1
|
Berghöfer J, Khaveh N, Mundlos S, Metzger J. Multi-tool copy number detection highlights common body size-associated variants in miniature pig breeds from different geographical regions. BMC Genomics 2025; 26:285. [PMID: 40121435 PMCID: PMC11929999 DOI: 10.1186/s12864-025-11446-8] [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: 10/30/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Copy number variations (CNVs) represent a common and highly specific type of variation in the genome, potentially influencing genetic diversity and mammalian phenotypic development. Structural variants, such as deletions, duplications, and insertions, have frequently been highlighted as key factors influencing traits in high-production pigs. However, comprehensive CNV analyses in miniature pig breeds are limited despite their value in biomedical research. RESULTS This study performed whole-genome sequencing in 36 miniature pigs from nine breeds from America, Asia and Oceania, and Europe. By employing a multi-tool approach (CNVpytor, Delly, GATK gCNV, Smoove), the accuracy of CNV identification was improved. In total, 34 homozygous CNVs overlapped with exonic regions in all samples, suggesting a role in expressing specific phenotypes such as uniform growth patterns, fertility, or metabolic function. In addition, 386 copy number variation regions (CNVRs) shared by all breeds were detected, covering 33.6 Mb (1.48% of the autosomal genome). Further, 132 exclusive CNVRs were identified for American breeds, 47 for Asian and Oceanian breeds, and 114 for European breeds. Functional enrichment analysis revealed genes within the common CNVRs involved in body height determination and other growth-related parameters. Exclusive CNVRs were located in the region of genes enriched for lipid metabolism in American minipigs, reproductive traits in Asian and Oceanian breeds, and cardiovascular features and body height in European breeds. In the selected groups, quantitative trait loci associated with body size, meat quality, reproduction, and disease susceptibility were highlighted. CONCLUSION This investigation of the CNV landscape of minipigs underlines the impact of selective breeding on structural variants and its role in the development of specific breed phenotypes across geographical areas. The multi-tool approach provides a valuable resource for future studies on the effects of artificial selection on livestock genomes.
Collapse
Affiliation(s)
- Jan Berghöfer
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
- Institute of Animal Genomics, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Nadia Khaveh
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Animal Genomics, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute for Medical and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Centre for Regenerative Therapies, Berlin, Germany
| | - Julia Metzger
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
- Institute of Animal Genomics, University of Veterinary Medicine Hanover, Hanover, Germany.
| |
Collapse
|
2
|
Duyzend MH, Cacheiro P, Jacobsen JO, Giordano J, Brand H, Wapner RJ, Talkowski ME, Robinson PN, Smedley D. Improving prenatal diagnosis through standards and aggregation. Prenat Diagn 2024; 44:454-464. [PMID: 38242839 PMCID: PMC11006584 DOI: 10.1002/pd.6522] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/21/2024]
Abstract
Advances in sequencing and imaging technologies enable enhanced assessment in the prenatal space, with a goal to diagnose and predict the natural history of disease, to direct targeted therapies, and to implement clinical management, including transfer of care, election of supportive care, and selection of surgical interventions. The current lack of standardization and aggregation stymies variant interpretation and gene discovery, which hinders the provision of prenatal precision medicine, leaving clinicians and patients without an accurate diagnosis. With large amounts of data generated, it is imperative to establish standards for data collection, processing, and aggregation. Aggregated and homogeneously processed genetic and phenotypic data permits dissection of the genomic architecture of prenatal presentations of disease and provides a dataset on which data analysis algorithms can be tuned to the prenatal space. Here we discuss the importance of generating aggregate data sets and how the prenatal space is driving the development of interoperable standards and phenotype-driven tools.
Collapse
Affiliation(s)
- Michael H. Duyzend
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Pilar Cacheiro
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Julius O.B. Jacobsen
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jessica Giordano
- Department of Obstetrics & Gynecology, Columbia University Medical Center, New York, NY, USA
| | - Harrison Brand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Ronald J. Wapner
- Department of Obstetrics & Gynecology, Columbia University Medical Center, New York, NY, USA
| | - Michael E. Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
- Program in Bioinformatics and Integrative Genomics, Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Peter N. Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| | - Damian Smedley
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
3
|
Abu-Toamih-Atamni HJ, Lone IM, Binenbaum I, Mott R, Pilalis E, Chatziioannou A, Iraqi FA. Mapping novel QTL and fine mapping of previously identified QTL associated with glucose tolerance using the collaborative cross mice. Mamm Genome 2024; 35:31-55. [PMID: 37978084 DOI: 10.1007/s00335-023-10025-0] [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: 07/08/2023] [Accepted: 10/08/2023] [Indexed: 11/19/2023]
Abstract
A chronic metabolic illness, type 2 diabetes (T2D) is a polygenic and multifactorial complicated disease. With an estimated 463 million persons aged 20 to 79 having diabetes, the number is expected to rise to 700 million by 2045, creating a significant worldwide health burden. Polygenic variants of diabetes are influenced by environmental variables. T2D is regarded as a silent illness that can advance for years before being diagnosed. Finding genetic markers for T2D and metabolic syndrome in groups with similar environmental exposure is therefore essential to understanding the mechanism of such complex characteristic illnesses. So herein, we demonstrated the exclusive use of the collaborative cross (CC) mouse reference population to identify novel quantitative trait loci (QTL) and, subsequently, suggested genes associated with host glucose tolerance in response to a high-fat diet. In this study, we used 539 mice from 60 different CC lines. The diabetogenic effect in response to high-fat dietary challenge was measured by the three-hour intraperitoneal glucose tolerance test (IPGTT) test after 12 weeks of dietary challenge. Data analysis was performed using a statistical software package IBM SPSS Statistic 23. Afterward, blood glucose concentration at the specific and between different time points during the IPGTT assay and the total area under the curve (AUC0-180) of the glucose clearance was computed and utilized as a marker for the presence and severity of diabetes. The observed AUC0-180 averages for males and females were 51,267.5 and 36,537.5 mg/dL, respectively, representing a 1.4-fold difference in favor of females with lower AUC0-180 indicating adequate glucose clearance. The AUC0-180 mean differences between the sexes within each specific CC line varied widely within the CC population. A total of 46 QTL associated with the different studied phenotypes, designated as T2DSL and its number, for Type 2 Diabetes Specific Locus and its number, were identified during our study, among which 19 QTL were not previously mapped. The genomic interval of the remaining 27 QTL previously reported, were fine mapped in our study. The genomic positions of 40 of the mapped QTL overlapped (clustered) on 11 different peaks or close genomic positions, while the remaining 6 QTL were unique. Further, our study showed a complex pattern of haplotype effects of the founders, with the wild-derived strains (mainly PWK) playing a significant role in the increase of AUC values.
Collapse
Affiliation(s)
- Hanifa J Abu-Toamih-Atamni
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel
| | - Iqbal M Lone
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel
| | - Ilona Binenbaum
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou Str, 11527, Athens, Greece
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Richard Mott
- Department of Genetics, University College of London, London, UK
| | | | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou Str, 11527, Athens, Greece
- e-NIOS Applications PC, 196 Syggrou Ave., 17671, Kallithea, Greece
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel.
| |
Collapse
|
4
|
Gargano MA, Matentzoglu N, Coleman B, Addo-Lartey EB, Anagnostopoulos A, Anderton J, Avillach P, Bagley AM, Bakštein E, Balhoff JP, Baynam G, Bello SM, Berk M, Bertram H, Bishop S, Blau H, Bodenstein DF, Botas P, Boztug K, Čady J, Callahan TJ, Cameron R, Carbon S, Castellanos F, Caufield JH, Chan LE, Chute C, Cruz-Rojo J, Dahan-Oliel N, Davids JR, de Dieuleveult M, de Souza V, de Vries BBA, de Vries E, DePaulo JR, Derfalvi B, Dhombres F, Diaz-Byrd C, Dingemans AJM, Donadille B, Duyzend M, Elfeky R, Essaid S, Fabrizzi C, Fico G, Firth HV, Freudenberg-Hua Y, Fullerton JM, Gabriel DL, Gilmour K, Giordano J, Goes FS, Moses RG, Green I, Griese M, Groza T, Gu W, Guthrie J, Gyori B, Hamosh A, Hanauer M, Hanušová K, He Y(O, Hegde H, Helbig I, Holasová K, Hoyt CT, Huang S, Hurwitz E, Jacobsen JOB, Jiang X, Joseph L, Keramatian K, King B, Knoflach K, Koolen DA, Kraus M, Kroll C, Kusters M, Ladewig MS, Lagorce D, Lai MC, Lapunzina P, Laraway B, Lewis-Smith D, Li X, Lucano C, Majd M, Marazita ML, Martinez-Glez V, McHenry TH, McInnis MG, McMurry JA, Mihulová M, Millett CE, Mitchell PB, Moslerová V, Narutomi K, Nematollahi S, Nevado J, et alGargano MA, Matentzoglu N, Coleman B, Addo-Lartey EB, Anagnostopoulos A, Anderton J, Avillach P, Bagley AM, Bakštein E, Balhoff JP, Baynam G, Bello SM, Berk M, Bertram H, Bishop S, Blau H, Bodenstein DF, Botas P, Boztug K, Čady J, Callahan TJ, Cameron R, Carbon S, Castellanos F, Caufield JH, Chan LE, Chute C, Cruz-Rojo J, Dahan-Oliel N, Davids JR, de Dieuleveult M, de Souza V, de Vries BBA, de Vries E, DePaulo JR, Derfalvi B, Dhombres F, Diaz-Byrd C, Dingemans AJM, Donadille B, Duyzend M, Elfeky R, Essaid S, Fabrizzi C, Fico G, Firth HV, Freudenberg-Hua Y, Fullerton JM, Gabriel DL, Gilmour K, Giordano J, Goes FS, Moses RG, Green I, Griese M, Groza T, Gu W, Guthrie J, Gyori B, Hamosh A, Hanauer M, Hanušová K, He Y(O, Hegde H, Helbig I, Holasová K, Hoyt CT, Huang S, Hurwitz E, Jacobsen JOB, Jiang X, Joseph L, Keramatian K, King B, Knoflach K, Koolen DA, Kraus M, Kroll C, Kusters M, Ladewig MS, Lagorce D, Lai MC, Lapunzina P, Laraway B, Lewis-Smith D, Li X, Lucano C, Majd M, Marazita ML, Martinez-Glez V, McHenry TH, McInnis MG, McMurry JA, Mihulová M, Millett CE, Mitchell PB, Moslerová V, Narutomi K, Nematollahi S, Nevado J, Nierenberg AA, Čajbiková NN, Nurnberger JI, Ogishima S, Olson D, Ortiz A, Pachajoa H, Perez de Nanclares G, Peters A, Putman T, Rapp CK, Rath A, Reese J, Rekerle L, Roberts A, Roy S, Sanders SJ, Schuetz C, Schulte EC, Schulze TG, Schwarz M, Scott K, Seelow D, Seitz B, Shen Y, Similuk MN, Simon ES, Singh B, Smedley D, Smith CL, Smolinsky JT, Sperry S, Stafford E, Stefancsik R, Steinhaus R, Strawbridge R, Sundaramurthi JC, Talapova P, Tenorio Castano JA, Tesner P, Thomas RH, Thurm A, Turnovec M, van Gijn ME, Vasilevsky NA, Vlčková M, Walden A, Wang K, Wapner R, Ware JS, Wiafe AA, Wiafe SA, Wiggins LD, Williams AE, Wu C, Wyrwoll MJ, Xiong H, Yalin N, Yamamoto Y, Yatham LN, Yocum AK, Young AH, Yüksel Z, Zandi PP, Zankl A, Zarante I, Zvolský M, Toro S, Carmody LC, Harris NL, Munoz-Torres MC, Danis D, Mungall CJ, Köhler S, Haendel MA, Robinson PN. The Human Phenotype Ontology in 2024: phenotypes around the world. Nucleic Acids Res 2024; 52:D1333-D1346. [PMID: 37953324 PMCID: PMC10767975 DOI: 10.1093/nar/gkad1005] [Show More Authors] [Citation(s) in RCA: 89] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023] Open
Abstract
The Human Phenotype Ontology (HPO) is a widely used resource that comprehensively organizes and defines the phenotypic features of human disease, enabling computational inference and supporting genomic and phenotypic analyses through semantic similarity and machine learning algorithms. The HPO has widespread applications in clinical diagnostics and translational research, including genomic diagnostics, gene-disease discovery, and cohort analytics. In recent years, groups around the world have developed translations of the HPO from English to other languages, and the HPO browser has been internationalized, allowing users to view HPO term labels and in many cases synonyms and definitions in ten languages in addition to English. Since our last report, a total of 2239 new HPO terms and 49235 new HPO annotations were developed, many in collaboration with external groups in the fields of psychiatry, arthrogryposis, immunology and cardiology. The Medical Action Ontology (MAxO) is a new effort to model treatments and other measures taken for clinical management. Finally, the HPO consortium is contributing to efforts to integrate the HPO and the GA4GH Phenopacket Schema into electronic health records (EHRs) with the goal of more standardized and computable integration of rare disease data in EHRs.
