1
|
DiLillo KM, Ruvuna L, Pratte KA, Serban KA, Labaki WW, Han MK, Freeman CM, Bowler RP, Curtis JL, Arnold KB. Validation of Systemic Complement Signatures in the Progression of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2024; 210:1269-1272. [PMID: 39311978 PMCID: PMC11568447 DOI: 10.1164/rccm.202311-2059le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 09/18/2024] [Indexed: 11/11/2024] Open
Affiliation(s)
| | | | | | - Karina A. Serban
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida; and
| | | | - MeiLan K. Han
- Division of Pulmonary and Critical Care Medicine, and
| | - Christine M. Freeman
- Division of Pulmonary and Critical Care Medicine, and
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan
- Research Service and
| | | | - Jeffrey L. Curtis
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | | |
Collapse
|
2
|
Chen BD, Lee C, Tapia AL, Reiner AP, Tang H, Kooperberg C, Manson JE, Li Y, Raffield LM. Proteome-wide association study using cis and trans variants and applied to blood cell and lipid-related traits in the Women's Health Initiative study. Genet Epidemiol 2024; 48:310-323. [PMID: 38940271 DOI: 10.1002/gepi.22578] [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: 09/11/2023] [Revised: 05/26/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
In most Proteome-Wide Association Studies (PWAS), variants near the protein-coding gene (±1 Mb), also known as cis single nucleotide polymorphisms (SNPs), are used to predict protein levels, which are then tested for association with phenotypes. However, proteins can be regulated through variants outside of the cis region. An intermediate GWAS step to identify protein quantitative trait loci (pQTL) allows for the inclusion of trans SNPs outside the cis region in protein-level prediction models. Here, we assess the prediction of 540 proteins in 1002 individuals from the Women's Health Initiative (WHI), split equally into a GWAS set, an elastic net training set, and a testing set. We compared the testing r2 between measured and predicted protein levels using this proposed approach, to the testing r2 using only cis SNPs. The two methods usually resulted in similar testing r2, but some proteins showed a significant increase in testing r2 with our method. For example, for cartilage acidic protein 1, the testing r2 increased from 0.101 to 0.351. We also demonstrate reproducible findings for predicted protein association with lipid and blood cell traits in WHI participants without proteomics data and in UK Biobank utilizing our PWAS weights.
Collapse
Affiliation(s)
- Brian D Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Chanhwa Lee
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Amanda L Tapia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Hua Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Yang H, Hou C, Chen W, Zeng Y, Qu Y, Sun Y, Hu Y, Tang X, Song H. Disease Modules Associated with Unfavorable Sleep Patterns and Their Genetic Determinants: A Prospective Cohort Study of the UK Biobank. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:415-429. [PMID: 39723226 PMCID: PMC11666895 DOI: 10.1007/s43657-023-00144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 12/28/2024]
Abstract
Despite the established associations between sleep-related traits and major diseases, comprehensive assessment on affected disease modules and their genetic determinants is lacking. Using multiple correspondence analysis and the k-means clustering algorithm, 235,826 eligible participants were clustered into distinct unfavorable sleep patterns [short sleep duration (n = 10,073), snoring (22,419), insomnia (102,771), insomnia and snoring (62,909)] and favorable sleep pattern groups (37,654). The associations of unfavorable sleep patterns with 134 diseases were estimated using Cox regression models; and comorbidity network analyses were applied for disease module identification. Genetic determinants associated with each disease module were identified by genome-wide association studies (GWAS). During an average follow-up of 10.80 years, unfavorable sleep patterns featured by 'short sleep duration', 'snoring', 'insomnia', and 'insomnia and snoring' were associated with increased risk of 0, 9, 10, and 19 diseases, respectively. Furthermore, comorbidity network analyses categorized these affected diseases into three disease modules, characterized by predominant diseases related to digestive system, circulatory and endocrine systems (snoring-related patterns only), and musculoskeletal system (insomnia-related patterns only). Using the number of affected diseases, as an index of a person's susceptibility to each disease module [i.e., susceptible score (SS)], GWAS analyses identified five, one, and three significant loci associated with the residual SS of these aforementioned disease modules, respectively, which mapped to several potential biological pathways, including those related to hormone regulation and catecholamine uptake. In conclusion, individuals with unfavorable sleep patterns, particularly snoring and insomnia, had increased risk of multiple diseases. The identification of three major disease modules with their relevant genetic determinants may facilitate strategy development for precision prevention of future health decline. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-023-00144-8.
