSEA and PPI network analyses, along with mRNA-drug interaction network, revealed 38 potential drugs corresponding to BIRC2, CAMK2G, CASP3, and IL8. ROC curve analysis showed that in GSE19274, FLOT2 with area under the ROC curve (AUC) of 0.850 and DAPK1 with AUC of 0.789. Conclusions Our study elucidates the key genes and functional pathways associated with necroptosis in NB, offering valuable insights to enhance our comprehension of the pathogenesis of NB, and improve prognosis assessment.
Collapse
Affiliation(s)
- Bing Gao
- Department of Public Health, International College, Krirk University, Bangkok, Thailand
| | - Shaochun Yan
- Center for Translational Medicine and Department of Laboratory Medicine, The Third People’s Hospital of Longgang District, Shenzhen, China
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Wei Xie
- Department of Public Health, International College, Krirk University, Bangkok, Thailand
- Center for Translational Medicine and Department of Laboratory Medicine, The Third People’s Hospital of Longgang District, Shenzhen, China
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guo Shao
- Department of Public Health, International College, Krirk University, Bangkok, Thailand
- Center for Translational Medicine and Department of Laboratory Medicine, The Third People’s Hospital of Longgang District, Shenzhen, China
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| |
Collapse
|
188
|
Xu Y, Han J, Fan Z, Liang S. Transcriptomics explores potential mechanisms for the development of Primary Sjogren's syndrome to diffuse large B-cell lymphoma in B cells. BMC Immunol 2024; 25:53. [PMID: 39080525 PMCID: PMC11287849 DOI: 10.1186/s12865-024-00646-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 07/26/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE Primary Sjogren's syndrome (pSS) is a prevalent autoimmune disease. The immune dysregulation it causes often leads to the development of diffuse large B-cell lymphoma (DLBCL) in clinical practice. However, how it contributes to these two disorders at the molecular level is not yet known. This study explored the potential molecular mechanisms associated with the differences between DLBCL and pSS. PATIENTS AND METHODS Gene expression matrices from discovery cohort 1, discovery cohort 2, and the validation cohort were downloaded from the GEO and TCGA databases. Weighted gene coexpression network analysis (WGCNA) was performed to identify the coexpression modules of DLBCL and pSS in discovery cohort 1 and obtain shared genes. GO and KEGG enrichment analyses and PPI network analysis were performed on the shared genes. Immune-related genes (IRGs) were intersected with shared genes to obtain common genes. Afterward, common genes were identified via machine learning methods. The immune infiltration analysis, miRNA-TF-hub gene regulatory chart, gene interactions of the hub genes, and gene‒drug target analysis were performed. Finally, STAT1 was identified as a possible essential gene by the above analysis, and immune infiltration and GSEA pathway analyses were performed in the high- and low-expression groups in discovery cohort 2. The diagnostic efficacy of the hub genes was assessed in the validation cohort, and clinical samples were collected for validation. RESULTS By WGCNA, one modular gene in each group was considered highly associated with the disease, and we obtained 28 shared genes. Enrichment analysis revealed shared genes involved in the viral response and regulation. We obtained four hub genes (ISG20, STAT1, TLR7, and RSAD2) via the algorithm. Hub genes and similar genes are primarily involved in regulating type I IFNs. The construction of a miRNA-TF-hub gene regulatory chart revealed that hsa-mir-155-5p, hsa-mir-146b-5p, hsa-mir-21-3p, and hsa-mir-126-3p play essential roles in both diseases. Hub genes were differentially expressed in B-cell memory according to immune infiltration analysis. Hub genes had a strong diagnostic effect on both diseases. STAT1 plays an essential role in immune cells in both diseases. CONCLUSION We identified hub susceptibility genes for DLBCL and pSS and identified hub genes and potential therapeutic targets that may act as biomarkers.
Collapse
Affiliation(s)
- Yanan Xu
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, P.R. China
| | - Jianxing Han
- Department of Stomatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Ziyi Fan
- Shanxi Bethune Hospital, Taiyuan, Shanxi, P.R. China
| | - Shufen Liang
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, P.R. China.
| |
Collapse
|
189
|
Cai H, Chen L, Yang S, Jiang R, Guo Y, He M, Luo Y, Hong G, Li H, Song K. Personalized differential expression analysis in triple-negative breast cancer. Brief Funct Genomics 2024; 23:495-506. [PMID: 38197537 DOI: 10.1093/bfgp/elad057] [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: 01/30/2023] [Revised: 11/16/2023] [Accepted: 12/04/2023] [Indexed: 01/11/2024] Open
Abstract
Identification of individual-level differentially expressed genes (DEGs) is a pre-step for the analysis of disease-specific biological mechanisms and precision medicine. Previous algorithms cannot balance accuracy and sufficient statistical power. Herein, RankCompV2, designed for identifying population-level DEGs based on relative expression orderings, was adjusted to identify individual-level DEGs. Furthermore, an optimized version of individual-level RankCompV2, named as RankCompV2.1, was designed based on the assumption that the rank positions of genes and relative rank differences of gene pairs would influence the identification of individual-level DEGs. In comparison to other individualized analysis algorithms, RankCompV2.1 performed better on statistical power, computational efficiency, and acquired coequal accuracy in both simulation and real paired cancer-normal data from ten cancer types. Besides, single sample GSEA and Gene Set Variation Analysis analysis showed that pathways enriched with up-regulated and down-regulated genes presented higher and lower enrichment scores, respectively. Furthermore, we identified 16 genes that were universally deregulated in 966 triple-negative breast cancer (TNBC) samples and interacted with Food and Drug Administration (FDA)-approved drugs or antineoplastic agents, indicating notable therapeutic targets for TNBC. In addition, we also identified genes with highly variable deregulation status and used these genes to cluster TNBC samples into three subgroups with different prognoses. The subgroup with the poorest outcome was characterized by down-regulated immune-regulated pathways, signal transduction pathways, and apoptosis-related pathways. Protein-protein interaction network analysis revealed that OAS family genes may be promising drug targets to activate tumor immunity in this subgroup. In conclusion, RankCompV2.1 is capable of identifying individual-level DEGs with high accuracy and statistical power, analyzing mechanisms of carcinogenesis and exploring therapeutic strategy.