Collapse
Affiliation(s)
| | | | - Ben Coleman
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | | - Joel Anderton
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Anita M Bagley
- Shriners Children's Northern California, Sacramento, CA, USA
| | - Eduard Bakštein
- National Institute of Mental Health, Klecany, Czech Republic
| | - James P Balhoff
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Gareth Baynam
- Rare Care Centre, Perth Children's Hospital, Perth, Australia
| | | | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Holli Bertram
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Somer Bishop
- Department of Psychiatry and Behavioral Sciences, UCSF Weil Institute for Neuroscience, San Francisco, CA, USA
| | - Hannah Blau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - David F Bodenstein
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | - Kaan Boztug
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Jolana Čady
- Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | - Tiffany J Callahan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, NY, NY, USA
| | | | - Seth J Carbon
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | - J Harry Caufield
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Lauren E Chan
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Christopher G Chute
- Schools of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Jaime Cruz-Rojo
- UDISGEN (Dysmorphology and Genetics Unit), 12 de Octubre Hospital, Madrid, Spain
| | - Noémi Dahan-Oliel
- Department of Clinical Research, Shriners Hospitals for Children, Montreal, Quebec, Canada
| | - Jon R Davids
- Shriners Children's Northern California, Sacramento, CA, USA
| | - Maud de Dieuleveult
- Département I&D, AP-HP, Banque Nationale de Données Maladies Rares, Paris, France
| | - Vinicius de Souza
- European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Bert B A de Vries
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - J Raymond DePaulo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Beata Derfalvi
- Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Ferdinand Dhombres
- Fetal Medicine Department, Armand Trousseau Hospital, Sorbonne University, GRC26, INSERM, Limics, Paris, France
| | - Claudia Diaz-Byrd
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Alexander J M Dingemans
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bruno Donadille
- St Antoine Hospital, Reference Center for Rare Growth Endocrine Disorders, Sorbonne University, AP-HP, INSERM, US14 - Orphanet, Plateforme Maladies Rares, Paris, France
| | | | - Reem Elfeky
- Department of Immunology, GOS Hospital for Children NHS Foundation Trust, University College London, London, UK
| | - Shahim Essaid
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Giovanna Fico
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Helen V Firth
- Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
| | - Yun Freudenberg-Hua
- Department of Psychiatry, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | | | - Davera L Gabriel
- School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | | | - Jessica Giordano
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rachel Gore Moses
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ian Green
- SNOMED International, London W2 6BD, UK
| | - Matthias Griese
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, German center for Lung research (DZL), Munich, Germany
| | - Tudor Groza
- Rare Care Centre, Perth Children's Hospital, Perth, Australia
| | | | - Julia Guthrie
- Department of Structural and Computational Biology, University of Vienna; Max Perutz Labs, Vienna, Austria
| | - Benjamin Gyori
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Ada Hamosh
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Marc Hanauer
- INSERM, US14 - Orphanet, Plateforme Maladies Rares, Paris, France
| | - Kateřina Hanušová
- Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | | | - Harshad Hegde
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Ingo Helbig
- Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kateřina Holasová
- Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | - Charles Tapley Hoyt
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | | | - Eric Hurwitz
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Julius O B Jacobsen
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Lisa Joseph
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, Bethesda, MD, USA
| | - Kamyar Keramatian
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Bryan King
- Department of Psychiatry and Behavioral Sciences, UCSF Weil Institute for Neuroscience, San Francisco, CA, USA
| | - Katrin Knoflach
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, German center for Lung research (DZL), Munich, Germany
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Megan L Kraus
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Carlo Kroll
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Maaike Kusters
- Immunology, NIHR Great Ormond Street Hospital BRC, London, UK
| | - Markus S Ladewig
- Department of Ophthalmology, University Clinic Marburg - Campus Fulda, Fulda, Germany
| | - David Lagorce
- INSERM, US14 - Orphanet, Plateforme Maladies Rares, Paris, France
| | - Meng-Chuan Lai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Pablo Lapunzina
- Institute of Medical and Molecular Genetics, Hospital Univ. La Paz, Madrid, Spain
| | - Bryan Laraway
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Lewis-Smith
- Translational and Clinical Research Institute, Henry Wellcome Building, Framlington Place, Newcastle University, Newcastle-Upon-Tyne NE14LP, UK
| | | | - Caterina Lucano
- INSERM, US14 - Orphanet, Plateforme Maladies Rares, Paris, France
| | - Marzieh Majd
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mary L Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victor Martinez-Glez
- Center for Genomic Medicine, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Sabadell, Spain
| | - Toby H McHenry
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Julie A McMurry
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michaela Mihulová
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - Caitlin E Millett
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Philip B Mitchell
- Discipline of Psychiatry & Mental Health, School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW, Australia
| | - Veronika Moslerová
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - Kenji Narutomi
- Okinawa Prefectural Nanbu Medical Center & Children's Medical Center
| | - Shahrzad Nematollahi
- School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada
| | - Julian Nevado
- Institute of Medical and Molecular Genetics, Hospital Univ. La Paz, Madrid, Spain
| | - Andrew A Nierenberg
- Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, MA, USA
| | - Nikola Novák Čajbiková
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - John I Nurnberger
- Stark Neurosciences Research Institute, Departments of Psychiatry and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Daniel Olson
- Data Collaboration Center, Data Science, Critical Path Institute, Tucson, AZ, USA
| | - Abigail Ortiz
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Harry Pachajoa
- Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| | - Guiomar Perez de Nanclares
- Molecular (epi) genetics lab, Bioaraba Health Research Institute, Araba University Hospital, Vitoria-Gasteiz, Spain
| | - Amy Peters
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Tim Putman
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christina K Rapp
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, German center for Lung research (DZL), Munich, Germany
| | - Ana Rath
- INSERM, US14 - Orphanet, Plateforme Maladies Rares, Paris, France
| | - Justin Reese
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Lauren Rekerle
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Angharad M Roberts
- National Heart & Lung Institute & MRC London Institute of Medical Sciences, Imperial College London, London W12 0HS, UK
| | - Suzy Roy
- SNOMED International, London W2 6BD, UK
| | - Stephan J Sanders
- Department of Paediatrics, Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Catharina Schuetz
- Universitätsklinikum Carl Gustav Carus, Medizinische Fakultät, TU, Dresden, Germany
| | - Eva C Schulte
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
| | - Thomas G Schulze
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Martin Schwarz
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - Katie Scott
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Dominik Seelow
- Exploratory Diagnostic Sciences, Berliner Institut für Gesundheitsforschung - Charité, Berlin, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center UKS, Homburg/Saar, Germany
| | | | - Morgan N Similuk
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric S Simon
- Eisenberg Family Depression Center, University of Michigan, Ann Arbor, MI, USA
| | - Balwinder Singh
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Damian Smedley
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Jake T Smolinsky
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Sarah Sperry
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | | | - Ray Stefancsik
- European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Robin Steinhaus
- Exploratory Diagnostic Sciences, Berliner Institut für Gesundheitsforschung - Charité, Berlin, Germany
| | - Rebecca Strawbridge
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | | | - Polina Talapova
- Institute for Research and Health Policy Studies, Tufts Medicine, Boston, MA 2111, USA
| | | | - Pavel Tesner
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - Rhys H Thomas
- Translational and Clinical Research Institute, Henry Wellcome Building, Framlington Place, Newcastle University, Newcastle-Upon-Tyne NE14LP, UK
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, Bethesda, MD, USA
| | - Marek Turnovec
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - Marielle E van Gijn
- Department of Genetics, University Medical Center Groningen, Groningen, Netherlands
| | | | - Markéta Vlčková
- Department of Biology and Medical Genetics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| | - Anita Walden
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kai Wang
- Chinese HPO Consortium, Beijing, China
| | - Ron Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - James S Ware
- National Heart & Lung Institute & MRC London Institute of Medical Sciences, Imperial College London, London W12 0HS, UK
| | | | | | - Lisa D Wiggins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Andrew E Williams
- Institute for Research and Health Policy Studies, Tufts Medicine, Boston, MA 2111, USA
| | - Chen Wu
- Chinese HPO Consortium, Beijing, China
| | - Margot J Wyrwoll
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Institute for Stem Cell Research, University of Edinburgh, Edinburgh, UK
| | - Hui Xiong
- Chinese HPO Consortium, Beijing, China
| | - Nefize Yalin
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Yasunori Yamamoto
- Database Center for Life Science, Joint Support-Center for Data Science Research, Research Organization of Information and Systems, Japan
| | - Lakshmi N Yatham
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Anastasia K Yocum
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Allan H Young
- Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, London SE5 8AF, UK
| | - Zafer Yüksel
- Department of Human Genetics, Bioscientia Healthcare GmbH, Ingelheim, Germany
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andreas Zankl
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Ignacio Zarante
- Institute of Human Genetics, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Miroslav Zvolský
- Institute of Health Information and Statistics of the Czech Republic, Prague, Czech Republic
| | - Sabrina Toro
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Leigh C Carmody
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Nomi L Harris
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Monica C Munoz-Torres
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel Danis
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Christopher J Mungall
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | - Melissa A Haendel
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Peter N Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
5
|
Wang Y, Lin Y, Wu S, Sun J, Meng Y, Jin E, Kong D, Duan G, Bei S, Fan Z, Wu G, Hao L, Song S, Tang B, Zhao W. BioKA: a curated and integrated biomarker knowledgebase for animals. Nucleic Acids Res 2024; 52:D1121-D1130. [PMID: 37843156 PMCID: PMC10767812 DOI: 10.1093/nar/gkad873] [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: 08/15/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023] Open
Abstract
Biomarkers play an important role in various area such as personalized medicine, drug development, clinical care, and molecule breeding. However, existing animals' biomarker resources predominantly focus on human diseases, leaving a significant gap in non-human animal disease understanding and breeding research. To address this limitation, we present BioKA (Biomarker Knowledgebase for Animals, https://ngdc.cncb.ac.cn/bioka), a curated and integrated knowledgebase encompassing multiple animal species, diseases/traits, and annotated resources. Currently, BioKA houses 16 296 biomarkers associated with 951 mapped diseases/traits across 31 species from 4747 references, including 11 925 gene/protein biomarkers, 1784 miRNA biomarkers, 1043 mutation biomarkers, 773 metabolic biomarkers, 357 circRNA biomarkers and 127 lncRNA biomarkers. Furthermore, BioKA integrates various annotations such as GOs, protein structures, protein-protein interaction networks, miRNA targets and so on, and constructs an interactive knowledge network of biomarkers including circRNA-miRNA-mRNA associations, lncRNA-miRNA associations and protein-protein associations, which is convenient for efficient data exploration. Moreover, BioKA provides detailed information on 308 breeds/strains of 13 species, and homologous annotations for 8784 biomarkers across 16 species, and offers three online application tools. The comprehensive knowledge provided by BioKA not only advances human disease research but also contributes to a deeper understanding of animal diseases and supports livestock breeding.
Collapse
Affiliation(s)
- Yibo Wang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yihao Lin
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sicheng Wu
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiani Sun
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuyan Meng
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Enhui Jin
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Demian Kong
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangya Duan
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shaoqi Bei
- Qilu University of Technology (Shandong Academy of Sciences), Shandong 250353, China
| | - Zhuojing Fan
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Gangao Wu
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Lili Hao
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Shuhui Song
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Bixia Tang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Wenming Zhao
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Liu X, Bishir M, Hodgkinson C, Goldman D, Chang SL. The mechanisms underlying alcohol-induced decreased splenic size: A network meta-analysis study. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:72-87. [PMID: 38059389 PMCID: PMC11161039 DOI: 10.1111/acer.15234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Organ weight change is widely accepted as a measure of toxicologic pathology. We and other groups have shown that excessive alcohol exposure leads to decreased spleen weight in rodents. This study explores the mechanisms underlying alcohol-induced splenic injury through a network meta-analysis. METHODS QIAGEN Ingenuity Pathway Analysis (IPA) and Mammalian Phenotype (MP) Ontology were used to identify alcohol-related molecules associated with the small spleen phenotype. Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and IPA bioinformatics tools were then used to analyze the biologic processes and enriched signaling pathways engaging these molecules. In addition, the "downstream effects analysis" algorithm was used to quantify alcohol's effects. RESULTS IPA identified 623 molecules affected by alcohol and a Venn diagram revealed that 26 of these molecules overlapped with those associated with the MP Ontology of small spleen. The 26 molecules are TGFB1, CASP8, MTOR, ESR1, CXCR4, CAMK4, NFKBIA, DRD2, BCL2, FAS, PEBP1, TRAF2, ATM, IGHM, EDNRB, MDM2, GLRA1, PRF1, TLR7, IFNG, ALOX5, FOXO1, IL15, APOE, IKBKG, and RORA. Some of the 26 molecules were also associated with the MP Ontology of abnormal white pulp and red pulp morphology of the spleen, abnormal splenic cell ratio, decreased splenocyte number, abnormal spleen physiology, increased splenocyte apoptosis, and reduced splenocyte proliferation. STRING and IPA "Core Analysis" showed that these molecules were mainly involved in pathways related to cell apoptosis, proliferation, migration, and immune responses. IPA's "Molecular Activity Predictor" tool showed that concurrent effects of activation and inhibition of these molecules led to decreased spleen size by modulating apoptosis, proliferation, and migration of splenocytes. CONCLUSIONS Our network meta-analysis revealed that excessive alcohol exposure can damage the spleen through a variety of molecular mechanisms, thereby affecting immune function and human health. We found that alcohol-mediated splenic atrophy is largely mediated by increased apoptosis signaling, migration of cells, and inhibition of splenocyte proliferation.
Collapse
Affiliation(s)
- Xiangqian Liu
- Institute of NeuroImmune Pharmacology, South Orange, New Jersey, USA
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Muhammed Bishir
- Institute of NeuroImmune Pharmacology, South Orange, New Jersey, USA
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, USA
| | - Colin Hodgkinson
- Laboratory of Neurogenetics, NIAAA, NIH, Rockville, Maryland, USA
| | - David Goldman
- Laboratory of Neurogenetics, NIAAA, NIH, Rockville, Maryland, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, South Orange, New Jersey, USA
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, USA
| |
Collapse
|
7
|
Bogue MA, Ball RL, Walton DO, Dunn MH, Kolishovski G, Berger A, Lamoureux A, Grubb SC, Gerring M, Kim M, Liang H, Emerson J, Stearns T, He H, Mukherjee G, Bluis J, Davis S, Desai S, Sundberg B, Kadakkuzha B, Kunde-Ramamoorthy G, Philip VM, Chesler EJ. Mouse phenome database: curated data repository with interactive multi-population and multi-trait analyses. Mamm Genome 2023; 34:509-519. [PMID: 37581698 PMCID: PMC10627943 DOI: 10.1007/s00335-023-10014-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023]
Abstract
The Mouse Phenome Database continues to serve as a curated repository and analysis suite for measured attributes of members of diverse mouse populations. The repository includes annotation to community standard ontologies and guidelines, a database of allelic states for 657 mouse strains, a collection of protocols, and analysis tools for flexible, interactive, user directed analyses that increasingly integrates data across traits and populations. The database has grown from its initial focus on a standard set of inbred strains to include heterogeneous mouse populations such as the Diversity Outbred and mapping crosses and well as Collaborative Cross, Hybrid Mouse Diversity Panel, and recombinant inbred strains. Most recently the system has expanded to include data from the International Mouse Phenotyping Consortium. Collectively these data are accessible by API and provided with an interactive tool suite that enables users' persistent selection, storage, and operation on collections of measures. The tool suite allows basic analyses, advanced functions with dynamic visualization including multi-population meta-analysis, multivariate outlier detection, trait pattern matching, correlation analyses and other functions. The data resources and analysis suite provide users a flexible environment in which to explore the basis of phenotypic variation in health and disease across the lifespan.
Collapse
Affiliation(s)
- Molly A Bogue
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA.
| | - Robyn L Ball
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - David O Walton
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Matthew H Dunn
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | | | - Anna Lamoureux
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Stephen C Grubb
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Matthew Gerring
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Matthew Kim
- University of British Columbia, Vancouver, BC, Canada
| | - Hongping Liang
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Jake Emerson
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Timothy Stearns
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Hao He
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | - John Bluis
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Sara Davis
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Sejal Desai
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Beth Sundberg
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | | | - Vivek M Philip
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | |
Collapse
|
8
|
Hill DP, Drabkin HJ, Smith CL, Van Auken KM, D’Eustachio P. Biochemical pathways represented by Gene Ontology-Causal Activity Models identify distinct phenotypes resulting from mutations in pathways. Genetics 2023; 225:iyad152. [PMID: 37579192 PMCID: PMC10550311 DOI: 10.1093/genetics/iyad152] [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: 07/13/2023] [Revised: 07/13/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023] Open
Abstract
Gene inactivation can affect the process(es) in which that gene acts and causally downstream ones, yielding diverse mutant phenotypes. Identifying the genetic pathways resulting in a given phenotype helps us understand how individual genes interact in a functional network. Computable representations of biological pathways include detailed process descriptions in the Reactome Knowledgebase and causal activity flows between molecular functions in Gene Ontology-Causal Activity Models (GO-CAMs). A computational process has been developed to convert Reactome pathways to GO-CAMs. Laboratory mice are widely used models of normal and pathological human processes. We have converted human Reactome GO-CAMs to orthologous mouse GO-CAMs, as a resource to transfer pathway knowledge between humans and model organisms. These mouse GO-CAMs allowed us to define sets of genes that function in a causally connected way. To demonstrate that individual variant genes from connected pathways result in similar but distinguishable phenotypes, we used the genes in our pathway models to cross-query mouse phenotype annotations in the Mouse Genome Database (MGD). Using GO-CAM representations of 2 related but distinct pathways, gluconeogenesis and glycolysis, we show that individual causal paths in gene networks give rise to discrete phenotypic outcomes resulting from perturbations of glycolytic and gluconeogenic genes. The accurate and detailed descriptions of gene interactions recovered in this analysis of well-studied processes suggest that this strategy can be applied to less well-understood processes in less well-studied model systems to predict phenotypic outcomes of novel gene variants and to identify potential gene targets in altered processes.