Collapse
Affiliation(s)
- Huazhen Yang
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Can Hou
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Wenwen Chen
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yu Zeng
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yuanyuan Qu
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yajing Sun
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yao Hu
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610000 China
| | - Huan Song
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavík, Iceland
| |
Collapse
|
4
|
Konigsberg IR, Vu T, Liu W, Litkowski EM, Pratte KA, Vargas LB, Gilmore N, Abdel-Hafiz M, Manichaikul A, Cho MH, Hersh CP, DeMeo DL, Banaei-Kashani F, Bowler RP, Lange LA, Kechris KJ. Proteomic networks and related genetic variants associated with smoking and chronic obstructive pulmonary disease. BMC Genomics 2024; 25:825. [PMID: 39223457 PMCID: PMC11370252 DOI: 10.1186/s12864-024-10619-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Studies have identified individual blood biomarkers associated with chronic obstructive pulmonary disease (COPD) and related phenotypes. However, complex diseases such as COPD typically involve changes in multiple molecules with interconnections that may not be captured when considering single molecular features. METHODS Leveraging proteomic data from 3,173 COPDGene Non-Hispanic White (NHW) and African American (AA) participants, we applied sparse multiple canonical correlation network analysis (SmCCNet) to 4,776 proteins assayed on the SomaScan v4.0 platform to derive sparse networks of proteins associated with current vs. former smoking status, airflow obstruction, and emphysema quantitated from high-resolution computed tomography scans. We then used NetSHy, a dimension reduction technique leveraging network topology, to produce summary scores of each proteomic network, referred to as NetSHy scores. We next performed a genome-wide association study (GWAS) to identify variants associated with the NetSHy scores, or network quantitative trait loci (nQTLs). Finally, we evaluated the replicability of the networks in an independent cohort, SPIROMICS. RESULTS We identified networks of 13 to 104 proteins for each phenotype and exposure in NHW and AA, and the derived NetSHy scores significantly associated with the variable of interests. Networks included known (sRAGE, ALPP, MIP1) and novel molecules (CA10, CPB1, HIS3, PXDN) and interactions involved in COPD pathogenesis. We observed 7 nQTL loci associated with NetSHy scores, 4 of which remained after conditional analysis. Networks for smoking status and emphysema, but not airflow obstruction, demonstrated a high degree of replicability across race groups and cohorts. CONCLUSIONS In this work, we apply state-of-the-art molecular network generation and summarization approaches to proteomic data from COPDGene participants to uncover protein networks associated with COPD phenotypes. We further identify genetic associations with networks. This work discovers protein networks containing known and novel proteins and protein interactions associated with clinically relevant COPD phenotypes across race groups and cohorts.
Collapse
Affiliation(s)
- Iain R Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Thao Vu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Weixuan Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Elizabeth M Litkowski
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Luciana B Vargas
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Niles Gilmore
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Mohamed Abdel-Hafiz
- Department of Computer Science and Engineering, University of Colorado - Denver, Denver, CO, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Farnoush Banaei-Kashani
- Department of Computer Science and Engineering, University of Colorado - Denver, Denver, CO, USA
| | | | - Leslie A Lange
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Katerina J Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA.