Collapse
Affiliation(s)
- Hao Cai
- Medical Big Data and Bioinformatics Research Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Liangbo Chen
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, China
| | - Shuxin Yang
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, China
| | - Ronghong Jiang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, China
| | - You Guo
- Medical Big Data and Bioinformatics Research Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Ming He
- Medical Big Data and Bioinformatics Research Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Yun Luo
- Medical Big Data and Bioinformatics Research Centre, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Guini Hong
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, China
| | - Hongdong Li
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, China
| | - Kai Song
- Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| |
Collapse
|
190
|
Gudivada IP, Amajala KC. Integrative Bioinformatics Analysis for Targeting Hub Genes in Hepatocellular Carcinoma Treatment. Curr Genomics 2024; 26:48-80. [PMID: 39911278 PMCID: PMC11793067 DOI: 10.2174/0113892029308243240709073945] [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: 03/22/2024] [Revised: 04/24/2024] [Accepted: 06/11/2024] [Indexed: 02/07/2025] Open
Abstract
Background The damage in the liver and hepatocytes is where the primary liver cancer begins, and this is referred to as Hepatocellular Carcinoma (HCC). One of the best methods for detecting changes in gene expression of hepatocellular carcinoma is through bioinformatics approaches. Objective This study aimed to identify potential drug target(s) hubs mediating HCC progression using computational approaches through gene expression and protein-protein interaction datasets. Methodology Four datasets related to HCC were acquired from the GEO database, and Differentially Expressed Genes (DEGs) were identified. Using Evenn, the common genes were chosen. Using the Fun Rich tool, functional associations among the genes were identified. Further, protein-protein interaction networks were predicted using STRING, and hub genes were identified using Cytoscape. The selected hub genes were subjected to GEPIA and Shiny GO analysis for survival analysis and functional enrichment studies for the identified hub genes. The up-regulating genes were further studied for immunohistopathological studies using HPA to identify gene/protein expression in normal vs HCC conditions. Drug Bank and Drug Gene Interaction Database were employed to find the reported drug status and targets. Finally, STITCH was performed to identify the functional association between the drugs and the identified hub genes. Results The GEO2R analysis for the considered datasets identified 735 upregulating and 284 downregulating DEGs. Functional gene associations were identified through the Fun Rich tool. Further, PPIN network analysis was performed using STRING. A comparative study was carried out between the experimental evidence and the other seven data evidence in STRING, revealing that most proteins in the network were involved in protein-protein interactions. Further, through Cytoscape plugins, the ranking of the genes was analyzed, and densely connected regions were identified, resulting in the selection of the top 20 hub genes involved in HCC pathogenesis. The identified hub genes were: KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Further, GEPIA and Shiny GO analyses provided insights into survival ratios and functional enrichment studied for the hub genes. The HPA database studies further found that upregulating genes were involved in changes in protein expression in Normal vs HCC tissues. These findings indicated that hub genes were certainly involved in the progression of HCC. STITCH database studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, could be used as leads to identify novel drugs, and identified hub genes could also be considered as potential and promising drug targets as they are involved in the gene-chemical interaction networks. Conclusion The present study involved various integrated bioinformatics approaches, analyzing gene expression and protein-protein interaction datasets, resulting in the identification of 20 top-ranked hubs involved in the progression of HCC. They are KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Gene-chemical interaction network studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, can be used as leads to identify novel drugs, and the identified hub genes can be promising drug targets. The current study underscores the significance of targeting these hub genes and utilizing existing molecules to generate new molecules to combat liver cancer effectively and can be further explored in terms of drug discovery research to develop treatments for HCC.
Collapse
Affiliation(s)
- Indu Priya Gudivada
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Krishna Chaitanya Amajala
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| |
Collapse
|
191
|
Chen Y, Li E, Chang Z, Zhang T, Song Z, Wu H, Cheng ZJ, Sun B. Identifying potential therapeutic targets in lung adenocarcinoma: a multi-omics approach integrating bulk and single-cell RNA sequencing with Mendelian randomization. Front Pharmacol 2024; 15:1433147. [PMID: 39092217 PMCID: PMC11291359 DOI: 10.3389/fphar.2024.1433147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Our research aimed to identify new therapeutic targets for Lung adenocarcinoma (LUAD), a major subtype of non-small cell lung cancer known for its low 5-year survival rate of 22%. By employing a comprehensive methodological approach, we analyzed bulk RNA sequencing data from 513 LUAD and 59 non-tumorous tissues, identifying 2,688 differentially expressed genes. Using Mendelian randomization (MR), we identified 74 genes with strong evidence for a causal effect on risk of LUAD. Survival analysis on these genes revealed significant differences in survival rates for 13 of them. Our pathway enrichment analysis highlighted their roles in immune response and cell communication, deepening our understanding. We also utilized single-cell RNA sequencing (scRNA-seq) to uncover cell type-specific gene expression patterns within LUAD, emphasizing the tumor microenvironment's heterogeneity. Pseudotime analysis further assisted in assessing the heterogeneity of tumor cell populations. Additionally, protein-protein interaction (PPI) network analysis was conducted to evaluate the potential druggability of these identified genes. The culmination of our efforts led to the identification of five genes (tier 1) with the most compelling evidence, including SECISBP2L, PRCD, SMAD9, C2orf91, and HSD17B13, and eight genes (tier 2) with convincing evidence for their potential as therapeutic targets.