Collapse
Affiliation(s)
- David P Hill
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | - Kimberly M Van Auken
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Peter D’Eustachio
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
9
|
Feng Q, Xia W, Feng Z, Tan Y, Zhang Y, Liu D, Zhang G. The accelerated organ senescence and proteotoxicity in thyrotoxicosis mice. J Cell Physiol 2023; 238:2481-2498. [PMID: 37750538 DOI: 10.1002/jcp.31108] [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: 03/08/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 09/27/2023]
Abstract
The mechanism of aging has always been the focus of research, because aging is related to disease susceptibility and seriously affects people's quality of life. The diseases also accelerate the aging process, especially the pathological changes of substantive organs, such as cardiac hypertrophy, severely shortened lifespan. So, lesions in organs are both a consequence and a cause of aging. However, the disease in a given organ is not in isolation but is a systemic problem. Our previous study found that thyrotoxicosis mice model has aging characteristics including immunosenescence, lipotoxicity, malnutrition. But all these characteristics will lead to organ senescence, therefore, this study continued to study the aging changes of important organs such as heart, liver, and kidney in thyrotoxicosis mice using tandem mass tags (TMT) proteomics method. The results showed that the excess thyroxine led to cardiac hypertrophy. In the liver, the ability to synthesize functional proteins, detoxify, and metabolism were declined. The effect on the kidney was the decreased ability of detoxify and metabolism. The main finding of the present study was that the acceleration of organ senescence by excess thyroxine was due to proteotoxicity. The shared cause of proteotoxicity in the three organs included the intensify of oxidative phosphorylation, the redundancy production of ribosomes, and the lack of splicing and ubiquitin proteasome system function. Totally, proteotoxicity was another parallel between thyrotoxicosis and aging in addition to lipotoxicity. Our research provided a convenient and appropriate animal model for exploring aging mechanism and antiaging drugs.
Collapse
Affiliation(s)
- Qin Feng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
| | - Wenkai Xia
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
| | - Zhong Feng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Yujun Tan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
| | - Yongxia Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, Shandong, China
| |
Collapse
|
10
|
Powell G, Simon MM, Pulit S, Mallon AM, Lindgren CM. Genic constraint against nonsynonymous variation across the mouse genome. BMC Genomics 2023; 24:562. [PMID: 37736706 PMCID: PMC10514939 DOI: 10.1186/s12864-023-09637-2] [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: 06/12/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Selective constraint, the depletion of variation due to negative selection, provides insights into the functional impact of variants and disease mechanisms. However, its characterization in mice, the most commonly used mammalian model, remains limited. This study aims to quantify mouse gene constraint using a new metric called the nonsynonymous observed expected ratio (NOER) and investigate its relationship with gene function. RESULTS NOER was calculated using whole-genome sequencing data from wild mouse populations (Mus musculus sp and Mus spretus). Positive correlations were observed between mouse gene constraint and the number of associated knockout phenotypes, indicating stronger constraint on pleiotropic genes. Furthermore, mouse gene constraint showed a positive correlation with the number of pathogenic variant sites in their human orthologues, supporting the relevance of mouse models in studying human disease variants. CONCLUSIONS NOER provides a resource for assessing the fitness consequences of genetic variants in mouse genes and understanding the relationship between gene constraint and function. The study's findings highlight the importance of pleiotropy in selective constraint and support the utility of mouse models in investigating human disease variants. Further research with larger sample sizes can refine constraint estimates in mice and enable more comprehensive comparisons of constraint between mouse and human orthologues.
Collapse
Affiliation(s)
- George Powell
- Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, Oxford, UK.
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK.
| | - Michelle M Simon
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Sara Pulit
- Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, Oxford, UK
| | - Ann-Marie Mallon
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Cecilia M Lindgren
- Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
11
|
Murray GC, Bubier JA, Zinder OJ, Harris B, Clark J, Christopher MC, Hanley C, Tjong H, Li M, Ngan CY, Reinholdt L, Burgess RW, Tadenev ALD. An allelic series of spontaneous Rorb mutant mice exhibit a gait phenotype, changes in retina morphology and behavior, and gene expression signatures associated with the unfolded protein response. G3 (BETHESDA, MD.) 2023; 13:jkad131. [PMID: 37300435 PMCID: PMC10411600 DOI: 10.1093/g3journal/jkad131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
The Retinoid-related orphan receptor beta (RORβ) gene encodes a developmental transcription factor and has 2 predominant isoforms created through alternative first exon usage; one specific to the retina and another present more broadly in the central nervous system, particularly regions involved in sensory processing. RORβ belongs to the nuclear receptor family and plays important roles in cell fate specification in the retina and cortical layer formation. In mice, loss of RORβ causes disorganized retina layers, postnatal degeneration, and production of immature cone photoreceptors. Hyperflexion or "high-stepping" of rear limbs caused by reduced presynaptic inhibition by Rorb-expressing inhibitory interneurons of the spinal cord is evident in RORβ-deficient mice. RORβ variants in patients are associated with susceptibility to various neurodevelopmental conditions, primarily generalized epilepsies, but including intellectual disability, bipolar, and autism spectrum disorders. The mechanisms by which RORβ variants confer susceptibility to these neurodevelopmental disorders are unknown but may involve aberrant neural circuit formation and hyperexcitability during development. Here we report an allelic series in 5 strains of spontaneous Rorb mutant mice with a high-stepping gait phenotype. We show retinal abnormalities in a subset of these mutants and demonstrate significant differences in various behavioral phenotypes related to cognition. Gene expression analyses in all 5 mutants reveal a shared over-representation of the unfolded protein response and pathways related to endoplasmic reticulum stress, suggesting a possible mechanism of susceptibility relevant to patients.
Collapse
Affiliation(s)
- George C Murray
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | | | | | | | - James Clark
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | - Harianto Tjong
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Meihong Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chew Yee Ngan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | | | - Robert W Burgess
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | | |
Collapse
|
12
|
Hill DP, Drabkin HJ, Smith CL, Van Auken KM, D’Eustachio P. Biochemical Pathways Represented by Gene Ontology Causal Activity Models Identify Distinct Phenotypes Resulting from Mutations in Pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541760. [PMID: 37293039 PMCID: PMC10245817 DOI: 10.1101/2023.05.22.541760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gene inactivation can affect the process(es) in which that gene acts and causally downstream ones, yielding diverse mutant phenotypes. Identifying the genetic pathways resulting in a given phenotype helps us understand how individual genes interact in a functional network. Computable representations of biological pathways include detailed process descriptions in the Reactome Knowledgebase, and causal activity flows between molecular functions in Gene Ontology-Causal Activity Models (GO-CAMs). A computational process has been developed to convert Reactome pathways to GO-CAMs. Laboratory mice are widely used models of normal and pathological human processes. We have converted human Reactome GO-CAMs to orthologous mouse GO-CAMs, as a resource to transfer pathway knowledge between humans and model organisms. These mouse GO-CAMs allowed us to define sets of genes that function in a connected and well-defined way. To test whether individual genes from well-defined pathways result in similar and distinguishable phenotypes, we used the genes in our pathway models to cross-query mouse phenotype annotations in the Mouse Genome Database (MGD). Using GO-CAM representations of two related but distinct pathways, gluconeogenesis and glycolysis, we can identify causal paths in gene networks that give rise to discrete phenotypic outcomes for perturbations of glycolysis and gluconeogenesis. The accurate and detailed descriptions of gene interactions recovered in this analysis of well-studied processes suggest that this strategy can be applied to less well-understood processes in less well-studied model systems to predict phenotypic outcomes of novel gene variants and to identify potential gene targets in altered processes.
Collapse
Affiliation(s)
| | | | | | - Kimberly M Van Auken
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena CA 91125 USA
| | - Peter D’Eustachio
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York NY 10016 USA
| |
Collapse
|
13
|
Khaveh N, Schachler K, Berghöfer J, Jung K, Metzger J. Altered hair root gene expression profiles highlight calcium signaling and lipid metabolism pathways to be associated with curly hair initiation and maintenance in Mangalitza pigs. Front Genet 2023; 14:1184015. [PMID: 37351343 PMCID: PMC10282778 DOI: 10.3389/fgene.2023.1184015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
Hair types have been under strong targeted selection in domestic animals for their impact on skin protection, thermoregulation and exterior morphology, and subsequent economic importance. In pigs, a very special hair phenotype was observed in Mangalitza, who expresses a thick coat of curly bristles and downy hair. Two breed-specific missense variants in TRPM2 and CYP4F3 were suggested to be associated with the Mangalitza pig's hair shape due to their role in hair follicle morphogenesis reported for human and mice. However, the mechanism behind this expression of a curly hair type is still unclear and needs to be explored. In our study, hair shafts were measured and investigated for the curvature of the hair in Mangalitza and crossbreeds in comparison to straight-coated pigs. For molecular studies, hair roots underwent RNA sequencing for a differential gene expression analysis using DESeq2. The output matrix of normalized counts was then used to construct weighted gene co-expression networks. The resulting hair root gene expression profiles highlighted 454 genes to be significantly differentially expressed for initiation of curly hair phenotype in newborn Mangalitza piglets versus post-initiation in later development. Furthermore, 2,554 genes showed a significant differential gene expression in curly hair in comparison to straight hair. Neither TRPM2 nor CYP4F3 were identified as differentially expressed. Incidence of the genes in weighted co-expression networks associated with TRPM2 and CYP4F3, and prominent interactions of subsequent proteins with lipids and calcium-related pathways suggested calcium signaling and/or lipid metabolism as essential players in the induction of the curly hair as well as an ionic calcium-dependency to be a prominent factor for the maintenance of this phenotype. Subsequently, our study highlights the complex interrelations and dependencies of mutant genes TRPM2 and CYP4F3 and associated gene expression patterns, allowing the initiation of curly hair type during the development of a piglet as well as the maintenance in adult individuals.
Collapse
Affiliation(s)
- Nadia Khaveh
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kathrin Schachler
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Jan Berghöfer
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Klaus Jung
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Julia Metzger
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
14
|
Recanatini M, Menestrina L. Network modeling helps to tackle the complexity of drug-disease systems. WIREs Mech Dis 2023:e1607. [PMID: 36958762 DOI: 10.1002/wsbm.1607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/03/2023] [Accepted: 03/03/2023] [Indexed: 03/25/2023]
Abstract
From the (patho)physiological point of view, diseases can be considered as emergent properties of living systems stemming from the complexity of these systems. Complex systems display some typical features, including the presence of emergent behavior and the organization in successive hierarchic levels. Drug treatments increase this complexity scenario, and from some years the use of network models has been introduced to describe drug-disease systems and to make predictions about them with regard to several aspects related to drug discovery. Here, we review some recent examples thereof with the aim to illustrate how network science tools can be very effective in addressing both tasks. We will examine the use of bipartite networks that lead to the important concept of "disease module", as well as the introduction of more articulated models, like multi-scale and multiplex networks, able to describe disease systems at increasing levels of organization. Examples of predictive models will then be discussed, considering both those that exploit approaches purely based on graph theory and those that integrate machine learning methods. A short account of both kinds of methodological applications will be provided. Finally, the point will be made on the present situation of modeling complex drug-disease systems highlighting some open issues. This article is categorized under: Neurological Diseases > Computational Models Infectious Diseases > Computational Models Cardiovascular Diseases > Computational Models.
Collapse
Affiliation(s)
- Maurizio Recanatini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, Bologna, 40126, Italy
| | - Luca Menestrina
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, Bologna, 40126, Italy
| |
Collapse
|
15
|
Beck T, Rowlands T, Shorter T, Brookes AJ. GWAS Central: an expanding resource for finding and visualising genotype and phenotype data from genome-wide association studies. Nucleic Acids Res 2023; 51:D986-D993. [PMID: 36350644 PMCID: PMC9825503 DOI: 10.1093/nar/gkac1017] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/10/2022] Open
Abstract
The GWAS Central resource gathers and curates extensive summary-level genome-wide association study (GWAS) data and puts a range of user-friendly but powerful website tools for the comparison and visualisation of GWAS data at the fingertips of researchers. Through our continued efforts to harmonise and import data received from GWAS authors and consortia, and data sets actively collected from public sources, the database now contains over 72.5 million P-values for over 5000 studies testing over 7.4 million unique genetic markers investigating over 1700 unique phenotypes. Here, we describe an update to integrate this extensive data collection with mouse disease model data to support insights into the functional impact of human genetic variation. GWAS Central has expanded to include mouse gene-phenotype associations observed during mouse gene knockout screens. To allow similar cross-species phenotypes to be compared, terms from mammalian and human phenotype ontologies have been mapped. New interactive interfaces to find, correlate and view human and mouse genotype-phenotype associations are included in the website toolkit. Additionally, the integrated browser for interrogating multiple association data sets has been updated and a GA4GH Beacon API endpoint has been added for discovering variants tested in GWAS. The GWAS Central resource is accessible at https://www.gwascentral.org/.
Collapse
Affiliation(s)
- Tim Beck
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
- Health Data Research UK (HDR UK), London, UK
| | - Thomas Rowlands
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Tom Shorter
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Anthony J Brookes
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK
- Health Data Research UK (HDR UK), London, UK
| |
Collapse
|
16
|
Bogue MA, Ball RL, Philip VM, Walton DO, Dunn M, Kolishovski G, Lamoureux A, Gerring M, Liang H, Emerson J, Stearns T, He H, Mukherjee G, Bluis J, Desai S, Sundberg B, Kadakkuzha B, Kunde-Ramamoorthy G, Chesler E. Mouse Phenome Database: towards a more FAIR-compliant and TRUST-worthy data repository and tool suite for phenotypes and genotypes. Nucleic Acids Res 2023; 51:D1067-D1074. [PMID: 36330959 PMCID: PMC9825561 DOI: 10.1093/nar/gkac1007] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/11/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
The Mouse Phenome Database (MPD; https://phenome.jax.org; RRID:SCR_003212), supported by the US National Institutes of Health, is a Biomedical Data Repository listed in the Trans-NIH Biomedical Informatics Coordinating Committee registry. As an increasingly FAIR-compliant and TRUST-worthy data repository, MPD accepts phenotype and genotype data from mouse experiments and curates, organizes, integrates, archives, and distributes those data using community standards. Data are accompanied by rich metadata, including widely used ontologies and detailed protocols. Data are from all over the world and represent genetic, behavioral, morphological, and physiological disease-related characteristics in mice at baseline or those exposed to drugs or other treatments. MPD houses data from over 6000 strains and populations, representing many reproducible strain types and heterogenous populations such as the Diversity Outbred where each mouse is unique but can be genotyped throughout the genome. A suite of analysis tools is available to aggregate, visualize, and analyze these data within and across studies and populations in an increasingly traceable and reproducible manner. We have refined existing resources and developed new tools to continue to provide users with access to consistent, high-quality data that has translational relevance in a modernized infrastructure that enables interaction with a suite of bioinformatics analytic and data services.
Collapse
Affiliation(s)
- Molly A Bogue
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | - Robyn L Ball
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | | | | | | | | | | | | | | | - Jake Emerson
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | - Tim Stearns
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | - Hao He
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | | | - John Bluis
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | - Sejal Desai
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | - Beth Sundberg
- The Jackson Laboratory, Bar Harbor Maine, 04609, USA
| | | | | | | |
Collapse
|
17
|
Murray GC, Bais P, Hatton CL, Tadenev ALD, Hoffmann BR, Stodola TJ, Morelli KH, Pratt SL, Schroeder D, Doty R, Fiehn O, John SWM, Bult CJ, Cox GA, Burgess RW. Mouse models of NADK2 deficiency analyzed for metabolic and gene expression changes to elucidate pathophysiology. Hum Mol Genet 2022; 31:4055-4074. [PMID: 35796562 PMCID: PMC9703942 DOI: 10.1093/hmg/ddac151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
NADK2 encodes the mitochondrial form of nicotinamide adenine dinucleotide (NAD) kinase, which phosphorylates NAD. Rare recessive mutations in human NADK2 are associated with a syndromic neurological mitochondrial disease that includes metabolic changes, such as hyperlysinemia and 2,4 dienoyl CoA reductase (DECR) deficiency. However, the full pathophysiology resulting from NADK2 deficiency is not known. Here, we describe two chemically induced mouse mutations in Nadk2-S326L and S330P-which cause severe neuromuscular disease and shorten lifespan. The S330P allele was characterized in detail and shown to have marked denervation of neuromuscular junctions by 5 weeks of age and muscle atrophy by 11 weeks of age. Cerebellar Purkinje cells also showed progressive degeneration in this model. Transcriptome profiling on brain and muscle was performed at early and late disease stages. In addition, metabolomic profiling was performed on the brain, muscle, liver and spinal cord at the same ages and on plasma at 5 weeks. Combined transcriptomic and metabolomic analyses identified hyperlysinemia, DECR deficiency and generalized metabolic dysfunction in Nadk2 mutant mice, indicating relevance to the human disease. We compared findings from the Nadk model to equivalent RNA sequencing and metabolomic datasets from a mouse model of infantile neuroaxonal dystrophy, caused by recessive mutations in Pla2g6. This enabled us to identify disrupted biological processes that are common between these mouse models of neurological disease, as well as those processes that are gene-specific. These findings improve our understanding of the pathophysiology of neuromuscular diseases and describe mouse models that will be useful for future preclinical studies.