| |
Collapse
|
5
|
Wen J, Liu J, Feng Q, Lu Y, Yuan K, Zhang X, Zhang C, Gao Y, Wang X, Mamatyusupu D, Xu S. Ancestral origins and post-admixture adaptive evolution of highland Tajiks. Natl Sci Rev 2024; 11:nwae284. [PMID: 40040643 PMCID: PMC11879426 DOI: 10.1093/nsr/nwae284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/04/2024] [Accepted: 08/04/2024] [Indexed: 03/06/2025] Open
Abstract
It remains debatable how many genes and how various the mechanisms are behind human adaptation to extreme environments, such as high altitudes. Despite extensive studies on Tibetans, Andeans and Ethiopians, new insights are expected to be provided with careful analysis of underrepresented highlanders living in a different geographical region, such as the Tajiks, who reside on the Pamir Plateau at an average altitude exceeding 4000 meters. Moreover, genetic admixture, as we observed in the current whole-genome deep-sequencing study of Xinjiang Tajiks (XJT), offers a unique opportunity to explore how admixture may facilitate adaptation to high-altitude environments. Compared with other extensively studied highlanders, XJT showed pronounced admixture patterns: most of their ancestry are derived from West Eurasians (34.5%-48.3%) and South Asians (21.4%-40.0%), and some minor ancestry from East Asians and Siberians (3.62%-17.5%). The greater genetic diversity in XJT than in their ancestral source populations provides a genetic basis for their adaptation to high-altitude environments. The admixture gain of functional adaptive components from ancestral populations could facilitate adaptation to high-altitude environments. Specifically, admixture-facilitated adaptation was strongly associated with skin-related candidate genes that respond to UV radiation (e.g. HERC2 and BNC2) and cardiovascular-system-related genes (e.g. MPI and BEST1). Notably, no adaptive variants of genes showing outstanding natural selection signatures in the Tibetan or Andean highlanders were identified in XJT, including EPAS1 and EGLN1, indicating that a different set of genes contributed to XJT's survival on the Pamir Plateau, although some genes underlying natural selection in XJT have been previously reported in other highlanders. Our results highlight the unique genetic adaptations in XJT and propose that admixture may play a vital role in facilitating high-altitude adaptation. By introducing and elevating diversity, admixture likely induces novel genetic factors that contribute to the survival of populations in extreme environments like the highlands.
Collapse
Affiliation(s)
- Jia Wen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiaojiao Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qidi Feng
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Lu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200438, China
| | - Kai Yuan
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoxi Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Zhang
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yang Gao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoji Wang
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dolikun Mamatyusupu
- College of the Life Sciences and Technology, Xinjiang University, Urumqi 830046, China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
6
|
Jiang MZ, Gaynor SM, Li X, Van Buren E, Stilp A, Buth E, Wang FF, Manansala R, Gogarten SM, Li Z, Polfus LM, Salimi S, Bis JC, Pankratz N, Yanek LR, Durda P, Tracy RP, Rich SS, Rotter JI, Mitchell BD, Lewis JP, Psaty BM, Pratte KA, Silverman EK, Kaplan RC, Avery C, North KE, Mathias RA, Faraday N, Lin H, Wang B, Carson AP, Norwood AF, Gibbs RA, Kooperberg C, Lundin J, Peters U, Dupuis J, Hou L, Fornage M, Benjamin EJ, Reiner AP, Bowler RP, Lin X, Auer PL, Raffield LM, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, TOPMed Inflammation Working Group. Whole genome sequencing based analysis of inflammation biomarkers in the Trans-Omics for Precision Medicine (TOPMed) consortium. Hum Mol Genet 2024; 33:1429-1441. [PMID: 38747556 PMCID: PMC11305684 DOI: 10.1093/hmg/ddae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 05/28/2024] Open
Abstract
Inflammation biomarkers can provide valuable insight into the role of inflammatory processes in many diseases and conditions. Sequencing based analyses of such biomarkers can also serve as an exemplar of the genetic architecture of quantitative traits. To evaluate the biological insight, which can be provided by a multi-ancestry, whole-genome based association study, we performed a comprehensive analysis of 21 inflammation biomarkers from up to 38 465 individuals with whole-genome sequencing from the Trans-Omics for Precision Medicine (TOPMed) program (with varying sample size by trait, where the minimum sample size was n = 737 for MMP-1). We identified 22 distinct single-variant associations across 6 traits-E-selectin, intercellular adhesion molecule 1, interleukin-6, lipoprotein-associated phospholipase A2 activity and mass, and P-selectin-that remained significant after conditioning on previously identified associations for these inflammatory biomarkers. We further expanded upon known biomarker associations by pairing the single-variant analysis with a rare variant set-based analysis that further identified 19 significant rare variant set-based associations with 5 traits. These signals were distinct from both significant single variant association signals within TOPMed and genetic signals observed in prior studies, demonstrating the complementary value of performing both single and rare variant analyses when analyzing quantitative traits. We also confirm several previously reported signals from semi-quantitative proteomics platforms. Many of these signals demonstrate the extensive allelic heterogeneity and ancestry-differentiated variant-trait associations common for inflammation biomarkers, a characteristic we hypothesize will be increasingly observed with well-powered, large-scale analyses of complex traits.