Collapse
Affiliation(s)
- Youpeng Chen
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Enzhong Li
- Department of Endocrinology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenglin Chang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Tingting Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenfeng Song
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Haojie Wu
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhangkai J. Cheng
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
192
|
Wang H, Nagarajan P, Winkler T, Bentley A, Miller C, Kraja A, Schwander K, Lee S, Wang W, Brown M, Morrison J, Giri A, O'Connell J, Bartz T, de Las Fuentes L, Gudmundsdottir V, Guo X, Harris S, Huang Z, Kals M, Kho M, Lefevre C, Luan J, Lyytikäinen LP, Mangino M, Milaneschi Y, Palmer N, Rao V, Rauramaa R, Shen B, Stadler S, Sun Q, Tang J, Thériault S, van der Graaf A, van der Most P, Wang Y, Weiss S, Westerman K, Yang Q, Yasuharu T, Zhao W, Zhu W, Altschul D, Ansari MAY, Anugu P, Argoty-Pantoja A, Arzt M, Aschard H, Attia J, Bazzano L, Breyer M, Brody J, Cade B, Chen HH, Chen YDI, Chen Z, de Vries P, Dimitrov L, Do A, Du J, Dupont C, Edwards T, Evans M, Faquih T, Felix S, Fisher-Hoch S, Floyd J, Graff M, Charles Gu C, Gu D, Hairston K, Hanley A, Heid I, Heikkinen S, Highland H, Hood M, Kähönen M, Karvonen-Gutierrez C, Kawaguchi T, Kazuya S, Tanika K, Komulainen P, Levy D, Lin H, Liu P, Marques-Vidal P, McCormick J, Mei H, Meigs J, Menni C, Nam K, Nolte I, Pacheco N, Petty L, Polikowsky H, Province M, Psaty B, Raffield L, Raitakari O, et alWang H, Nagarajan P, Winkler T, Bentley A, Miller C, Kraja A, Schwander K, Lee S, Wang W, Brown M, Morrison J, Giri A, O'Connell J, Bartz T, de Las Fuentes L, Gudmundsdottir V, Guo X, Harris S, Huang Z, Kals M, Kho M, Lefevre C, Luan J, Lyytikäinen LP, Mangino M, Milaneschi Y, Palmer N, Rao V, Rauramaa R, Shen B, Stadler S, Sun Q, Tang J, Thériault S, van der Graaf A, van der Most P, Wang Y, Weiss S, Westerman K, Yang Q, Yasuharu T, Zhao W, Zhu W, Altschul D, Ansari MAY, Anugu P, Argoty-Pantoja A, Arzt M, Aschard H, Attia J, Bazzano L, Breyer M, Brody J, Cade B, Chen HH, Chen YDI, Chen Z, de Vries P, Dimitrov L, Do A, Du J, Dupont C, Edwards T, Evans M, Faquih T, Felix S, Fisher-Hoch S, Floyd J, Graff M, Charles Gu C, Gu D, Hairston K, Hanley A, Heid I, Heikkinen S, Highland H, Hood M, Kähönen M, Karvonen-Gutierrez C, Kawaguchi T, Kazuya S, Tanika K, Komulainen P, Levy D, Lin H, Liu P, Marques-Vidal P, McCormick J, Mei H, Meigs J, Menni C, Nam K, Nolte I, Pacheco N, Petty L, Polikowsky H, Province M, Psaty B, Raffield L, Raitakari O, Rich S, Riha R, Risch L, Risch M, Ruiz-Narvaez E, Scott R, Sitlani C, Smith J, Sofer T, Teder-Laving M, Völker U, Vollenweider P, Wang G, van Dijk KWI, Wilson O, Xia R, Yao J, Young K, Zhang R, Zhu X, Below J, Böger C, Conen D, Cox S, Dörr M, Feitosa M, Fox E, Franceschini N, Gharib S, Gudnason V, Harlow S, He J, Holliday E, Kutalik Z, Lakka T, Lawlor D, Lee S, Lehtimäki T, Li C, Liu CT, Mägi R, Matsuda F, Morrison A, Penninx BWJH, Peyser P, Rotter J, Snieder H, Spector T, Wagenknecht L, Wareham N, Zonderman A, North K, Fornage M, Hung A, Manning A, Gauderman W, Chen H, Munroe P, Rao D, van Heemst D, Redline S, Noordam R. A Large-Scale Genome-Wide Study of Gene-Sleep Duration Interactions for Blood Pressure in 811,405 Individuals from Diverse Populations. RESEARCH SQUARE 2024:rs.3.rs-4163414. [PMID: 39070651 PMCID: PMC11276021 DOI: 10.21203/rs.3.rs-4163414/v1] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Although both short and long sleep duration are associated with elevated hypertension risk, our understanding of their interplay with biological pathways governing blood pressure remains limited. To address this, we carried out genome-wide cross-population gene-by-short-sleep and long-sleep duration interaction analyses for three blood pressure traits (systolic, diastolic, and pulse pressure) in 811,405 individuals from diverse population groups. We discover 22 novel gene-sleep duration interaction loci for blood pressure, mapped to 23 genes. Investigating these genes' functional implications shed light on neurological, thyroidal, bone metabolism, and hematopoietic pathways that necessitate future investigation for blood pressure management that caters to sleep health lifestyle. Non-overlap between short sleep (12) and long sleep (10) interactions underscores the plausible nature of distinct influences of both sleep duration extremes in cardiovascular health. Several of our loci are specific towards a particular population background or sex, emphasizing the importance of addressing heterogeneity entangled in gene-environment interactions, when considering precision medicine design approaches for blood pressure management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michael Brown
- The University of Texas Health Science Center at Houston
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Nicholette Palmer
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai
| | | | | | | | | | - Quan Sun
- University of North Carolina, USA
| | | | | | | | | | | | - Stefan Weiss
- University Medicine Greifswald & University of Greifswald
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus
| | | | | | | | | | | | | | | | | | | | | | | | | | - Joseph McCormick
- The University of Texas Health Science Center at Houston (UTHealth) School of Public Health
| | - Hao Mei
- University of Mississippi Medical Center
| | | | | | | | - Ilja Nolte
- University of Groningen, University Medical Center Groningen
| | | | | | | | | | | | | | - Olli Raitakari
- Turku University Hospital and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku
| | | | | | | | | | | | - Rodney Scott
- University of Newcastle and the Hunter Medical Research Institute
| | | | | | | | | | | | | | | | | | | | - Rui Xia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jiang He
- Tulane University School of Public Health and Tropical Medicine
| | | | | | | | | | | | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | | | | | | | | | | | - Patricia Peyser
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Jerome Rotter
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center
| | | | | | | | | | | | | | - Myriam Fornage
- 1. Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center
- 2. Human Genetics Center, Department of Epidemiology, School of Public Health
| | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Lai D, Zhang M, Green N, Abreu M, Schwantes-An TH, Parker C, Zhang S, Jin F, Sun A, Zhang P, Edenberg H, Liu Y, Foroud T. Genome-wide meta-analyses of cross substance use disorders in European, African, and Latino ancestry populations. RESEARCH SQUARE 2024:rs.3.rs-3955955. [PMID: 39070649 PMCID: PMC11275984 DOI: 10.21203/rs.3.rs-3955955/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Genetic risks for substance use disorders (SUDs) are due to both SUD-specific and SUD-shared genes. We performed the largest multivariate analyses to date to search for SUD-shared genes using samples of European (EA), African (AA), and Latino (LA) ancestries. By focusing on variants having cross-SUD and cross-ancestry concordant effects, we identified 45 loci. Through gene-based analyses, gene mapping, and gene prioritization, we identified 250 SUD-shared genes. These genes are highly expressed in amygdala, cortex, hippocampus, hypothalamus, and thalamus, primarily in neuronal cells. Cross-SUD concordant variants explained ~ 50% of the heritability of each SUD in EA. The top 5% individuals having the highest polygenic scores were approximately twice as likely to have SUDs as others in EA and LA. Polygenic scores had higher predictability in females than in males in EA. Using real-world data, we identified five drugs targeting identified SUD-shared genes that may be repurposed to treat SUDs.