Collapse
Affiliation(s)
- G C Murray
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - P Bais
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - C L Hatton
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - A L D Tadenev
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - B R Hoffmann
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - T J Stodola
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - K H Morelli
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - S L Pratt
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - D Schroeder
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - R Doty
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
| | - O Fiehn
- West Coast Metabolomics Center, University of California Davis, 451 Health Science Dr., Davis, CA 95618, USA
| | - S W M John
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10032, USA
| | - C J Bult
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - G A Cox
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - R W Burgess
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME 04609, USA
- The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
18
|
Faubel RJ, Santos Canellas VS, Gaesser J, Beluk NH, Feinstein TN, Wang Y, Yankova M, Karunakaran KB, King SM, Ganapathiraju MK, Lo CW. Flow blockage disrupts cilia-driven fluid transport in the epileptic brain. Acta Neuropathol 2022; 144:691-706. [PMID: 35980457 DOI: 10.1007/s00401-022-02463-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/28/2023]
Abstract
A carpet of ependymal motile cilia lines the brain ventricular system, forming a network of flow channels and barriers that pattern cerebrospinal fluid (CSF) flow at the surface. This CSF transport system is evolutionary conserved, but its physiological function remains unknown. Here we investigated its potential role in epilepsy with studies focused on CDKL5 deficiency disorder (CDD), a neurodevelopmental disorder with early-onset epilepsy refractory to seizure medications and the most common cause of infant epilepsy. CDKL5 is a highly conserved X-linked gene suggesting its function in regulating cilia length and motion in the green alga Chlamydomonas might have implication in the etiology of CDD. Examination of the structure and function of airway motile cilia revealed both the CDD patients and the Cdkl5 knockout mice exhibit cilia lengthening and abnormal cilia motion. Similar defects were observed for brain ventricular cilia in the Cdkl5 knockout mice. Mapping ependymal cilia generated flow in the ventral third ventricle (v3V), a brain region with important physiological functions showed altered patterning of flow. Tracing of cilia-mediated inflow into v3V with fluorescent dye revealed the appearance of a flow barrier at the inlet of v3V in Cdkl5 knockout mice. Analysis of mice with a mutation in another epilepsy-associated kinase, Yes1, showed the same disturbance of cilia motion and flow patterning. The flow barrier was also observed in the Foxj1± and FOXJ1CreERT:Cdkl5y/fl mice, confirming the contribution of ventricular cilia to the flow disturbances. Importantly, mice exhibiting altered cilia-driven flow also showed increased susceptibility to anesthesia-induced seizure-like activity. The cilia-driven flow disturbance arises from altered cilia beating orientation with the disrupted polarity of the cilia anchoring rootlet meshwork. Together these findings indicate motile cilia disturbances have an essential role in CDD-associated seizures and beyond, suggesting cilia regulating kinases may be a therapeutic target for medication-resistant epilepsy.
Collapse
Affiliation(s)
- Regina J Faubel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA
| | - Veronica S Santos Canellas
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA
| | - Jenna Gaesser
- Division of Child Neurology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA
| | - Nancy H Beluk
- Division of Radiology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Tim N Feinstein
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA
| | - Yong Wang
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Am Faßberg 17, 37077, Göttingen, Germany
| | - Maya Yankova
- Department of Molecular Biology and Biophysics, And Electron Microscopy Facility, University of Connecticut Health Center, Farmington, CT, 06030-3305, USA
| | - Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, 560012, India
| | - Stephen M King
- Department of Molecular Biology and Biophysics, And Electron Microscopy Facility, University of Connecticut Health Center, Farmington, CT, 06030-3305, USA
| | - Madhavi K Ganapathiraju
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA.
| |
Collapse
|
19
|
Alghamdi SM, Schofield PN, Hoehndorf R. How much do model organism phenotypes contribute to the computational identification of human disease genes? Dis Model Mech 2022; 15:275986. [PMID: 35758016 PMCID: PMC9366895 DOI: 10.1242/dmm.049441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 06/13/2022] [Indexed: 12/04/2022] Open
Abstract
Computing phenotypic similarity helps identify new disease genes and diagnose rare diseases. Genotype–phenotype data from orthologous genes in model organisms can compensate for lack of human data and increase genome coverage. In the past decade, cross-species phenotype comparisons have proven valuble, and several ontologies have been developed for this purpose. The relative contribution of different model organisms to computational identification of disease-associated genes is not fully explored. We used phenotype ontologies to semantically relate phenotypes resulting from loss-of-function mutations in model organisms to disease-associated phenotypes in humans. Semantic machine learning methods were used to measure the contribution of different model organisms to the identification of known human gene–disease associations. We found that mouse genotype–phenotype data provided the most important dataset in the identification of human disease genes by semantic similarity and machine learning over phenotype ontologies. Other model organisms' data did not improve identification over that obtained using the mouse alone, and therefore did not contribute significantly to this task. Our work impacts on the development of integrated phenotype ontologies, as well as for the use of model organism phenotypes in human genetic variant interpretation. This article has an associated First Person interview with the first author of the paper. Editor's choice: We investigated the use of model organism phenotypes in the computational identification of disease genes, identifying several data biases and concluding that mouse model phenotypes contribute most to computational disease gene identification.
Collapse
Affiliation(s)
- Sarah M Alghamdi
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, 23955 Thuwal, Saudi Arabia
| | - Paul N Schofield
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, CB2 3EG, Cambridge, UK
| | - Robert Hoehndorf
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, 4700 KAUST, 23955 Thuwal, Saudi Arabia
| |
Collapse
|
20
|
Lindlöf A. The Vulnerability of the Developing Brain: Analysis of Highly Expressed Genes in Infant C57BL/6 Mouse Hippocampus in Relation to Phenotypic Annotation Derived From Mutational Studies. Bioinform Biol Insights 2022; 16:11779322211062722. [PMID: 35023907 PMCID: PMC8743926 DOI: 10.1177/11779322211062722] [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: 06/22/2021] [Accepted: 11/03/2021] [Indexed: 12/06/2022] Open
Abstract
The hippocampus has been shown to have a major role in learning and memory, but also to participate in the regulation of emotions. However, its specific role(s) in memory is still unclear. Hippocampal damage or dysfunction mainly results in memory issues, especially in the declarative memory but, in animal studies, has also shown to lead to hyperactivity and difficulty in inhibiting responses previously taught. The brain structure is affected in neuropathological disorders, such as Alzheimer's, epilepsy, and schizophrenia, and also by depression and stress. The hippocampus structure is far from mature at birth and undergoes substantial development throughout infant and juvenile life. The aim of this study was to survey genes highly expressed throughout the postnatal period in mouse hippocampus and which have also been linked to an abnormal phenotype through mutational studies to achieve a greater understanding about hippocampal functions during postnatal development. Publicly available gene expression data from C57BL/6 mouse hippocampus was analyzed; from a total of 5 time points (at postnatal day 1, 10, 15, 21, and 30), 547 genes highly expressed in all of these time points were selected for analysis. Highly expressed genes are considered to be of potential biological importance and appear to be multifunctional, and hence any dysfunction in such a gene will most likely have a large impact on the development of abilities during the postnatal and juvenile period. Phenotypic annotation data downloaded from Mouse Genomic Informatics database were analyzed for these genes, and the results showed that many of them are important for proper embryo development and infant survival, proper growth, and increase in body size, as well as for voluntary movement functions, motor coordination, and balance. The results also indicated an association with seizures that have primarily been characterized by uncontrolled motor activity and the development of proper grooming abilities. The complete list of genes and their phenotypic annotation data have been compiled in a file for easy access.
Collapse
|
21
|
Quantitative phosphoproteomics reveals GSK3A substrate network is involved in the cryodamage of sperm motility. Biosci Rep 2021; 41:229867. [PMID: 34596222 PMCID: PMC8521533 DOI: 10.1042/bsr20211326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022] Open
Abstract
During sperm cryopreservation, the most significant phenotype of cryodamage is the decrease in sperm motility. Several proteomics studies have already been performed to search for key regulators at the protein level. However, sperm functions are known to be highly regulated by phosphorylation signaling. Here, we constructed a quantitative phosphoproteome to investigate the expression change of phosphorylated sites during sperm cryopreservation. A total of 3107 phosphorylated sites are identified and 848 of them are found to be significantly differentially expressed (DE). Bioinformatics analysis showed that the corresponding genes of these regulated sites are highly associated with sperm motility, providing a connection between the molecular basis and the phenotype of cryodamage. We then performed kinase enrichment analysis and successfully identified glycogen synthase kinase-3α (GSK3A) as the key kinase that may play an important role in the regulation of sperm motility. We further constructed a GSK3A centric network that could help us better understand the molecular mechanism of cryodamage in sperm motility. Finally, we also verified that GSK3A was abnormally activated during this process. The presented phosphoproteome and functional associations provide abundant research resources for us to learn the regulation of sperm functions, as well as to optimize the cryoprotectant for sperm cryopreservation.
Collapse
|
22
|
Ringwald M, Richardson JE, Baldarelli RM, Blake JA, Kadin JA, Smith C, Bult CJ. Mouse Genome Informatics (MGI): latest news from MGD and GXD. Mamm Genome 2021; 33:4-18. [PMID: 34698891 PMCID: PMC8913530 DOI: 10.1007/s00335-021-09921-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/21/2021] [Indexed: 12/01/2022]
Abstract
The Mouse Genome Informatics (MGI) database system combines multiple expertly curated community data resources into a shared knowledge management ecosystem united by common metadata annotation standards. MGI's mission is to facilitate the use of the mouse as an experimental model for understanding the genetic and genomic basis of human health and disease. MGI is the authoritative source for mouse gene, allele, and strain nomenclature and is the primary source of mouse phenotype annotations, functional annotations, developmental gene expression information, and annotations of mouse models with human diseases. MGI maintains mouse anatomy and phenotype ontologies and contributes to the development of the Gene Ontology and Disease Ontology and uses these ontologies as standard terminologies for annotation. The Mouse Genome Database (MGD) and the Gene Expression Database (GXD) are MGI's two major knowledgebases. Here, we highlight some of the recent changes and enhancements to MGD and GXD that have been implemented in response to changing needs of the biomedical research community and to improve the efficiency of expert curation. MGI can be accessed freely at http://www.informatics.jax.org .
Collapse
|
23
|
Konopka T, Ng S, Smedley D. Diffusion enables integration of heterogeneous data and user-driven learning in a desktop knowledge-base. PLoS Comput Biol 2021; 17:e1009283. [PMID: 34379637 PMCID: PMC8382188 DOI: 10.1371/journal.pcbi.1009283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 08/23/2021] [Accepted: 07/16/2021] [Indexed: 11/20/2022] Open
Abstract
Integrating reference datasets (e.g. from high-throughput experiments) with unstructured and manually-assembled information (e.g. notes or comments from individual researchers) has the potential to tailor bioinformatic analyses to specific needs and to lead to new insights. However, developing bespoke analysis pipelines from scratch is time-consuming, and general tools for exploring such heterogeneous data are not available. We argue that by treating all data as text, a knowledge-base can accommodate a range of bioinformatic data types and applications. We show that a database coupled to nearest-neighbor algorithms can address common tasks such as gene-set analysis as well as specific tasks such as ontology translation. We further show that a mathematical transformation motivated by diffusion can be effective for exploration across heterogeneous datasets. Diffusion enables the knowledge-base to begin with a sparse query, impute more features, and find matches that would otherwise remain hidden. This can be used, for example, to map multi-modal queries consisting of gene symbols and phenotypes to descriptions of diseases. Diffusion also enables user-driven learning: when the knowledge-base cannot provide satisfactory search results in the first instance, users can improve the results in real-time by adding domain-specific knowledge. User-driven learning has implications for data management, integration, and curation.
Collapse
Affiliation(s)
- Tomasz Konopka
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Sandra Ng
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Damian Smedley
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
24
|
Dorman A, Binenbaum I, Abu-Toamih Atamni HJ, Chatziioannou A, Tomlinson I, Mott R, Iraqi FA. Genetic mapping of novel modifiers for Apc Min induced intestinal polyps' development using the genetic architecture power of the collaborative cross mice. BMC Genomics 2021; 22:566. [PMID: 34294033 PMCID: PMC8299641 DOI: 10.1186/s12864-021-07890-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 07/14/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Familial adenomatous polyposis is an inherited genetic disease, characterized by colorectal polyps. It is caused by inactivating mutations in the Adenomatous polyposis coli (Apc) gene. Mice carrying a nonsense mutation in the Apc gene at R850, which is designated ApcMin/+ (Multiple intestinal neoplasia), develop intestinal adenomas. Several genetic modifier loci of Min (Mom) were previously mapped, but so far, most of the underlying genes have not been identified. To identify novel modifier loci associated with ApcMin/+, we performed quantitative trait loci (QTL) analysis for polyp development using 49 F1 crosses between different Collaborative Cross (CC) lines and C57BL/6 J-ApcMin/+mice. The CC population is a genetic reference panel of recombinant inbred lines, each line independently descended from eight genetically diverse founder strains. C57BL/6 J-ApcMin/+ males were mated with females from 49 CC lines. F1 offspring were terminated at 23 weeks and polyp counts from three sub-regions (SB1-3) of small intestinal and colon were recorded. RESULTS The number of polyps in all these sub-regions and colon varied significantly between the different CC lines. At 95% genome-wide significance, we mapped nine novel QTL for variation in polyp number, with distinct QTL associated with each intestinal sub-region. QTL confidence intervals varied in width between 2.63-17.79 Mb. We extracted all genes in the mapped QTL at 90 and 95% CI levels using the BioInfoMiner online platform to extract, significantly enriched pathways and key linker genes, that act as regulatory and orchestrators of the phenotypic landscape associated with the ApcMin/+ mutation. CONCLUSIONS Genomic structure of the CC lines has allowed us to identify novel modifiers and confirmed some of the previously mapped modifiers. Key genes involved mainly in metabolic and immunological processes were identified. Future steps in this analysis will be to identify regulatory elements - and possible epistatic effects - located in the mapped QTL.