Collapse
Affiliation(s)
- Min-Zhi Jiang
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
| | - Sheila M Gaynor
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, United States
- Regeneron Genetics Center, 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Xihao Li
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
- Department of Biostatistics, 135 Dauer Drive, 4115D McGavran-Greenberg Hall, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Eric Van Buren
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, United States
| | - Adrienne Stilp
- Department of Biostatistics, 4333 Brooklyn Ave NE, University of Washington, Seattle, WA 98105, United States
| | - Erin Buth
- Department of Biostatistics, 4333 Brooklyn Ave NE, University of Washington, Seattle, WA 98105, United States
| | - Fei Fei Wang
- Department of Biostatistics, 4333 Brooklyn Ave NE, University of Washington, Seattle, WA 98105, United States
| | - Regina Manansala
- Centre for Health Economics Research & Modelling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO) WHO Collaborating Centre, University of Antwerp, Campus Drie Eiken - Building S; Universiteitsplein 1 2610 Antwerpen, Belgium
| | - Stephanie M Gogarten
- Department of Biostatistics, 4333 Brooklyn Ave NE, University of Washington, Seattle, WA 98105, United States
| | - Zilin Li
- School of Mathematics and Statistics, Northeast Normal University, 5268 Renmin Street, Changchun, JL 130024, China
| | - Linda M Polfus
- Advanced Analytics, Ambry Genetics, 1 Enterprise, Aliso Viejo, CA 92656, United States
| | - Shabnam Salimi
- Department of Epidemiology and Public Health, Division of Gerontology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave NE, Box 359458, Seattle, WA 98195, United States
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN 55455, United States
| | - Lisa R Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, 1830 E Monument St Rm 8024, Baltimore, MD 21287, United States
| | - Peter Durda
- Department of Pathology & Laboratory Medicine, University of Vermont Larner College of Medicine, 360 South Park Drive, Colchester, VT 05446, United States
| | - Russell P Tracy
- Department of Pathology & Laboratory Medicine, University of Vermont Larner College of Medicine, 360 South Park Drive, Colchester, VT 05446, United States
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, 200 Jeanette Lancaster Way, Charlottesville, VA 22903, United States
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA 90502, United States
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, 670 W. Baltimore St., Baltimore, MD 21201, United States
| | - Joshua P Lewis
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, 670 W. Baltimore St., Baltimore, MD 21201, United States
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave NE, Box 359458, Seattle, WA 98195, United States
- Departments of Epidemiology and Health Systems and Population Health, University of Washington, 4333 Brooklyn Ave NE, Seattle, WA 98101, United States
| | - Katherine A Pratte
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, United States
| | - Edwin K Silverman
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | - Christy Avery
- Department of Epidemiology, University of North Carolina at Chapel Hill, 137 East Franklin Street, Chapel Hill, NC 27599, United States
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, 137 East Franklin Street, Chapel Hill, NC 27599, United States
| | - Rasika A Mathias
- Department of Medicine, Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Cir JHAAC Room 3B53, Baltimore, MD 21287, United States
| | - Nauder Faraday
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD 21287, United States
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, 55 Lake Ave North, Worcester, MA 01655, United States
| | - Biqi Wang
- Department of Medicine, University of Massachusetts Chan Medical School, 55 Lake Ave North, Worcester, MA 01655, United States
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, 350 W. Woodrow Wilson Avenue, Suite 701, Jackson, MS 39213, United States
| | - Arnita F Norwood
- Department of Medicine, University of Mississippi Medical Center, 350 W. Woodrow Wilson Avenue, Suite 701, Jackson, MS 39213, United States
| | - Richard A Gibbs
- Department of Molecular and Human Genetics, Human Genome Sequencing Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA 98109, United States
| | - Jessica Lundin
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA 98109, United States
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA 98109, United States
| | - Josée Dupuis