Collapse
Affiliation(s)
- Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | | | | | | | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | | | | | | | - Anna Sun
- Indiana University School of Medicine
| | | | | | | | | |
Collapse
|
194
|
Bai Z, Hao J, Chen M, Yao K, Zheng L, Liu L, Hu J, Guo K, Lv Y, Li F. Integrating plasma proteomics with genome-wide association data to identify novel drug targets for inflammatory bowel disease. Sci Rep 2024; 14:16251. [PMID: 39009667 PMCID: PMC11250821 DOI: 10.1038/s41598-024-66780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disease that includes Crohn's disease (CD) and ulcerative colitis (UC). Although genome-wide association studies (GWASs) have identified many relevant genetic risk loci, the impact of these loci on protein abundance and their potential utility as clinical therapeutic targets remain uncertain. Therefore, this study aimed to investigate the pathogenesis of IBD and identify effective therapeutic targets through a comprehensive and integrated analysis. We systematically integrated GWAS data related to IBD, UC and CD (N = 25,305) by the study of de Lange KM with the human blood proteome (N = 7213) by the Atherosclerosis Risk in Communities (ARIC) study. Proteome-wide association study (PWAS), mendelian randomisation (MR) and Bayesian colocalisation analysis were used to identify proteins contributing to the risk of IBD. Integrative analysis revealed that genetic variations in IBD, UC and CD affected the abundance of five (ERAP2, RIPK2, TALDO1, CADM2 and RHOC), three (VSIR, HGFAC and CADM2) and two (MST1 and FLRT3) cis-regulated plasma proteins, respectively (P < 0.05). Among the proteins identified via Bayesian colocalisation analysis, CADM2 was found to be an important common protein between IBD and UC. A drug and five druggable target genes were identified from DGIdb after Bayesian colocalisation analysis. Our study's findings from genetic and proteomic approaches have identified compelling proteins that may serve as important leads for future functional studies and potential drug targets for IBD (UC and CD).
Collapse
Affiliation(s)
- Zhongyuan Bai
- First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Jiawei Hao
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Miaoran Chen
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Kaixin Yao
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Leilei Zheng
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Liu Liu
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Jingxi Hu
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Kaiqing Guo
- Hepatobiliary Pancreatogastric Surgery, Shanxi Province Cancer Hospital, Taiyuan, China.
- Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China.
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Yongqiang Lv
- Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China.
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
- Department of Scientific Research, Shanxi Province Cancer Hospital, Taiyuan, China.
| | - Feng Li
- Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China.
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
- Central Laboratory, Shanxi Province Cancer Hospital, Taiyuan, China.
| |
Collapse
|
195
|
Jiang H, He Y, Lan X, Xie X. Identification and validation of potential common biomarkers for papillary thyroid carcinoma and Hashimoto's thyroiditis through bioinformatics analysis and machine learning. Sci Rep 2024; 14:15578. [PMID: 38971817 PMCID: PMC11227570 DOI: 10.1038/s41598-024-66162-2] [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: 12/20/2023] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
There is a growing body of evidence suggesting that Hashimoto's thyroiditis (HT) may contribute to an increased risk of papillary thyroid carcinoma (PTC). However, the exact relationship between HT and PTC is still not fully understood. The objective of this study was to identify potential common biomarkers that may be associated with both PTC and HT. Three microarray datasets from the GEO database and RNA-seq dataset from TCGA database were collected to identify shared differentially expressed genes (DEGs) between HT and PTC. A total of 101 genes was identified as common DEGs, primarily enriched inflammation- and immune-related pathways through GO and KEGG analysis. We performed protein-protein interaction analysis and identified six significant modules comprising a total of 29 genes. Subsequently, tree hub genes (CD53, FCER1G, TYROBP) were selected using random forest (RF) algorithms for the development of three diagnostic models. The artificial neural network (ANN) model demonstrates superior performance. Notably, CD53 exerted the greatest influence on the ANN model output. We analyzed the protein expressions of the three genes using the Human Protein Atlas database. Moreover, we observed various dysregulated immune cells that were significantly associated with the hub genes through immune infiltration analysis. Immunofluorescence staining confirmed the differential expression of CD53, FCER1G, and TYROBP, as well as the results of immune infiltration analysis. Lastly, we hypothesise that benzylpenicilloyl polylysine and aspirinmay be effective in the treatment of HT and PTC and may prevent HT carcinogenesis. This study indicates that CD53, FCER1G, and TYROBP play a role in the development of HT and PTC, and may contribute to the progression of HT to PTC. These hub genes could potentially serve as diagnostic markers and therapeutic targets for PTC and HT.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Ultrasound, The Second Affiliated Hospital of Anhui Medical Universty, Hefei, 230601, Anhui, China
| | - Yanbin He
- Dian Diagnostics Group Co., Ltd, Hangzhou, 310000, Zhejiang, China
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, 310030, Zhejiang, China
| | - Xiaofeng Lan
- Department of Ultrasound, The Second Affiliated Hospital of Anhui Medical Universty, Hefei, 230601, Anhui, China
| | - Xiang Xie
- Department of Ultrasound, The Second Affiliated Hospital of Anhui Medical Universty, Hefei, 230601, Anhui, China.