Collapse
Affiliation(s)
- Alexandra Dorman
- Department of Clinical Microbiology & Immunology, Sackler Faculty of Medicine, Ramat Aviv, 69978 Tel-Aviv, Israel
| | - Ilona Binenbaum
- Department of Biology, University of Patras, Patras, Greece
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Hanifa J. Abu-Toamih Atamni
- Department of Clinical Microbiology & Immunology, Sackler Faculty of Medicine, Ramat Aviv, 69978 Tel-Aviv, Israel
| | | | - Ian Tomlinson
- Cancer Research UK Edinburgh Centre, Charles and Ethel Barr Chair of Cancer Research, University of Edinburgh, Edinburgh, UK
| | - Richard Mott
- Department of Genetics, University Collage of London, London, UK
| | - Fuad A. Iraqi
- Department of Clinical Microbiology & Immunology, Sackler Faculty of Medicine, Ramat Aviv, 69978 Tel-Aviv, Israel
| |
Collapse
|
25
|
Requena F, Abdallah HH, García A, Nitschké P, Romana S, Malan V, Rausell A. CNVxplorer: a web tool to assist clinical interpretation of CNVs in rare disease patients. Nucleic Acids Res 2021; 49:W93-W103. [PMID: 34019647 PMCID: PMC8262689 DOI: 10.1093/nar/gkab347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/12/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
Copy Number Variants (CNVs) are an important cause of rare diseases. Array-based Comparative Genomic Hybridization tests yield a ∼12% diagnostic rate, with ∼8% of patients presenting CNVs of unknown significance. CNVs interpretation is particularly challenging on genomic regions outside of those overlapping with previously reported structural variants or disease-associated genes. Recent studies showed that a more comprehensive evaluation of CNV features, leveraging both coding and non-coding impacts, can significantly improve diagnostic rates. However, currently available CNV interpretation tools are mostly gene-centric or provide only non-interactive annotations difficult to assess in the clinical practice. Here, we present CNVxplorer, a web server suited for the functional assessment of CNVs in a clinical diagnostic setting. CNVxplorer mines a comprehensive set of clinical, genomic, and epigenomic features associated with CNVs. It provides sequence constraint metrics, impact on regulatory elements and topologically associating domains, as well as expression patterns. Analyses offered cover (a) agreement with patient phenotypes; (b) visualizations of associations among genes, regulatory elements and transcription factors; (c) enrichment on functional and pathway annotations and (d) co-occurrence of terms across PubMed publications related to the query CNVs. A flexible evaluation workflow allows dynamic re-interrogation in clinical sessions. CNVxplorer is publicly available at http://cnvxplorer.com.
Collapse
Affiliation(s)
- Francisco Requena
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Clinical Bioinformatics Laboratory, Imagine Institute, INSERM UMR1163, F-75015 Paris, France
| | - Hamza Hadj Abdallah
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Service de Cytogénétique, Hôpital Necker-Enfants Malades, APHP, F-75015 Paris, France
| | - Alejandro García
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Clinical Bioinformatics Laboratory, Imagine Institute, INSERM UMR1163, F-75015 Paris, France
| | - Patrick Nitschké
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Plateforme de Bioinformatique, Université Paris Descartes, F-75015 Paris, France
| | - Sergi Romana
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Service de Cytogénétique, Hôpital Necker-Enfants Malades, APHP, F-75015 Paris, France
| | - Valérie Malan
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Service de Cytogénétique, Hôpital Necker-Enfants Malades, APHP, F-75015 Paris, France
| | - Antonio Rausell
- Université de Paris, Institut Imagine, F-75006 Paris, France
- Clinical Bioinformatics Laboratory, Imagine Institute, INSERM UMR1163, F-75015 Paris, France
- Service de Génétique Moleculaire, Hôpital Necker-Enfants Malades, APHP, F-75015, Paris, France
| |
Collapse
|
26
|
Jeon M, Jagodnik KM, Kropiwnicki E, Stein DJ, Ma'ayan A. Prioritizing Pain-Associated Targets with Machine Learning. Biochemistry 2021; 60:1430-1446. [PMID: 33606503 DOI: 10.1021/acs.biochem.0c00930] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
While hundreds of genes have been associated with pain, much of the molecular mechanisms of pain remain unknown. As a result, current analgesics are limited to few clinically validated targets. Here, we trained a machine learning (ML) ensemble model to predict new targets for 17 categories of pain. The model utilizes features from transcriptomics, proteomics, and gene ontology to prioritize targets for modulating pain. We focused on identifying novel G-protein-coupled receptors (GPCRs), ion channels, and protein kinases because these proteins represent the most successful drug target families. The performance of the model to predict novel pain targets is 0.839 on average based on AUROC, while the predictions for arthritis had the highest accuracy (AUROC = 0.929). The model predicts hundreds of novel targets for pain; for example, GPR132 and GPR109B are highly ranked GPCRs for rheumatoid arthritis. Overall, gene-pain association predictions cluster into three groups that are enriched for cytokine, calcium, and GABA-related cell signaling pathways. These predictions can serve as a foundation for future experimental exploration to advance the development of safer and more effective analgesics.
Collapse
Affiliation(s)
- Minji Jeon
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Kathleen M Jagodnik
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Eryk Kropiwnicki
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Daniel J Stein
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1603, New York, New York 10029, United States
| |
Collapse
|
27
|
Lim N, Tesar S, Belmadani M, Poirier-Morency G, Mancarci BO, Sicherman J, Jacobson M, Leong J, Tan P, Pavlidis P. Curation of over 10 000 transcriptomic studies to enable data reuse. Database (Oxford) 2021; 2021:6143045. [PMID: 33599246 PMCID: PMC7904053 DOI: 10.1093/database/baab006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/09/2020] [Accepted: 01/28/2021] [Indexed: 01/07/2023]
Abstract
Vast amounts of transcriptomic data reside in public repositories, but effective reuse remains challenging. Issues include unstructured dataset metadata, inconsistent data processing and quality control, and inconsistent probe-gene mappings across microarray technologies. Thus, extensive curation and data reprocessing are necessary prior to any reuse. The Gemma bioinformatics system was created to help address these issues. Gemma consists of a database of curated transcriptomic datasets, analytical software, a web interface and web services. Here we present an update on Gemma's holdings, data processing and analysis pipelines, our curation guidelines, and software features. As of June 2020, Gemma contains 10 811 manually curated datasets (primarily human, mouse and rat), over 395 000 samples and hundreds of curated transcriptomic platforms (both microarray and RNA sequencing). Dataset topics were represented with 10 215 distinct terms from 12 ontologies, for a total of 54 316 topic annotations (mean topics/dataset = 5.2). While Gemma has broad coverage of conditions and tissues, it captures a large majority of available brain-related datasets, accounting for 34% of its holdings. Users can access the curated data and differential expression analyses through the Gemma website, RESTful service and an R package. Database URL: https://gemma.msl.ubc.ca/home.html.
Collapse
Affiliation(s)
- Nathaniel Lim
- Genome Science and Technology Graduate Program, University of British Columbia, Vancouver, BC V6T1Z4, Canada,Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | - Stepan Tesar
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | - Manuel Belmadani
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | - Guillaume Poirier-Morency
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | - Burak Ogan Mancarci
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada,Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Jordan Sicherman
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada,Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Matthew Jacobson
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | - Justin Leong
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | - Patrick Tan
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T1Z4, Canada
| | | |
Collapse
|
28
|
Baldarelli RM, Smith CM, Finger JH, Hayamizu TF, McCright IJ, Xu J, Shaw DR, Beal JS, Blodgett O, Campbell J, Corbani LE, Frost PJ, Giannatto SC, Miers DB, Kadin JA, Richardson JE, Ringwald M. The mouse Gene Expression Database (GXD): 2021 update. Nucleic Acids Res 2021; 49:D924-D931. [PMID: 33104772 PMCID: PMC7778941 DOI: 10.1093/nar/gkaa914] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 01/05/2023] Open
Abstract
The Gene Expression Database (GXD; www.informatics.jax.org/expression.shtml) is an extensive and well-curated community resource of mouse developmental gene expression information. For many years, GXD has collected and integrated data from RNA in situ hybridization, immunohistochemistry, RT-PCR, northern blot, and western blot experiments through curation of the scientific literature and by collaborations with large-scale expression projects. Since our last report in 2019, we have continued to acquire these classical types of expression data; developed a searchable index of RNA-Seq and microarray experiments that allows users to quickly and reliably find specific mouse expression studies in ArrayExpress (https://www.ebi.ac.uk/arrayexpress/) and GEO (https://www.ncbi.nlm.nih.gov/geo/); and expanded GXD to include RNA-Seq data. Uniformly processed RNA-Seq data are imported from the EBI Expression Atlas and then integrated with the other types of expression data in GXD, and with the genetic, functional, phenotypic and disease-related information in Mouse Genome Informatics (MGI). This integration has made the RNA-Seq data accessible via GXD’s enhanced searching and filtering capabilities. Further, we have embedded the Morpheus heat map utility into the GXD user interface to provide additional tools for display and analysis of RNA-Seq data, including heat map visualization, sorting, filtering, hierarchical clustering, nearest neighbors analysis and visual enrichment.
Collapse
Affiliation(s)
| | | | | | - Terry F Hayamizu
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Jingxia Xu
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - David R Shaw
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Jonathan S Beal
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Olin Blodgett
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Jeffrey Campbell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Lori E Corbani
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Pete J Frost
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Dave B Miers
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - James A Kadin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Martin Ringwald
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| |
Collapse
|
29
|
Köhler S, Gargano M, Matentzoglu N, Carmody LC, Lewis-Smith D, Vasilevsky NA, Danis D, Balagura G, Baynam G, Brower AM, Callahan TJ, Chute CG, Est JL, Galer PD, Ganesan S, Griese M, Haimel M, Pazmandi J, Hanauer M, Harris NL, Hartnett M, Hastreiter M, Hauck F, He Y, Jeske T, Kearney H, Kindle G, Klein C, Knoflach K, Krause R, Lagorce D, McMurry JA, Miller JA, Munoz-Torres M, Peters RL, Rapp CK, Rath AM, Rind SA, Rosenberg A, Segal MM, Seidel MG, Smedley D, Talmy T, Thomas Y, Wiafe SA, Xian J, Yüksel Z, Helbig I, Mungall CJ, Haendel MA, Robinson PN. The Human Phenotype Ontology in 2021. Nucleic Acids Res 2021; 49:D1207-D1217. [PMID: 33264411 PMCID: PMC7778952 DOI: 10.1093/nar/gkaa1043] [Citation(s) in RCA: 674] [Impact Index Per Article: 168.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/11/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
The Human Phenotype Ontology (HPO, https://hpo.jax.org) was launched in 2008 to provide a comprehensive logical standard to describe and computationally analyze phenotypic abnormalities found in human disease. The HPO is now a worldwide standard for phenotype exchange. The HPO has grown steadily since its inception due to considerable contributions from clinical experts and researchers from a diverse range of disciplines. Here, we present recent major extensions of the HPO for neurology, nephrology, immunology, pulmonology, newborn screening, and other areas. For example, the seizure subontology now reflects the International League Against Epilepsy (ILAE) guidelines and these enhancements have already shown clinical validity. We present new efforts to harmonize computational definitions of phenotypic abnormalities across the HPO and multiple phenotype ontologies used for animal models of disease. These efforts will benefit software such as Exomiser by improving the accuracy and scope of cross-species phenotype matching. The computational modeling strategy used by the HPO to define disease entities and phenotypic features and distinguish between them is explained in detail.We also report on recent efforts to translate the HPO into indigenous languages. Finally, we summarize recent advances in the use of HPO in electronic health record systems.
Collapse
Affiliation(s)
| | - Michael Gargano
- Monarch Initiative
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Nicolas Matentzoglu
- Monarch Initiative
- Semanticly Ltd, London, UK
- European Bioinformatics Institute (EMBL-EBI)
| | - Leigh C Carmody
- Monarch Initiative
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - David Lewis-Smith
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Clinical Neurosciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Nicole A Vasilevsky
- Monarch Initiative
- Oregon Clinical & Translational Research Institute, Oregon Health & Science University
| | | | - Ganna Balagura
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Child Health, University of Genoa, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS ‘G. Gaslini’ Institute, Genoa, Italy
| | - Gareth Baynam
- Western Australian Register of Developmental Anomalies, King Edward memorial Hospital, Perth, Australia
- Telethon Kids Institute and the Division of Paediatrics, Faculty of Helath and Medical Sciences, University of Western Australia, Perth, Australia
| | - Amy M Brower
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD, USA
| | - Tiffany J Callahan
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Colorado, USA
| | | | - Johanna L Est
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peter D Galer
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shiva Ganesan
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthias Griese
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Ludwig-Maximilians University, German Center for Lung Research (DZL), Munich, Germany
| | - Matthias Haimel
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Julia Pazmandi
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| | - Marc Hanauer
- INSERM, US14––Orphanet, Plateforme Maladies Rares, Paris, France
| | - Nomi L Harris
- Monarch Initiative
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley CA, USA
| | - Michael J Hartnett
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD, USA
| | - Maximilian Hastreiter
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian Hauck
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Yongqun He
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tim Jeske
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hugh Kearney
- FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, Ireland
| | - Gerhard Kindle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI). Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
- Centre for Biobanking FREEZE, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katrin Knoflach
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Ludwig-Maximilians University, German Center for Lung Research (DZL), Munich, Germany
| | - Roland Krause
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - David Lagorce
- INSERM, US14––Orphanet, Plateforme Maladies Rares, Paris, France
| | - Julie A McMurry
- Monarch Initiative
- Translational and Integrative Sciences Center, Department of Environmental and Molecular Toxicology, Oregon State University, OR, USA
| | - Jillian A Miller
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD, USA
| | - Monica C Munoz-Torres
- Monarch Initiative
- Translational and Integrative Sciences Center, Department of Environmental and Molecular Toxicology, Oregon State University, OR, USA
| | - Rebecca L Peters
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD, USA
| | - Christina K Rapp
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Ludwig-Maximilians University, German Center for Lung Research (DZL), Munich, Germany
| | - Ana M Rath
- INSERM, US14––Orphanet, Plateforme Maladies Rares, Paris, France
| | - Shahmir A Rind
- WA Register of Developmental Anomalies
- Curtin University, Western Australia, Australia
| | - Avi Z Rosenberg
- Division of Kidney-Urologic Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Markus G Seidel
- Research Unit for Pediatric Hematology and Immunology, Division of Pediatric Hemato-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Damian Smedley
- The William Harvey Research Institute, Charterhouse Square Barts and the London School of Medicine and Dentistry Queen Mary University of London, London EC1M 6BQ, UK
| | - Tomer Talmy
- Genomic Research Department, Emedgene Technologies, Tel Aviv, Israel
- Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Yarlalu Thomas
- West Australian Register of Developmental Anomalies, East Perth, WA, Australia
| | | | - Julie Xian
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, PA, USA
| | - Zafer Yüksel
- Human Genetics, Bioscientia GmbH, Ingelheim, Germany
| | - Ingo Helbig
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christopher J Mungall
- Monarch Initiative
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley CA, USA
| | - Melissa A Haendel
- Monarch Initiative
- Oregon Clinical & Translational Research Institute, Oregon Health & Science University
- Translational and Integrative Sciences Center, Department of Environmental and Molecular Toxicology, Oregon State University, OR, USA
| | - Peter N Robinson
- Monarch Initiative
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| |
Collapse
|
30
|
Kanza S, Graham Frey J. Semantic Technologies in Drug Discovery. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11520-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
31
|
Binenbaum I, Atamni HAT, Fotakis G, Kontogianni G, Koutsandreas T, Pilalis E, Mott R, Himmelbauer H, Iraqi FA, Chatziioannou AA. Container-aided integrative QTL and RNA-seq analysis of Collaborative Cross mice supports distinct sex-oriented molecular modes of response in obesity. BMC Genomics 2020; 21:761. [PMID: 33143653 PMCID: PMC7640698 DOI: 10.1186/s12864-020-07173-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/21/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The Collaborative Cross (CC) mouse population is a valuable resource to study the genetic basis of complex traits, such as obesity. Although the development of obesity is influenced by environmental factors, underlying genetic mechanisms play a crucial role in the response to these factors. The interplay between the genetic background and the gene expression pattern can provide further insight into this response, but we lack robust and easily reproducible workflows to integrate genomic and transcriptomic information in the CC mouse population. RESULTS We established an automated and reproducible integrative workflow to analyse complex traits in the CC mouse genetic reference panel at the genomic and transcriptomic levels. We implemented the analytical workflow to assess the underlying genetic mechanisms of host susceptibility to diet induced obesity and integrated these results with diet induced changes in the hepatic gene expression of susceptible and resistant mice. Hepatic gene expression differs significantly between obese and non-obese mice, with a significant sex effect, where male and female mice exhibit different responses and coping mechanisms. CONCLUSION Integration of the data showed that different genes but similar pathways are involved in the genetic susceptibility and disturbed in diet induced obesity. Genetic mechanisms underlying susceptibility to high-fat diet induced obesity are different in female and male mice. The clear distinction we observed in the systemic response to the high-fat diet challenge and to obesity between male and female mice points to the need for further research into distinct sex-related mechanisms in metabolic disease.
Collapse
Affiliation(s)
- Ilona Binenbaum
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
- Department of Biology, University of Patras, Patras, Greece
| | - Hanifa Abu-Toamih Atamni
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Georgios Fotakis
- Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
- e-NIOS PC, Kallithea, Athens, Greece
| | - Georgia Kontogianni
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Theodoros Koutsandreas
- e-NIOS PC, Kallithea, Athens, Greece
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Eleftherios Pilalis
- e-NIOS PC, Kallithea, Athens, Greece
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Richard Mott
- Department of Genetics, University College of London, London, UK
| | - Heinz Himmelbauer
- Institute of Computational Biology, Department of Biotechnology, University of Life Sciences and Natural Resources, Vienna (BOKU), Vienna, Austria
- Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Aristotelis A Chatziioannou
- e-NIOS PC, Kallithea, Athens, Greece.
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
32
|
Fernando PC, Mabee PM, Zeng E. Integration of anatomy ontology data with protein-protein interaction networks improves the candidate gene prediction accuracy for anatomical entities. BMC Bioinformatics 2020; 21:442. [PMID: 33028186 PMCID: PMC7542696 DOI: 10.1186/s12859-020-03773-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023] Open
Abstract
Background Identification of genes responsible for anatomical entities is a major requirement in many fields including developmental biology, medicine, and agriculture. Current wet lab techniques used for this purpose, such as gene knockout, are high in resource and time consumption. Protein–protein interaction (PPI) networks are frequently used to predict disease genes for humans and gene candidates for molecular functions, but they are rarely used to predict genes for anatomical entities. Moreover, PPI networks suffer from network quality issues, which can be a limitation for their usage in predicting candidate genes. Therefore, we developed an integrative framework to improve the candidate gene prediction accuracy for anatomical entities by combining existing experimental knowledge about gene-anatomical entity relationships with PPI networks using anatomy ontology annotations. We hypothesized that this integration improves the quality of the PPI networks by reducing the number of false positive and false negative interactions and is better optimized to predict candidate genes for anatomical entities. We used existing Uberon anatomical entity annotations for zebrafish and mouse genes to construct gene networks by calculating semantic similarity between the genes. These anatomy-based gene networks were semantic networks, as they were constructed based on the anatomy ontology annotations that were obtained from the experimental data in the literature. We integrated these anatomy-based gene networks with mouse and zebrafish PPI networks retrieved from the STRING database and compared the performance of their network-based candidate gene predictions. Results According to evaluations of candidate gene prediction performance tested under four different semantic similarity calculation methods (Lin, Resnik, Schlicker, and Wang), the integrated networks, which were semantically improved PPI networks, showed better performances by having higher area under the curve values for receiver operating characteristic and precision-recall curves than PPI networks for both zebrafish and mouse. Conclusion Integration of existing experimental knowledge about gene-anatomical entity relationships with PPI networks via anatomy ontology improved the candidate gene prediction accuracy and optimized them for predicting candidate genes for anatomical entities.
Collapse
Affiliation(s)
- Pasan C Fernando
- Department of Biology, University of South Dakota, Vermillion, SD, USA.
| | - Paula M Mabee
- Department of Biology, University of South Dakota, Vermillion, SD, USA.,National Ecological Observatory Network, Battelle Memorial Institute, 1685 38th St., Suite 100, Boulder, CO, 80301, USA
| | - Erliang Zeng
- Division of Biostatistics and Computational Biology, College of Dentistry, University of Iowa, Iowa City, IA, USA. .,Department of Preventive and Community Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, USA. .,Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA. .,Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
33
|
Bogue MA, Philip VM, Walton DO, Grubb SC, Dunn MH, Kolishovski G, Emerson J, Mukherjee G, Stearns T, He H, Sinha V, Kadakkuzha B, Kunde-Ramamoorthy G, Chesler EJ. Mouse Phenome Database: a data repository and analysis suite for curated primary mouse phenotype data. Nucleic Acids Res 2020; 48:D716-D723. [PMID: 31696236 PMCID: PMC7145612 DOI: 10.1093/nar/gkz1032] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/27/2023] Open
Abstract
The Mouse Phenome Database (MPD; https://phenome.jax.org) is a widely accessed and highly functional data repository housing primary phenotype data for the laboratory mouse accessible via APIs and providing tools to analyze and visualize those data. Data come from investigators around the world and represent a broad scope of phenotyping endpoints and disease-related traits in naïve mice and those exposed to drugs, environmental agents or other treatments. MPD houses rigorously curated per-animal data with detailed protocols. Public ontologies and controlled vocabularies are used for annotation. In addition to phenotype tools, genetic analysis tools enable users to integrate and interpret genome–phenome relations across the database. Strain types and populations include inbred, recombinant inbred, F1 hybrid, transgenic, targeted mutants, chromosome substitution, Collaborative Cross, Diversity Outbred and other mapping populations. Our new analysis tools allow users to apply selected data in an integrated fashion to address problems in trait associations, reproducibility, polygenic syndrome model selection and multi-trait modeling. As we refine these tools and approaches, we will continue to provide users a means to identify consistent, quality studies that have high translational relevance.
Collapse
Affiliation(s)
- Molly A Bogue
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | - Vivek M Philip
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | - David O Walton
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | | | - Matthew H Dunn
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | | | - Jake Emerson
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | | | | | - Hao He
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | - Vinita Sinha
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | | | | | | |
Collapse
|
34
|
Shefchek KA, Harris NL, Gargano M, Matentzoglu N, Unni D, Brush M, Keith D, Conlin T, Vasilevsky N, Zhang XA, Balhoff JP, Babb L, Bello SM, Blau H, Bradford Y, Carbon S, Carmody L, Chan LE, Cipriani V, Cuzick A, Della Rocca M, Dunn N, Essaid S, Fey P, Grove C, Gourdine JP, Hamosh A, Harris M, Helbig I, Hoatlin M, Joachimiak M, Jupp S, Lett KB, Lewis SE, McNamara C, Pendlington ZM, Pilgrim C, Putman T, Ravanmehr V, Reese J, Riggs E, Robb S, Roncaglia P, Seager J, Segerdell E, Similuk M, Storm AL, Thaxon C, Thessen A, Jacobsen JOB, McMurry JA, Groza T, Köhler S, Smedley D, Robinson PN, Mungall CJ, Haendel MA, Munoz-Torres MC, Osumi-Sutherland D. The Monarch Initiative in 2019: an integrative data and analytic platform connecting phenotypes to genotypes across species. Nucleic Acids Res 2020; 48:D704-D715. [PMID: 31701156 PMCID: PMC7056945 DOI: 10.1093/nar/gkz997] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022] Open
Abstract
In biology and biomedicine, relating phenotypic outcomes with genetic variation and environmental factors remains a challenge: patient phenotypes may not match known diseases, candidate variants may be in genes that haven’t been characterized, research organisms may not recapitulate human or veterinary diseases, environmental factors affecting disease outcomes are unknown or undocumented, and many resources must be queried to find potentially significant phenotypic associations. The Monarch Initiative (https://monarchinitiative.org) integrates information on genes, variants, genotypes, phenotypes and diseases in a variety of species, and allows powerful ontology-based search. We develop many widely adopted ontologies that together enable sophisticated computational analysis, mechanistic discovery and diagnostics of Mendelian diseases. Our algorithms and tools are widely used to identify animal models of human disease through phenotypic similarity, for differential diagnostics and to facilitate translational research. Launched in 2015, Monarch has grown with regards to data (new organisms, more sources, better modeling); new API and standards; ontologies (new Mondo unified disease ontology, improvements to ontologies such as HPO and uPheno); user interface (a redesigned website); and community development. Monarch data, algorithms and tools are being used and extended by resources such as GA4GH and NCATS Translator, among others, to aid mechanistic discovery and diagnostics.
Collapse
Affiliation(s)
- Kent A Shefchek
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Nomi L Harris
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Michael Gargano
- The Jackson Laboratory For Genomic Medicine, Farmington, CT 06032, USA
| | - Nicolas Matentzoglu
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Deepak Unni
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Matthew Brush
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Daniel Keith
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Tom Conlin
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Nicole Vasilevsky
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - James P Balhoff
- Renaissance Computing Institute at UNC, Chapel Hill, NC 27517, USA
| | - Larry Babb
- Broad Institute, Cambridge, MA 02142, USA
| | | | - Hannah Blau
- The Jackson Laboratory For Genomic Medicine, Farmington, CT 06032, USA
| | - Yvonne Bradford
- Institute of Neuroscience, University of Oregon, Eugene, OR 97401, USA
| | - Seth Carbon
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Leigh Carmody
- The Jackson Laboratory For Genomic Medicine, Farmington, CT 06032, USA
| | - Lauren E Chan
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Valentina Cipriani
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | | | - Maria Della Rocca
- Office of Rare Diseases Research (ORDR), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nathan Dunn
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Shahim Essaid
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Petra Fey
- dictyBase, Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chris Grove
- California Institute of Technology, Pasadena, CA 91125, USA
| | - Jean-Phillipe Gourdine
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ada Hamosh
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.,Department of Neuropediatrics, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany.,Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Maureen Hoatlin
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Marcin Joachimiak
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Simon Jupp
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Kenneth B Lett
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Suzanna E Lewis
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | | | - Zoë M Pendlington
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | | | - Tim Putman
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Vida Ravanmehr
- The Jackson Laboratory For Genomic Medicine, Farmington, CT 06032, USA
| | - Justin Reese
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Erin Riggs
- Autism & Developmental Medicine Institute, Geisinger, Danville, PA 17837, USA
| | - Sofia Robb
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Paola Roncaglia
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | | | - Erik Segerdell
- Xenbase, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Morgan Similuk
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea L Storm
- Office of Rare Diseases Research (ORDR), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Courtney Thaxon
- University of North Carolina Medical School, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Anne Thessen
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Julius O B Jacobsen
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Julie A McMurry
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA
| | | | - Sebastian Köhler
- Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Damian Smedley
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Peter N Robinson
- The Jackson Laboratory For Genomic Medicine, Farmington, CT 06032, USA
| | - Christopher J Mungall
- Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Melissa A Haendel
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.,Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Monica C Munoz-Torres
- Center for Genome Research and Biocomputing, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - David Osumi-Sutherland
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| |
Collapse
|
35
|
Sickle Cell Disease Ontology Working Group
AdekileAdekunleAnieKofi AHamdaCherif BenBrownBiobeleBukiniDaimaCampbellAndrewChaouchMelekChimusaEmileChunda-LiyokaCatherineDennis-AntwiJemimaDerebailVimal KFlor-ParkMiriamGeardAmyGhediraKaisHaendelMelissaHanchardNeil AHotchkissJadeJonasMarioIbrahimMuntaserIngramClairInusaBabaJimohAdijat OzohuJuppSimonKamgaKarenKashimZainab AbimbolaKnight-MaddenJenniferLandouréGuidaLopez-SallPhilomeneMakaniJulieMalasaLeonardMasekoamengTshepisoMazanduGastonMnikaKhuthalaMulderNicolaMunungNchangwi SyntiaMunubeDeogratiasMwitaLiberataNembawareVictoriaNnoduObiageliOfori-AcquahSolomonOhene-FrempongKwakuOsei-AkotoAlexPaintsilVivianPanjiSumirRahimyMohamed CherifRoyalCharmaineSangedaRaphael ZTayoBamideleTiouiriInesTluwayFurahiniTreadwellMarshaTshiloloLeonVasilevskyNicoleWaiswaKasadhakawo MusaWonkamAmbroise. The Sickle Cell Disease Ontology: enabling universal sickle cell-based knowledge representation. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2020; 2019:5626537. [PMID: 31769834 PMCID: PMC6878945 DOI: 10.1093/database/baz118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/27/2022]
Abstract
Sickle cell disease (SCD) is one of the most common monogenic diseases in humans with multiple phenotypic expressions that can manifest as both acute and chronic complications. Although described more than a century ago, challenges in comprehensive disease management and collaborative research on this disease are compounded by the complex molecular and clinical phenotypes of SCD, environmental and psychosocial factors, limited therapeutic options and ambiguous terminology. This ambiguous terminology has hampered the integration and interoperability of existing SCD knowledge, and SCD research translation. The SCD Ontology (SCDO), which is a community-driven integrative and universal knowledge representation system for SCD, overcomes this issue by providing a controlled vocabulary developed by a group of experts in both SCD and ontology design. SCDO is the first and most comprehensive standardized human- and machine-readable resource that unambiguously represents terminology and concepts about SCD for researchers, patients and clinicians. It is built around the central concept ‘hemoglobinopathy’, allowing inclusion of non-SCD haemoglobinopathies, such as thalassaemias, which may interfere with or influence SCD phenotypic manifestations. This collaboratively developed ontology constitutes a comprehensive knowledge management system and standardized terminology of various SCD-related factors. The SCDO will promote interoperability of different research datasets, facilitate seamless data sharing and collaborations, including meta-analyses within the SCD community, and support the development and curation of data-basing and clinical informatics in SCD.
Collapse
Affiliation(s)
- Sickle Cell Disease Ontology Working Group
AdekileAdekunleAnieKofi AHamdaCherif BenBrownBiobeleBukiniDaimaCampbellAndrewChaouchMelekChimusaEmileChunda-LiyokaCatherineDennis-AntwiJemimaDerebailVimal KFlor-ParkMiriamGeardAmyGhediraKaisHaendelMelissaHanchardNeil AHotchkissJadeJonasMarioIbrahimMuntaserIngramClairInusaBabaJimohAdijat OzohuJuppSimonKamgaKarenKashimZainab AbimbolaKnight-MaddenJenniferLandouréGuidaLopez-SallPhilomeneMakaniJulieMalasaLeonardMasekoamengTshepisoMazanduGastonMnikaKhuthalaMulderNicolaMunungNchangwi SyntiaMunubeDeogratiasMwitaLiberataNembawareVictoriaNnoduObiageliOfori-AcquahSolomonOhene-FrempongKwakuOsei-AkotoAlexPaintsilVivianPanjiSumirRahimyMohamed CherifRoyalCharmaineSangedaRaphael ZTayoBamideleTiouiriInesTluwayFurahiniTreadwellMarshaTshiloloLeonVasilevskyNicoleWaiswaKasadhakawo MusaWonkamAmbroise
- Computational Biology Division, Institute of Infectious Disease and Molecular Medicine, N1.05, Werner Beit North, Faculty of Health Sciences, Anzio Road, Observatory, 7925 Cape Town, South Africa
- Division of Human Genetics, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925
Cape Town, South Africa
- Corresponding author: Tel: 0027 21 4066058;
| |
Collapse
|
36
|
Köhler S, Carmody L, Vasilevsky N, Jacobsen JOB, Danis D, Gourdine JP, Gargano M, Harris NL, Matentzoglu N, McMurry JA, Osumi-Sutherland D, Cipriani V, Balhoff JP, Conlin T, Blau H, Baynam G, Palmer R, Gratian D, Dawkins H, Segal M, Jansen AC, Muaz A, Chang WH, Bergerson J, Laulederkind SJF, Yüksel Z, Beltran S, Freeman AF, Sergouniotis PI, Durkin D, Storm AL, Hanauer M, Brudno M, Bello SM, Sincan M, Rageth K, Wheeler MT, Oegema R, Lourghi H, Della Rocca MG, Thompson R, Castellanos F, Priest J, Cunningham-Rundles C, Hegde A, Lovering RC, Hajek C, Olry A, Notarangelo L, Similuk M, Zhang XA, Gómez-Andrés D, Lochmüller H, Dollfus H, Rosenzweig S, Marwaha S, Rath A, Sullivan K, Smith C, Milner JD, Leroux D, Boerkoel CF, Klion A, Carter MC, Groza T, Smedley D, Haendel MA, Mungall C, Robinson PN. Expansion of the Human Phenotype Ontology (HPO) knowledge base and resources. Nucleic Acids Res 2020; 47:D1018-D1027. [PMID: 30476213 PMCID: PMC6324074 DOI: 10.1093/nar/gky1105] [Citation(s) in RCA: 438] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022] Open
Abstract
The Human Phenotype Ontology (HPO)—a standardized vocabulary of phenotypic abnormalities associated with 7000+ diseases—is used by thousands of researchers, clinicians, informaticians and electronic health record systems around the world. Its detailed descriptions of clinical abnormalities and computable disease definitions have made HPO the de facto standard for deep phenotyping in the field of rare disease. The HPO’s interoperability with other ontologies has enabled it to be used to improve diagnostic accuracy by incorporating model organism data. It also plays a key role in the popular Exomiser tool, which identifies potential disease-causing variants from whole-exome or whole-genome sequencing data. Since the HPO was first introduced in 2008, its users have become both more numerous and more diverse. To meet these emerging needs, the project has added new content, language translations, mappings and computational tooling, as well as integrations with external community data. The HPO continues to collaborate with clinical adopters to improve specific areas of the ontology and extend standardized disease descriptions. The newly redesigned HPO website (www.human-phenotype-ontology.org) simplifies browsing terms and exploring clinical features, diseases, and human genes.