- Department of Interventional Ultrasound, The Second Affiliated Hospital of Anhui Medical Universty, Hefei, 230601, Anhui, China.
| |
Collapse
|
196
|
Strom NI, Verhulst B, Bacanu SA, Cheesman R, Purves KL, Gedik H, Mitchell BL, Kwong AS, Faucon AB, Singh K, Medland S, Colodro-Conde L, Krebs K, Hoffmann P, Herms S, Gehlen J, Ripke S, Awasthi S, Palviainen T, Tasanko EM, Peterson RE, Adkins DE, Shabalin AA, Adams MJ, Iveson MH, Campbell A, Thomas LF, Winsvold BS, Drange OK, Børte S, Ter Kuile AR, Nguyen TH, Meier SM, Corfield EC, Hannigan L, Levey DF, Czamara D, Weber H, Choi KW, Pistis G, Couvy-Duchesne B, Van der Auwera S, Teumer A, Karlsson R, Garcia-Argibay M, Lee D, Wang R, Bjerkeset O, Stordal E, Bäckmann J, Salum GA, Zai CC, Kennedy JL, Zai G, Tiwari AK, Heilmann-Heimbach S, Schmidt B, Kaprio J, Kennedy MM, Boden J, Havdahl A, Middeldorp CM, Lopes FL, Akula N, McMahon FJ, Binder EB, Fehm L, Ströhle A, Castelao E, Tiemeier H, Stein DJ, Whiteman D, Olsen C, Fuller Z, Wang X, Wray NR, Byrne EM, Lewis G, Timpson NJ, Davis LK, Hickie IB, Gillespie NA, Milani L, Schumacher J, Woldbye DP, Forstner AJ, Nöthen MM, Hovatta I, Horwood J, Copeland WE, Maes HH, McIntosh AM, Andreassen OA, Zwart JA, Mors O, Børglum AD, Mortensen PB, Ask H, Reichborn-Kjennerud T, Najman JM, et alStrom NI, Verhulst B, Bacanu SA, Cheesman R, Purves KL, Gedik H, Mitchell BL, Kwong AS, Faucon AB, Singh K, Medland S, Colodro-Conde L, Krebs K, Hoffmann P, Herms S, Gehlen J, Ripke S, Awasthi S, Palviainen T, Tasanko EM, Peterson RE, Adkins DE, Shabalin AA, Adams MJ, Iveson MH, Campbell A, Thomas LF, Winsvold BS, Drange OK, Børte S, Ter Kuile AR, Nguyen TH, Meier SM, Corfield EC, Hannigan L, Levey DF, Czamara D, Weber H, Choi KW, Pistis G, Couvy-Duchesne B, Van der Auwera S, Teumer A, Karlsson R, Garcia-Argibay M, Lee D, Wang R, Bjerkeset O, Stordal E, Bäckmann J, Salum GA, Zai CC, Kennedy JL, Zai G, Tiwari AK, Heilmann-Heimbach S, Schmidt B, Kaprio J, Kennedy MM, Boden J, Havdahl A, Middeldorp CM, Lopes FL, Akula N, McMahon FJ, Binder EB, Fehm L, Ströhle A, Castelao E, Tiemeier H, Stein DJ, Whiteman D, Olsen C, Fuller Z, Wang X, Wray NR, Byrne EM, Lewis G, Timpson NJ, Davis LK, Hickie IB, Gillespie NA, Milani L, Schumacher J, Woldbye DP, Forstner AJ, Nöthen MM, Hovatta I, Horwood J, Copeland WE, Maes HH, McIntosh AM, Andreassen OA, Zwart JA, Mors O, Børglum AD, Mortensen PB, Ask H, Reichborn-Kjennerud T, Najman JM, Stein MB, Gelernter J, Milaneschi Y, Penninx BW, Boomsma DI, Maron E, Erhardt-Lehmann A, Rück C, Kircher TT, Melzig CA, Alpers GW, Arolt V, Domschke K, Smoller JW, Preisig M, Martin NG, Lupton MK, Luik AI, Reif A, Grabe HJ, Larsson H, Magnusson PK, Oldehinkel AJ, Hartman CA, Breen G, Docherty AR, Coon H, Conrad R, Lehto K, Deckert J, Eley TC, Mattheisen M, Hettema JM. Genome-wide association study of major anxiety disorders in 122,341 European-ancestry cases identifies 58 loci and highlights GABAergic signaling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.03.24309466. [PMID: 39006447 PMCID: PMC11245051 DOI: 10.1101/2024.07.03.24309466] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The major anxiety disorders (ANX; including generalized anxiety disorder, panic disorder, and phobias) are highly prevalent, often onset early, persist throughout life, and cause substantial global disability. Although distinct in their clinical presentations, they likely represent differential expressions of a dysregulated threat-response system. Here we present a genome-wide association meta-analysis comprising 122,341 European ancestry ANX cases and 729,881 controls. We identified 58 independent genome-wide significant ANX risk variants and 66 genes with robust biological support. In an independent sample of 1,175,012 self-report ANX cases and 1,956,379 controls, 51 of the 58 associated variants were replicated. As predicted by twin studies, we found substantial genetic correlation between ANX and depression, neuroticism, and other internalizing phenotypes. Follow-up analyses demonstrated enrichment in all major brain regions and highlighted GABAergic signaling as one potential mechanism underlying ANX genetic risk. These results advance our understanding of the genetic architecture of ANX and prioritize genes for functional follow-up studies.