Collapse
Affiliation(s)
- Sebastian Köhler
- Charité Centrum für Therapieforschung, Charité-Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany.,Einstein Center Digital Future, Berlin 10117, Germany.,Monarch Initiative, monarchinitiative.org
| | - Leigh Carmody
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Nicole Vasilevsky
- Monarch Initiative, monarchinitiative.org.,Oregon Health & Science University, Portland, OR 97217, USA
| | - Julius O B Jacobsen
- Monarch Initiative, monarchinitiative.org.,Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London EC1M 6BQ, UK
| | - Daniel Danis
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Jean-Philippe Gourdine
- Monarch Initiative, monarchinitiative.org.,Oregon Health & Science University, Portland, OR 97217, USA
| | - Michael Gargano
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Nomi L Harris
- Monarch Initiative, monarchinitiative.org.,Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Nicolas Matentzoglu
- Monarch Initiative, monarchinitiative.org.,European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - Julie A McMurry
- Monarch Initiative, monarchinitiative.org.,Linus Pauling institute, Oregon State University, Corvallis, OR, USA
| | - David Osumi-Sutherland
- Monarch Initiative, monarchinitiative.org.,European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - Valentina Cipriani
- Monarch Initiative, monarchinitiative.org.,William Harvey Research Institute, Queen Mary University College of London.,UCL Genetics Institute, University College of London.,UCL Institute of Ophthalmology, University College of London
| | - James P Balhoff
- Monarch Initiative, monarchinitiative.org.,Renaissance Computing Institute, University of North Carolina at Chapel Hill
| | - Tom Conlin
- Monarch Initiative, monarchinitiative.org.,Linus Pauling institute, Oregon State University, Corvallis, OR, USA
| | - Hannah Blau
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Gareth Baynam
- Western Australian Register of Developmental Anomalies and Genetic Services of Western Australia, Department of Health, Government of Western Australia, WA, Australia.,School of Paediatrics and Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Spatial Sciences, Department of Science and Engineering, Curtin University, Perth, WA, Australia.,The Office of Population Health Genomics, Department of Health, Government of Western Australia, Perth, WA, Australia
| | - Richard Palmer
- Spatial Sciences, Department of Science and Engineering, Curtin University, Perth, WA, Australia
| | - Dylan Gratian
- Western Australian Register of Developmental Anomalies and Genetic Services of Western Australia, Department of Health, Government of Western Australia, WA, Australia
| | - Hugh Dawkins
- The Office of Population Health Genomics, Department of Health, Government of Western Australia, Perth, WA, Australia
| | | | - Anna C Jansen
- Neurogenetics Research Group, Vrije Universiteit Brussel, Brussels, Belgium.,Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel, Brussels, Belgium
| | - Ahmed Muaz
- Monarch Initiative, monarchinitiative.org.,Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Willie H Chang
- Centre for Computational Medicine, Hospital for Sick Children and Department of Computer Science, University of Toronto, Toronto, Canada
| | - Jenna Bergerson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stanley J F Laulederkind
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, 8701 Watertown Plank Road Milwaukee, WI 53226, USA
| | | | - Sergi Beltran
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona 08028, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Daniel Durkin
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrea L Storm
- ICF, Rockville, MD, USA.,National Center for Advancing Translational Sciences, Office of Rare Diseases Research, National Institutes of Health, Bethesda, MD, USA
| | - Marc Hanauer
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Michael Brudno
- Centre for Computational Medicine, Hospital for Sick Children and Department of Computer Science, University of Toronto, Toronto, Canada
| | | | - Murat Sincan
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, USA
| | - Kayli Rageth
- Sanford Imagenetics, Sanford Health, Sioux Falls, SD, USA
| | - Matthew T Wheeler
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, the Netherlands
| | - Halima Lourghi
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Maria G Della Rocca
- ICF, Rockville, MD, USA.,National Center for Advancing Translational Sciences, Office of Rare Diseases Research, National Institutes of Health, Bethesda, MD, USA
| | - Rachel Thompson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | - James Priest
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ayushi Hegde
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ruth C Lovering
- Institute of Cardiovascular Science, University College London, UK
| | | | - Annie Olry
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Luigi Notarangelo
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Morgan Similuk
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xingmin A Zhang
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - David Gómez-Andrés
- Child Neurology Unit. Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Hanns Lochmüller
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona 08028, Spain.,Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Hélène Dollfus
- Centre for Rare Eye Diseases CARGO, SENSGENE FSMR Network, Strasbourg University Hospital, Strasbourg, France
| | - Sergio Rosenzweig
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - Shruti Marwaha
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Ana Rath
- INSERM, US14-Orphanet, Plateforme Maladies Rares, 75014 Paris, France
| | - Kathleen Sullivan
- Department of Pediatrics, Division of Allergy Immunology, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | | | - Joshua D Milner
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dorothée Leroux
- Centre for Rare Eye Diseases CARGO, SENSGENE FSMR Network, Strasbourg University Hospital, Strasbourg, France
| | | | - Amy Klion
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melody C Carter
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tudor Groza
- Monarch Initiative, monarchinitiative.org.,Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Damian Smedley
- Monarch Initiative, monarchinitiative.org.,Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London EC1M 6BQ, UK
| | - Melissa A Haendel
- Monarch Initiative, monarchinitiative.org.,Oregon Health & Science University, Portland, OR 97217, USA.,Linus Pauling institute, Oregon State University, Corvallis, OR, USA
| | - Chris Mungall
- Monarch Initiative, monarchinitiative.org.,Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Peter N Robinson
- Monarch Initiative, monarchinitiative.org.,The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.,Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| |
Collapse
|
37
|
Smith CM, Hayamizu TF, Finger JH, Bello SM, McCright IJ, Xu J, Baldarelli RM, Beal JS, Campbell J, Corbani LE, Frost PJ, Lewis JR, Giannatto SC, Miers D, Shaw DR, Kadin JA, Richardson JE, Smith CL, Ringwald M. The mouse Gene Expression Database (GXD): 2019 update. Nucleic Acids Res 2020; 47:D774-D779. [PMID: 30335138 PMCID: PMC6324054 DOI: 10.1093/nar/gky922] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/04/2018] [Indexed: 11/13/2022] Open
Abstract
The mouse Gene Expression Database (GXD) is an extensive, well-curated community resource freely available at www.informatics.jax.org/expression.shtml. Covering all developmental stages, GXD includes data from RNA in situ hybridization, immunohistochemistry, RT-PCR, northern blot and western blot experiments in wild-type and mutant mice. GXD's gene expression information is integrated with the other data in Mouse Genome Informatics and interconnected with other databases, placing these data in the larger biological and biomedical context. Since the last report, the ability of GXD to provide insights into the molecular mechanisms of development and disease has been greatly enhanced by the addition of new data and by the implementation of new web features. These include: improvements to the Differential Gene Expression Data Search, facilitating searches for genes that have been shown to be exclusively expressed in a specified structure and/or developmental stage; an enhanced anatomy browser that now provides access to expression data and phenotype data for a given anatomical structure; direct access to the wild-type gene expression data for the tissues affected in a specific mutant; and a comparison matrix that juxtaposes tissues where a gene is normally expressed against tissues, where mutations in that gene cause abnormalities.
Collapse
Affiliation(s)
| | - Terry F Hayamizu
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Susan M Bello
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Jingxia Xu
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Jonathan S Beal
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Jeffrey Campbell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Lori E Corbani
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Pete J Frost
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Jill R Lewis
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Dave Miers
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - David R Shaw
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - James A Kadin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Cynthia L Smith
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Martin Ringwald
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| |
Collapse
|
38
|
Bioinformatic framework for analysis of transcription factor changes as the molecular link between replicative cellular senescence signaling pathways and carcinogenesis. Biogerontology 2020; 21:357-366. [PMID: 32100207 DOI: 10.1007/s10522-020-09866-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/22/2020] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a natural condition of irreversible cell cycle arrest and apoptotic resistance that occurs in cells exposed to various stress factors, such as replicative stress or overexpression of oncogenes. Unraveling the complex regulation of senescence in cells is essential to strengthen senescence-related therapeutic approaches in cancer, as cellular senescence plays a dual role in tumorigenesis, having both anti- and pro-tumorigenic effects. In our study we created a model of replicative cellular senescence, based on transcriptomic data, including an extra intermediate time-point prior to cells entering senescence, to elucidate the interplay of networks governing cellular senescence with networks involved in tumorigenesis. We reveal specific changes that occur in transcription factor activity at different timepoints before and after cells entering senescence and model the signaling networks that govern these changes.
Collapse
|
39
|
Data-driven method to enhance craniofacial and oral phenotype vocabularies. J Am Dent Assoc 2019; 150:933-939.e2. [PMID: 31668172 DOI: 10.1016/j.adaj.2019.05.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND A significant amount of clinical information captured as free-text narratives could be better used for several applications, such as clinical decision support, ontology development, evidence-based practice, and research. The Human Phenotype Ontology (HPO) is specifically used for semantic comparisons for diagnostic purposes. All these functions require quality coverage of the domain of interest. The authors used natural language processing to capture craniofacial and oral phenotype signatures from electronic health records and then used these signatures for evaluation of existing oral phenotype ontology coverage. METHODS The authors applied a text-processing pipeline based on the clinical Text Analysis and Knowledge Extraction System to annotate the clinical notes with Unified Medical Language System codes. The authors extracted the disease or disorder phenotype terms, which were then compared with HPO terms and their synonyms. RESULTS The authors retrieved 2,153 deidentified clinical notes from 558 patients. Finally, 2,416 unique diseases or disorders phenotype terms were extracted, which included 210 craniofacial or oral phenotype terms. Twenty-six of these phenotypes were not found in the HPO. CONCLUSIONS The authors demonstrated that natural language processing tools could extract relevant phenotype terms from clinical narratives, which could help identify gaps in existing ontologies and enhance craniofacial and dental phenotyping vocabularies. PRACTICAL IMPLICATIONS The expansion of terms in the dental, oral, and craniofacial domains in the HPO is particularly important as the dental community moves toward electronic health records.
Collapse
|
40
|
Network-Based Functional Prediction Augments Genetic Association To Predict Candidate Genes for Histamine Hypersensitivity in Mice. G3-GENES GENOMES GENETICS 2019; 9:4223-4233. [PMID: 31645420 PMCID: PMC6893195 DOI: 10.1534/g3.119.400740] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genetic mapping is a primary tool of genetics in model organisms; however, many quantitative trait loci (QTL) contain tens or hundreds of positional candidate genes. Prioritizing these genes for validation is often ad hoc and biased by previous findings. Here we present a technique for prioritizing positional candidates based on computationally inferred gene function. Our method uses machine learning with functional genomic networks, whose links encode functional associations among genes, to identify network-based signatures of functional association to a trait of interest. We demonstrate the method by functionally ranking positional candidates in a large locus on mouse Chr 6 (45.9 Mb to 127.8 Mb) associated with histamine hypersensitivity (Histh). Histh is characterized by systemic vascular leakage and edema in response to histamine challenge, which can lead to multiple organ failure and death. Although Histh risk is strongly influenced by genetics, little is known about its underlying molecular or genetic causes, due to genetic and physiological complexity of the trait. To dissect this complexity, we ranked genes in the Histh locus by predicting functional association with multiple Histh-related processes. We integrated these predictions with new single nucleotide polymorphism (SNP) association data derived from a survey of 23 inbred mouse strains and congenic mapping data. The top-ranked genes included Cxcl12, Ret, Cacna1c, and Cntn3, all of which had strong functional associations and were proximal to SNPs segregating with Histh. These results demonstrate the power of network-based computational methods to nominate highly plausible quantitative trait genes even in challenging cases involving large QTL and extreme trait complexity.
Collapse
|
41
|
Kolishovski G, Lamoureux A, Hale P, Richardson JE, Recla JM, Adesanya O, Simons A, Kunde-Ramamoorthy G, Bult CJ. The JAX Synteny Browser for mouse-human comparative genomics. Mamm Genome 2019; 30:353-361. [PMID: 31776723 PMCID: PMC6892358 DOI: 10.1007/s00335-019-09821-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/20/2019] [Indexed: 10/30/2022]
Abstract
Visualizing regions of conserved synteny between two genomes is supported by numerous software applications. However, none of the current applications allow researchers to select genome features to display or highlight in blocks of synteny based on the annotated biological properties of the features (e.g., type, function, and/or phenotype association). To address this usability gap, we developed an interactive web-based conserved synteny browser, The Jackson Laboratory (JAX) Synteny Browser. The browser allows researchers to highlight or selectively display genome features in the reference and/or the comparison genome according to the biological attributes of the features. Although the current implementation for the browser is limited to the reference genomes for the laboratory mouse and human, the software platform is intentionally genome agnostic. The JAX Synteny Browser software can be deployed for any two genomes where genome coordinates for syntenic blocks are defined and for which biological attributes of the features in one or both genomes are available in widely used standard bioinformatics file formats. The JAX Synteny Browser is available at: http://syntenybrowser.jax.org/. The code base is available from GitHub: https://github.com/TheJacksonLaboratory/syntenybrowser and is distributed under the Creative Commons Attribution license (CC BY).
Collapse
Affiliation(s)
- Georgi Kolishovski
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
| | - Anna Lamoureux
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
| | - Paul Hale
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
| | - Joel E Richardson
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
| | - Jill M Recla
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
| | - Omoluyi Adesanya
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
- Institute of Public Health, Washington University of St. Louis, St. Louis, MO, 63110, USA
| | - Al Simons
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA
| | | | - Carol J Bult
- The Jackson Laboratory for Mammalian Genomics, Bar Harbor, ME, 04609, USA.
| |
Collapse
|
42
|
Zhang W, Zhang H, Yang H, Li M, Xie Z, Li W. Computational resources associating diseases with genotypes, phenotypes and exposures. Brief Bioinform 2019; 20:2098-2115. [PMID: 30102366 PMCID: PMC6954426 DOI: 10.1093/bib/bby071] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/01/2018] [Indexed: 12/16/2022] Open
Abstract
The causes of a disease and its therapies are not only related to genotypes, but also associated with other factors, including phenotypes, environmental exposures, drugs and chemical molecules. Distinguishing disease-related factors from many neutral factors is critical as well as difficult. Over the past two decades, bioinformaticians have developed many computational resources to integrate the omics data and discover associations among these factors. However, researchers and clinicians are experiencing difficulties in choosing appropriate resources from hundreds of relevant databases and software tools. Here, in order to assist the researchers and clinicians, we systematically review the public computational resources of human diseases related to genotypes, phenotypes, environment factors, drugs and chemical exposures. We briefly describe the development history of these computational resources, followed by the details of the relevant databases and software tools. We finally conclude with a discussion of current challenges and future opportunities as well as prospects on this topic.
Collapse
Affiliation(s)
- Wenliang Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Haiyue Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Huan Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Miaoxin Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhi Xie
- State Key Lab of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 500040, China
| | - Weizhong Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
43
|
Paquola C, Bethlehem RAI, Seidlitz J, Wagstyl K, Romero-Garcia R, Whitaker KJ, Vos de Wael R, Williams GB, NSPN Consortium, Vértes PE, Margulies DS, Bernhardt B, Bullmore ET. Shifts in myeloarchitecture characterise adolescent development of cortical gradients. eLife 2019; 8:e50482. [PMID: 31724948 PMCID: PMC6855802 DOI: 10.7554/elife.50482] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/02/2019] [Indexed: 11/21/2022] Open
Abstract
We studied an accelerated longitudinal cohort of adolescents and young adults (n = 234, two time points) to investigate dynamic reconfigurations in myeloarchitecture. Intracortical profiles were generated using magnetization transfer (MT) data, a myelin-sensitive magnetic resonance imaging contrast. Mixed-effect models of depth specific intracortical profiles demonstrated two separate processes i) overall increases in MT, and ii) flattening of the MT profile related to enhanced signal in mid-to-deeper layers, especially in heteromodal and unimodal association cortices. This development was independent of morphological changes. Enhanced MT in mid-to-deeper layers was found to spatially co-localise specifically with gene expression markers of oligodendrocytes. Interregional covariance analysis revealed that these intracortical changes contributed to a gradual differentiation of higher-order from lower-order systems. Depth-dependent trajectories of intracortical myeloarchitectural development contribute to the maturation of structural hierarchies in the human neocortex, providing a model for adolescent development that bridges microstructural and macroscopic scales of brain organisation.
Collapse
Affiliation(s)
- Casey Paquola
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMcGill UniversityMontrealCanada
| | - Richard AI Bethlehem
- Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
- Autism Research Centre, Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
| | - Jakob Seidlitz
- Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
- Developmental Neurogenomics UnitNational Institute of Mental HealthBethesdaUnited States
| | - Konrad Wagstyl
- Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
| | | | - Kirstie J Whitaker
- Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
- The Alan Turing InstituteLondonUnited Kingdom
| | - Reinder Vos de Wael
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMcGill UniversityMontrealCanada
| | - Guy B Williams
- Department of Clinical Neurosciences, Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUnited Kingdom
| | | | - Petra E Vértes
- Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
- The Alan Turing InstituteLondonUnited Kingdom
| | - Daniel S Margulies
- FrontlabInstitut du Cerveau et de la Moelle épinière, UPMC UMRS 1127, Inserm U 1127, CNRS UMR 7225ParisFrance
| | - Boris Bernhardt
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and HospitalMcGill UniversityMontrealCanada
| | - Edward T Bullmore
- Department of PsychiatryUniversity of CambridgeCambridgeUnited Kingdom
- Department of Clinical Neurosciences, Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
44
|
Karunakaran KB, Chaparala S, Ganapathiraju MK. Potentially repurposable drugs for schizophrenia identified from its interactome. Sci Rep 2019; 9:12682. [PMID: 31481665 PMCID: PMC6722087 DOI: 10.1038/s41598-019-48307-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
We previously presented the protein-protein interaction network of schizophrenia associated genes, and from it, the drug-protein interactome which showed the drugs that target any of the proteins in the interactome. Here, we studied these drugs further to identify whether any of them may potentially be repurposable for schizophrenia. In schizophrenia, gene expression has been described as a measurable aspect of the disease reflecting the action of risk genes. We studied each of the drugs from the interactome using the BaseSpace Correlation Engine, and shortlisted those that had a negative correlation with differential gene expression of schizophrenia. This analysis resulted in 12 drugs whose differential gene expression (drug versus normal) had an anti-correlation with differential expression for schizophrenia (disorder versus normal). Some of these drugs were already being tested for their clinical activity in schizophrenia and other neuropsychiatric disorders. Several proteins in the protein interactome of the targets of several of these drugs were associated with various neuropsychiatric disorders. The network of genes with opposite drug-induced versus schizophrenia-associated expression profiles were significantly enriched in pathways relevant to schizophrenia etiology and GWAS genes associated with traits or diseases that had a pathophysiological overlap with schizophrenia. Drugs that targeted the same genes as the shortlisted drugs, have also demonstrated clinical activity in schizophrenia and other related disorders. This integrated computational analysis will help translate insights from the schizophrenia drug-protein interactome to clinical research - an important step, especially in the field of psychiatric drug development which faces a high failure rate.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Indian Institute of Science, Bengaluru, India
| | | | - Madhavi K Ganapathiraju
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, USA.
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
45
|
Bogue MA, Grubb SC, Walton DO, Philip VM, Kolishovski G, Stearns T, Dunn MH, Skelly DA, Kadakkuzha B, TeHennepe G, Kunde-Ramamoorthy G, Chesler EJ. Mouse Phenome Database: an integrative database and analysis suite for curated empirical phenotype data from laboratory mice. Nucleic Acids Res 2019; 46:D843-D850. [PMID: 29136208 PMCID: PMC5753241 DOI: 10.1093/nar/gkx1082] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/19/2017] [Indexed: 12/25/2022] Open
Abstract
The Mouse Phenome Database (MPD; https://phenome.jax.org) is a widely used resource that provides access to primary experimental trait data, genotypic variation, protocols and analysis tools for mouse genetic studies. Data are contributed by investigators worldwide and represent a broad scope of phenotyping endpoints and disease-related traits in naïve mice and those exposed to drugs, environmental agents or other treatments. MPD houses individual animal data with detailed, searchable protocols, and makes these data available to other resources via API. MPD provides rigorous curation of experimental data and supporting documentation using relevant ontologies and controlled vocabularies. Most data in MPD are from inbreds and other reproducible strains such that the data are cumulative over time and across laboratories. The resource has been expanded to include the QTL Archive and other primary phenotype data from mapping crosses as well as advanced high-diversity mouse populations including the Collaborative Cross and Diversity Outbred mice. Furthermore, MPD provides a means of assessing replicability and reproducibility across experimental conditions and protocols, benchmarking assays in users’ own laboratories, identifying sensitized backgrounds for making new mouse models with genome editing technologies, analyzing trait co-inheritance, finding the common genetic basis for multiple traits and assessing sex differences and sex-by-genotype interactions.
Collapse
Affiliation(s)
- Molly A Bogue
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | | | | | - Tim Stearns
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Smith CL, Blake JA, Kadin JA, Richardson JE, Bult CJ. Mouse Genome Database (MGD)-2018: knowledgebase for the laboratory mouse. Nucleic Acids Res 2019; 46:D836-D842. [PMID: 29092072 PMCID: PMC5753350 DOI: 10.1093/nar/gkx1006] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/19/2017] [Indexed: 12/23/2022] Open
Abstract
The Mouse Genome Database (MGD; http://www.informatics.jax.org) is the key community mouse database which supports basic, translational and computational research by providing integrated data on the genetics, genomics, and biology of the laboratory mouse. MGD serves as the source for biological reference data sets related to mouse genes, gene functions, phenotypes and disease models with an increasing emphasis on the association of these data to human biology and disease. We report here on recent enhancements to this resource, including improved access to mouse disease model and human phenotype data and enhanced relationships of mouse models to human disease.
Collapse
Affiliation(s)
- Cynthia L Smith
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Judith A Blake
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - James A Kadin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Carol J Bult
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
47
|
Jia J, An Z, Ming Y, Guo Y, Li W, Liang Y, Guo D, Li X, Tai J, Chen G, Jin Y, Liu Z, Ni X, Shi T. eRAM: encyclopedia of rare disease annotations for precision medicine. Nucleic Acids Res 2019; 46:D937-D943. [PMID: 29106618 PMCID: PMC5753383 DOI: 10.1093/nar/gkx1062] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/24/2017] [Indexed: 01/12/2023] Open
Abstract
Rare diseases affect over a hundred million people worldwide, most of these patients are not accurately diagnosed and effectively treated. The limited knowledge of rare diseases forms the biggest obstacle for improving their treatment. Detailed clinical phenotyping is considered as a keystone of deciphering genes and realizing the precision medicine for rare diseases. Here, we preset a standardized system for various types of rare diseases, called encyclopedia of Rare disease Annotations for Precision Medicine (eRAM). eRAM was built by text-mining nearly 10 million scientific publications and electronic medical records, and integrating various data in existing recognized databases (such as Unified Medical Language System (UMLS), Human Phenotype Ontology, Orphanet, OMIM, GWAS). eRAM systematically incorporates currently available data on clinical manifestations and molecular mechanisms of rare diseases and uncovers many novel associations among diseases. eRAM provides enriched annotations for 15 942 rare diseases, yielding 6147 human disease related phenotype terms, 31 661 mammalians phenotype terms, 10,202 symptoms from UMLS, 18 815 genes and 92 580 genotypes. eRAM can not only provide information about rare disease mechanism but also facilitate clinicians to make accurate diagnostic and therapeutic decisions towards rare diseases. eRAM can be freely accessed at http://www.unimd.org/eram/.
Collapse
Affiliation(s)
- Jinmeng Jia
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhongxin An
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yue Ming
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, the Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, The Ministry of Education Key Laboratory of Major Diseases in Children, Center for Medical Genetics, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yunxiang Liang
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dongming Guo
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xin Li
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jun Tai
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, the Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Geng Chen
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, the Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Zhimei Liu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, the Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, the Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Tieliu Shi
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
48
|
Recla JM, Bubier JA, Gatti DM, Ryan JL, Long KH, Robledo RF, Glidden NC, Hou G, Churchill GA, Maser RS, Zhang ZW, Young EE, Chesler EJ, Bult CJ. Genetic mapping in Diversity Outbred mice identifies a Trpa1 variant influencing late-phase formalin response. Pain 2019; 160:1740-1753. [PMID: 31335644 PMCID: PMC6668363 DOI: 10.1097/j.pain.0000000000001571] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Identification of genetic variants that influence susceptibility to pain is key to identifying molecular mechanisms and targets for effective and safe therapeutic alternatives to opioids. To identify genes and variants associated with persistent pain, we measured late-phase response to formalin injection in 275 male and female Diversity Outbred mice genotyped for over 70,000 single nucleotide polymorphisms. One quantitative trait locus reached genome-wide significance on chromosome 1 with a support interval of 3.1 Mb. This locus, Nociq4 (nociceptive sensitivity quantitative trait locus 4; MGI: 5661503), harbors the well-known pain gene Trpa1 (transient receptor potential cation channel, subfamily A, member 1). Trpa1 is a cation channel known to play an important role in acute and chronic pain in both humans and mice. Analysis of Diversity Outbred founder strain allele effects revealed a significant effect of the CAST/EiJ allele at Trpa1, with CAST/EiJ carrier mice showing an early, but not late, response to formalin relative to carriers of the 7 other inbred founder alleles (A/J, C57BL/6J, 129S1/SvImJ, NOD/ShiLtJ, NZO/HlLtJ, PWK/PhJ, and WSB/EiJ). We characterized possible functional consequences of sequence variants in Trpa1 by assessing channel conductance, TRPA1-TRPV1 interactions, and isoform expression. The phenotypic differences observed in CAST/EiJ relative to C57BL/6J carriers were best explained by Trpa1 isoform expression differences, implicating a splice junction variant as the causal functional variant. This study demonstrates the utility of advanced, high-precision genetic mapping populations in resolving specific molecular mechanisms of variation in pain sensitivity.
Collapse
Affiliation(s)
- Jill M. Recla
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
- IGERT Program in Functional Genomics, Graduate School of Biomedical Sciences and Engineering, The University of Maine, Orono, ME 04469, USA
| | - Jason A. Bubier
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Daniel M. Gatti
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Jennifer L. Ryan
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Katie H. Long
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Nicole C. Glidden
- Department of Genetics and Genome Sciences, UCONN Health, 400 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Guoqiang Hou
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | - Richard S. Maser
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Zhong-wei Zhang
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Erin E. Young
- Department of Genetics and Genome Sciences, UCONN Health, 400 Farmington Avenue, Farmington, CT 06030-6403, USA
- School of Nursing, University of Connecticut, 231 Glenbrook Rd, Unit 4026, Storrs, CT 06269-4026, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269-4026, USA
| | | | - Carol J. Bult
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| |
Collapse
|
49
|
Bubier J, Hill D, Mukherjee G, Reynolds T, Baker EJ, Berger A, Emerson J, Blake JA, Chesler EJ. Curating gene sets: challenges and opportunities for integrative analysis. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2019; 2019:5381578. [PMID: 30888410 PMCID: PMC6424415 DOI: 10.1093/database/baz036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/20/2022]
Abstract
Genomic data interpretation often requires analyses that move from a gene-by-gene focus to a focus on sets of genes that are associated with biological phenomena such as molecular processes, phenotypes, diseases, drug interactions or environmental conditions. Unique challenges exist in the curation of gene sets beyond the challenges in curation of individual genes. Here we highlight a literature curation workflow whereby gene sets are curated from peer-reviewed published data into GeneWeaver (GW), a data repository and analysis platform. We describe the system features that allow for a flexible yet precise curation procedure. We illustrate the value of curation by gene sets through analysis of independently curated sets that relate to the integrated stress response, showing that sets curated from independent sources all share significant Jaccard similarity. A suite of reproducible analysis tools is provided in GW as services to carry out interactive functional investigation of user-submitted gene sets within the context of over 150 000 gene sets constructed from publicly available resources and published gene lists. A curation interface supports the ability of users to design and maintain curation workflows of gene sets, including assigning, reviewing and releasing gene sets within a curation project context.
Collapse
Affiliation(s)
| | - David Hill
- The Jackson Laboratory, Bar Harbor, ME, USA
| | | | - Timothy Reynolds
- Institute of Biomedical Studies, Baylor University, Waco, TX, USA
| | - Erich J Baker
- Institute of Biomedical Studies, Baylor University, Waco, TX, USA
| | | | | | | | | |
Collapse
|
50
|
Zhao Y, Smith JR, Wang SJ, Dwinell MR, Shimoyama M. Quantitative phenotype analysis to identify, validate and compare rat disease models. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2019; 2019:5424140. [PMID: 30938777 PMCID: PMC6444380 DOI: 10.1093/database/baz037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/08/2019] [Accepted: 02/27/2019] [Indexed: 12/18/2022]
Abstract
The laboratory rat has been widely used as an animal model in biomedical research. There are many strains exhibiting a wide variety of phenotypes. Capturing these phenotypes in a centralized database provides researchers with an easy method for choosing the appropriate strains for their studies. Existing resources have provided some preliminary work in rat phenotype databases. However, existing resources suffer from problems such as small number of animals, lack of updating, web interface queries limitations and lack of standardized metadata. The Rat Genome Database (RGD) PhenoMiner tool has provided the first step in this effort by standardizing and integrating data from individual studies. Our work, mainly utilizing data curated in RGD, involves the following key steps: (i) we developed a meta-analysis pipeline to automatically integrate data from heterogeneous sources and to produce expected ranges (standardized phenotype ranges) for different strains and phenotypes under different experimental conditions; (ii) we created tools to visualize expected ranges for individual strains and strain groups. We developed a meta-analysis pipeline and an interactive web interface that summarizes and visualizes expected ranges produced from the meta-analysis pipeline. Automation of the pipeline allows for updates as additional data becomes available. The interactive web interface provides curators and researchers with a platform for identifying and validating expected ranges for a variety of quantitative phenotypes. The data analysis result and visualization tools will promote an understanding of rat disease models, guide researchers to choose optimal strains for their research needs and encourage data sharing from different research hubs. Such resources also help to promote research reproducibility. The interactive platforms created in this project will continue to provide a valuable resource for translational research efforts.
Collapse
Affiliation(s)
- Yiqing Zhao
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, USA.,Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Jennifer R Smith
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shur-Jen Wang
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Melinda R Dwinell
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mary Shimoyama
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|