Collapse
Affiliation(s)
- Nora I Strom
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Brad Verhulst
- Psychiatry and Behavioral Sciences, Texas A&M University, College Station, Texas, USA
| | | | - Rosa Cheesman
- PROMENTA Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Kirstin L Purves
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Hüseyin Gedik
- Institute for Genomics in Health, Department of Psychiatry and Behavioral Sciences, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Life Sciences, Integrative Life Sciences Doctoral Program, Virginia Commonwealth University, Richmond, Virginia, USA
- Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Brittany L Mitchell
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, Queensland University , Brisbane, Queensland, Australia
| | - Alex S Kwong
- Bristol Medical School, Population Health Sciences, MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Annika B Faucon
- Division of Medicine, Human Genetics, Vanderbilt University, Nashville, Tennessee, USA
| | - Kritika Singh
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sarah Medland
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lucia Colodro-Conde
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Department of Biomedicine, Human Genomics Research Group, University of Basel; University Hospital Basel, Basel, Switzerland
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Medical Genetics and Pathology, Medical Faculty, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, Human Genomics Research Group, University of Basel; University Hospital Basel, Basel, Switzerland
| | - Jan Gehlen
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Stephan Ripke
- Dept. of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Swapnil Awasthi
- Dept. of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Teemu Palviainen
- Helsinki Institute of Life Science, Institute for Molecular Medicine Finland - FIMM, University of Helsinki, Helsinki, Finland
| | - Elisa M Tasanko
- Faculty of Medicine, Department of Psychology and Logopedics, SleepWell Research Program, University of Helsinki, Helsinki, Finland
| | - Roseann E Peterson
- Institute for Genomics in Health, Department of Psychiatry and Behavioral Sciences, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Psychiatry, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Daniel E Adkins
- School of Medicine, Department of Psychiatry, University of Utah, Salt Lake City, Utah, USA
| | - Andrey A Shabalin
- School of Medicine, Department of Psychiatry, University of Utah, Salt Lake City, Utah, USA
| | - Mark J Adams
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Matthew H Iveson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- College of Medicine and Veterinary Medicine, Institute of Genetics and Cancer; Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Laurent F Thomas
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Bendik S Winsvold
- Division of Clinical Neuroscience, Department of Research and Innovation, Oslo University Hospital, Oslo, Norway
- Department of Public Health and Nursing, HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Ole Kristian Drange
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
- Division of Mental Health, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- NORMENT Centre, University of Oslo, Oslo, Norway
- Centre of Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Psychiatry, Sørlandet Hospital, Kristiansand, Norway
| | - Sigrid Børte
- Division of Clinical Neuroscience, Department of Research and Innovation; Musculoskeletal Health, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Public Health and Nursing, HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Abigail R Ter Kuile
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
- Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom
| | - Tan-Hoang Nguyen
- Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sandra M Meier
- Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Elizabeth C Corfield
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute , Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Laurie Hannigan
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Daniel F Levey
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- Psychiatry, Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Darina Czamara
- Department of Genes and Environment, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Heike Weber
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - Karmel W Choi
- Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
- Psychiatry, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Giorgio Pistis
- Psychiatric Epidemiology and Psychopathology Research Center, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Prilly, Switzerland
| | - Baptiste Couvy-Duchesne
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- ARAMIS laboratory, Paris Brain Institute, Paris, France
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Miguel Garcia-Argibay
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Donghyung Lee
- Department of Statistics, Miami University, Oxford, Ohio, USA
| | - Rujia Wang
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ottar Bjerkeset
- Faculty of Nursing and Health Science, Nord University, Levanger, Norway
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Eystein Stordal
- Department of Psychiatry, Hospital Namsos, Nord-Trøndelag Health Trustt, Namsos, Norway
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Julia Bäckmann
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Giovanni A Salum
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Child Psychiatry, National Institute of Developmental Psychiatry, São Paulo, Brazil
| | - Clement C Zai
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Sciences Department, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Division of Neurosciences and Clinical Translation, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - James L Kennedy
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Sciences Department, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Division of Neurosciences and Clinical Translation, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Gwyneth Zai
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Sciences Department, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Division of Neurosciences and Clinical Translation, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Arun K Tiwari
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Sciences Department, Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Division of Neurosciences and Clinical Translation, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Börge Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Jaakko Kaprio
- Helsinki Institute of Life Science, Institute for Molecular Medicine Finland - FIMM, University of Helsinki, Helsinki, Finland
| | - Martin M Kennedy
- Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Joseph Boden
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Alexandra Havdahl
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- PROMENTA Centre, Department of Psychology, University of Oslo, Oslo, Norway
- Bristol Medical School, Population Health Sciences, MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Christel M Middeldorp
- Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, Queensland, Australia
| | - Fabiana L Lopes
- National Institute of Mental Health, Human Genetics Branch, National Institutes of Health, Bethesda, Maryland, USA
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Nirmala Akula
- National Institute of Mental Health, Genetic Basis of Mood and Anxiety Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Francis J McMahon
- National Institute of Mental Health, Genetic Basis of Mood and Anxiety Disorders, National Institutes of Health, Bethesda, Maryland, USA
- Psychiatry & Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Elisabeth B Binder
- Department of Genes and Environment, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Lydia Fehm
- Department of Psychology, Zentrum für Psychotherapie, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Ströhle
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Enrique Castelao
- Psychiatric Epidemiology and Psychopathology Research Center, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Prilly, Switzerland
| | - Henning Tiemeier
- Social and Behavioral Science, T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Child and Adolescent Psychiatry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dan J Stein
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - David Whiteman
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Catherine Olsen
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Enda M Byrne
- Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Glyn Lewis
- UCL Division of Psychiatry, University College London, London, UK
| | - Nicholas J Timpson
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
- Bristol Medical School, Population Health Sciences, MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Lea K Davis
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Sydney, Australia
| | | | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - David P Woldbye
- Department of Neuroscience, Laboratory of Neural Plasticity, University of Copenhagen, Copenhagen, Denmark
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Iiris Hovatta
- Faculty of Medicine, Department of Psychology and Logopedics and SleepWell Research Program, University of Helsinki, Helsinki, Finland
| | - John Horwood
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - William E Copeland
- UVM Medical Center, Department of Psychiatry, University of Vermont, Burlington, Vermont, USA
| | - Hermine H Maes
- Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
- Psychiatry, Virginia Commonwealth University, Richmond, Virginia, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Andrew M McIntosh
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ole A Andreassen
- NORMENT Centre, University of Oslo, Oslo, Norway
- Centre of Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - John-Anker Zwart
- Division of Clinical Neuroscience, Department of Research and Innovation; Musculoskeletal Health, Oslo University Hospital, Oslo, Norway
- Department of Public Health and Nursing, HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Mors
- Department of Psychiatry, Psychosis Research Unit, Aarhus University Hospital, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus University, Aarhus, Denmark
| | - Anders D Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalised Medicine, Aarhus University, Aarhus, Denmark
| | - Preben B Mortensen
- The National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Helga Ask
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Ted Reichborn-Kjennerud
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- NORMENT Centre, University of Oslo, Oslo, Norway
| | - Jackob M Najman
- Faculty of Medicine, School of Public Health, University of Queensland, Herston, Queensland, Australia
| | - Murray B Stein
- Psychiatry, University of California San Diego, La Jolla, CA, USA
- School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Joel Gelernter
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- Psychiatry Research, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
- Departments of Genetics and Neuroscience, Yale University of Medicine, New Haven, Connecticut, USA
| | - Yuri Milaneschi
- Amsterdam Neuroscience; Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Brenda W Penninx
- Amsterdam Neuroscience; Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Dorret I Boomsma
- Twin Register and Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Eduard Maron
- Psychiatry, University of Tartu, Tartu, Estonia
- Department of Medicine, Centre for Neuropsychopharmacology,, Division of Brain Sciences, Imperial College London, London, UK
| | - Angelika Erhardt-Lehmann
- Department of Genes and Environment, Max-Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Christian Rück
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Tilo T Kircher
- Department of Psychiatry, University of Marburg, Marburg, Germany
| | - Christiane A Melzig
- Psychology, Clinical Psychology, Experimental Psychopathology and Psychotherapy, University of Marburg, Marburg, Germany
- Psychology, Biological and Clinical Psychology, University of Greifswald, Greifswald, Germany
| | - Georg W Alpers
- School of Social Sciences, Department of Psychology, University of Mannheim, Mannheim, Germany
| | - Volker Arolt
- Department of Mental Health, Institute for Translational Psychiatry, University of Muenster, Muenster, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Center for Mental Health (DZPG), Partner Site Berlin, Berlin, Germany
| | - Jordan W Smoller
- Psychiatry, Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Martin Preisig
- Psychiatric Epidemiology and Psychopathology Research Center, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Prilly, Switzerland
| | - Nicholas G Martin
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Michelle K Lupton
- Brain and Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, Queensland University , Brisbane, Queensland, Australia
- Faculty of Health, Queensland University of technology, Queensland, Australia
| | - Annemarie I Luik
- Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt - Goethe University, Frankfurt, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Henrik Larsson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik K Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Albertine J Oldehinkel
- Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Catharina A Hartman
- Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anna R Docherty
- School of Medicine, Psychiatry, University of Utah, Salt Lake City, Utah, USA
- School of Medicine, Psychiatry; Huntsman Mental Health Institute, University of Utah, Salt Lake City, Utah, USA
- Psychiatry, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Hilary Coon
- School of Medicine, Psychiatry, University of Utah, Salt Lake City, Utah, USA
| | - Rupert Conrad
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Münster, Münster, Germany
| | - Kelli Lehto
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Thalia C Eley
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Manuel Mattheisen
- Community Health and Epidemiology, Dalhousie University, Halifax, Nova Scotia, Canada
- Computer Science, Dalhousie University, Halifax, Nova Scotia, Canada
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| | - John M Hettema
- Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas, USA
| |
Collapse
|
197
|
Tao T, Mo X, Zhao L. Identifying novel potential drug targets for endometriosis via plasma proteome screening. Front Endocrinol (Lausanne) 2024; 15:1416978. [PMID: 39036049 PMCID: PMC11257892 DOI: 10.3389/fendo.2024.1416978] [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: 04/13/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Background Endometriosis (EM) is a chronic painful condition that predominantly affects women of reproductive age. Currently, surgery or medication can only provide limited symptom relief. This study used a comprehensive genetic analytical approach to explore potential drug targets for EM in the plasma proteome. Methods In this study, 2,923 plasma proteins were selected as exposure and EM as outcome for two-sample Mendelian randomization (MR) analyses. The plasma proteomic data were derived from the UK Biobank Pharmaceutical Proteomics Project (UKB-PPP), while the EM dataset from the FinnGen consortium R10 release data. Several sensitivity analyses were performed, including summary-data-based MR (SMR) analyses, heterogeneity in dependent instruments (HEIDI) test, reverse MR analyses, steiger detection test, and bayesian co-localization analyses. Furthermore, proteome-wide association study (PWAS) and single-cell transcriptomic analyses were also conducted to validate the findings. Results Six significant (p < 3.06 × 10-5) plasma protein-EM pairs were identified by MR analyses. These included EPHB4 (OR = 1.40, 95% CI: 1.20 - 1.63), FSHB (OR = 3.91, 95% CI: 3.13 - 4.87), RSPO3 (OR = 1.60, 95% CI: 1.38 - 1.86), SEZ6L2 (OR = 1.44, 95% CI: 1.23 - 1.68) and WASHC3 (OR = 2.00, 95% CI: 1.54 - 2.59) were identified as risk factors, whereas KDR (OR = 0.80, 95% CI: 0.75 - 0.90) was found to be a protective factor. All six plasma proteins passed the SMR test (P < 8.33 × 10-3), but only four plasma proteins passed the HEIDI heterogeneity test (PHEIDI > 0.05), namely FSHB, RSPO3, SEZ6L2 and EPHB4. These four proteins showed strong evidence of co-localization (PPH4 > 0.7). In particular, RSPO3 and EPHB4 were replicated in the validated PWAS. Single-cell analyses revealed high expression of SEZ6L2 and EPHB4 in stromal and epithelial cells within EM lesions, while RSPO3 exhibited elevated expression in stromal cells and fibroblasts. Conclusion Our study identified FSHB, RSPO3, SEZ6L2, and EPHB4 as potential drug targets for EM and highlighted the critical role of stromal and epithelial cells in disease development. These findings provide new insights into the diagnosis and treatment of EM.
Collapse
Affiliation(s)
- Tian Tao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Mo
- Department of Gynaecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Liangbin Zhao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
198
|
Li YS, Jiang HC. Integrating molecular pathway with genome-wide association data for causality identification in breast cancer. Discov Oncol 2024; 15:254. [PMID: 38954227 PMCID: PMC11219684 DOI: 10.1007/s12672-024-01125-7] [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: 05/21/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE The study purpose was to explore the causal association between pyruvate metabolism and breast cancer (BC), as well as the molecular role of key metabolic genes, by using bioinformatics and Mendelian randomization (MR) analysis. METHODS We retrieved and examined diverse datasets from the GEO database to ascertain differentially acting genes (DAGs) in BC via differential expression analysis. Following this, we performed functional and pathway enrichment analyses to ascertain noteworthy molecular functions and metabolic pathways in BC. Employing MR analysis, we established a causal association between pyruvate metabolism and the susceptibility to BC. Additionally, utilizing the DGIdb database, we identified potential targeted medications that act on genes implicated in the pyruvate metabolic pathway and formulated a competing endogenous RNA (ceRNA) regulatory network in BC. RESULTS We collected the datasets GSE54002, GSE70947, and GSE22820, and identified a total of 1127 DEGs between the BC and NC groups. GO and KEGG enrichment analysis showed that the molecular functions of these DEGs mainly included mitotic nuclear division, extracellular matrix, signaling receptor activator activity, etc. Metabolic pathways were mainly concentrated in PI3K-Akt signaling pathway, Cytokine-cytokine receptor binding and Pyruvate, Tyrosine, Propanoate and Phenylalanine metabolism, etc. In addition, MR analysis demonstrated a causal relationship between pyruvate metabolism and BC risk. Finally, we constructed a regulatory network between pathway genes (ADH1B, ACSS2, ACACB, ADH1A, ALDH2, and ADH1C) and targeted drugs, as well as a ceRNA (lncRNA-miRNA-mRNA) regulatory network for BC, further revealing their interactions. CONCLUSIONS Our research revealed a causal association between pyruvate metabolism and BC risk, found that ADH1B, ACSS2, ACACB, ADH1A, ALDH2, and ADH1C takes place an important part in the development of BC in the molecular mechanisms related to pyruvate metabolism, and identified some potential targeted small molecule drugs.
Collapse
Affiliation(s)
- Yan-Shuang Li
- Department of Breast Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hong-Chuan Jiang
- Department of Breast Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
199
|
Kahnert K, Soattin L, Mills RW, Wilson C, Maurya S, Sorrentino A, Al-Othman S, Tikhomirov R, van de Vegte YJ, Hansen FB, Achter J, Hu W, Zi M, Smith M, van der Harst P, Olesen MS, Boisen Olsen K, Banner J, Jensen THL, Zhang H, Boyett MR, D’Souza A, Lundby A. Proteomics couples electrical remodelling to inflammation in a murine model of heart failure with sinus node dysfunction. Cardiovasc Res 2024; 120:927-942. [PMID: 38661182 PMCID: PMC11218694 DOI: 10.1093/cvr/cvae054] [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: 11/01/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 04/26/2024] Open
Abstract
AIMS In patients with heart failure (HF), concomitant sinus node dysfunction (SND) is an important predictor of mortality, yet its molecular underpinnings are poorly understood. Using proteomics, this study aimed to dissect the protein and phosphorylation remodelling within the sinus node in an animal model of HF with concurrent SND. METHODS AND RESULTS We acquired deep sinus node proteomes and phosphoproteomes in mice with heart failure and SND and report extensive remodelling. Intersecting the measured (phospho)proteome changes with human genomics pharmacovigilance data, highlighted downregulated proteins involved in electrical activity such as the pacemaker ion channel, Hcn4. We confirmed the importance of ion channel downregulation for sinus node physiology using computer modelling. Guided by the proteomics data, we hypothesized that an inflammatory response may drive the electrophysiological remodeling underlying SND in heart failure. In support of this, experimentally induced inflammation downregulated Hcn4 and slowed pacemaking in the isolated sinus node. From the proteomics data we identified proinflammatory cytokine-like protein galectin-3 as a potential target to mitigate the effect. Indeed, in vivo suppression of galectin-3 in the animal model of heart failure prevented SND. CONCLUSION Collectively, we outline the protein and phosphorylation remodeling of SND in heart failure, we highlight a role for inflammation in electrophysiological remodelling of the sinus node, and we present galectin-3 signalling as a target to ameliorate SND in heart failure.
Collapse
Affiliation(s)
- Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Luca Soattin
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Robert W Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Claire Wilson
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Svetlana Maurya
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Sami Al-Othman
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Roman Tikhomirov
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), 72 Du Cane Road, London W12 0NN, UK
| | - Yordi J van de Vegte
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Finn B Hansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Jonathan Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Wei Hu
- Department of Physics & Astronomy, Biological Physics Group, University of Manchester, Manchester, UK
| | - Min Zi
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Matthew Smith
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), 72 Du Cane Road, London W12 0NN, UK
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, Netherlands Heart Institute, Utrecht, the Netherlands
| | - Morten S Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Kristine Boisen Olsen
- Department of Forensic Medicine, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Jytte Banner
- Department of Forensic Medicine, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | | | - Henggui Zhang
- Department of Physics & Astronomy, Biological Physics Group, University of Manchester, Manchester, UK
| | - Mark R Boyett
- Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Alicia D’Souza
- Division of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), 72 Du Cane Road, London W12 0NN, UK
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| |
Collapse
|
200
|
Zhao H, Liu Y, Zhang X, Liao Y, Zhang H, Han X, Guo L, Fan B, Wang W, Lu C. Identifying novel proteins for suicide attempt by integrating proteomes from brain and blood with genome-wide association data. Neuropsychopharmacology 2024; 49:1255-1265. [PMID: 38317018 PMCID: PMC11224332 DOI: 10.1038/s41386-024-01807-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/28/2023] [Accepted: 01/16/2024] [Indexed: 02/07/2024]
Abstract
Genome-wide association studies (GWASs) have identified risk loci for suicide attempt (SA), but deciphering how they confer risk for SA remains largely unknown. This study aims to identify the key proteins and gain insights into SA pathogenesis. We integrated data from the brain proteome (N = 376) and blood proteome (N = 35,559) and combined it with the largest SA GWAS summary statistics to date (N = 518,612). A comprehensive set of methods was employed, including Mendelian randomization (MR), Steiger filtering, Bayesian colocalization, proteome‑wide association studies (PWAS), transcript-levels, cell-type specificity, correlation, and protein-protein interaction (PPI) network analysis. Validation was performed using other protein datasets and the SA dataset from FinnGen study. We identified ten proteins (GLRX5, GMPPB, B3GALTL, FUCA2, TTLL12, ADCK1, MMAA, HIBADH, ACP1, DOC2A) associated with SA in brain proteomics. GLRX5, GMPPB, and FUCA2 showed strong colocalization evidence and were supported by PWAS and transcript-level analysis, and were predominantly expressed in glutamatergic neuronal cells. In blood proteomics, one significant protein (PEAR1) and three near-significant proteins (NDE1, EVA1C, B4GALT2) were identified, but lacked colocalization evidence. Moreover, despite the limited correlation between the same protein in brain and blood, the PPI network analysis provided new insights into the interaction between brain and blood in SA. Furthermore, GLRX5 was associated with the GSTP1, the target of Clozapine. The comprehensive analysis provides strong evidence supporting a causal association between three genetically determined brain proteins (GLRX5, GMPPB, and FUCA2) with SA. These findings offer valuable insights into SA's underlying mechanisms and potential therapeutic approaches.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, China
| | - Yifeng Liu
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Xuening Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuhua Liao
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Huimin Zhang
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Xue Han
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, China
| | - Beifang Fan
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China.
| | - Wanxin Wang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, China.
| | - Ciyong Lu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|