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, 2001 McGill College Avenue, Montreal, QC H3A 1G1, Canada
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA 02118, United States
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N Lake Shore Drive, Chicago, IL 60611, United States
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, United States
| | - Emelia J Benjamin
- Department of Medicine, Cardiovascular Medicine, Boston Medical Center, Boston University Chobanian and Avedisian School of Medicine, 72 East Newton Street, Boston, MA 02118, United States
- Department of Epidemiology, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA 02118, United States
- Boston University and National Heart, Lung, and Blood Institute’s Framingham Heart Study, 73 Mount Wayte Ave #2, Framingham, MA 01702, United States
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, 4333 Brooklyn Ave NE, Seattle, WA 98105, United States
| | - Russell P Bowler
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, United States
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, United States
| | - Paul L Auer
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599, United States
| | | |
Collapse
|
7
|
Quintanilha JCF, Sibley AB, Liu Y, Niedzwiecki D, Halabi S, Rogers L, O'Neil B, Kindler H, Kelly W, Venook A, McLeod HL, Ratain MJ, Nixon AB, Innocenti F, Owzar K. Common variation in a long non-coding RNA gene modulates variation of circulating TGF-β2 levels in metastatic colorectal cancer patients (Alliance). BMC Genomics 2024; 25:473. [PMID: 38745123 PMCID: PMC11092225 DOI: 10.1186/s12864-024-10354-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Herein, we report results from a genome-wide study conducted to identify protein quantitative trait loci (pQTL) for circulating angiogenic and inflammatory protein markers in patients with metastatic colorectal cancer (mCRC). The study was conducted using genotype, protein marker, and baseline clinical and demographic data from CALGB/SWOG 80405 (Alliance), a randomized phase III study designed to assess outcomes of adding VEGF or EGFR inhibitors to systemic chemotherapy in mCRC patients. Germline DNA derived from blood was genotyped on whole-genome array platforms. The abundance of protein markers was quantified using a multiplex enzyme-linked immunosorbent assay from plasma derived from peripheral venous blood collected at baseline. A robust rank-based method was used to assess the statistical significance of each variant and protein pair against a strict genome-wide level. A given pQTL was tested for validation in two external datasets of prostate (CALGB 90401) and pancreatic cancer (CALGB 80303) patients. Bioinformatics analyses were conducted to further establish biological bases for these findings. RESULTS The final analysis was carried out based on data from 540,021 common typed genetic variants and 23 protein markers from 869 genetically estimated European patients with mCRC. Correcting for multiple testing, the analysis discovered a novel cis-pQTL in LINC02869, a long non-coding RNA gene, for circulating TGF-β2 levels (rs11118119; AAF = 0.11; P-value < 1.4e-14). This finding was validated in a cohort of 538 prostate cancer patients from CALGB 90401 (AAF = 0.10, P-value < 3.3e-25). The analysis also validated a cis-pQTL we had previously reported for VEGF-A in advanced pancreatic cancer, and additionally identified trans-pQTLs for VEGF-R3, and cis-pQTLs for CD73. CONCLUSIONS This study has provided evidence of a novel cis germline genetic variant that regulates circulating TGF-β2 levels in plasma of patients with advanced mCRC and prostate cancer. Moreover, the validation of previously identified pQTLs for VEGF-A, CD73, and VEGF-R3, potentiates the validity of these associations.
Collapse
Affiliation(s)
- Julia C F Quintanilha
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Donna Niedzwiecki
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Susan Halabi
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Layne Rogers
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Bert O'Neil
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Hedy Kindler
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - William Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Alan Venook
- Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Howard L McLeod
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Utah Tech University, St George, UT, USA
| | - Mark J Ratain
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA.
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Konigsberg IR, Vu T, Liu W, Litkowski EM, Pratte KA, Vargas LB, Gilmore N, Abdel-Hafiz M, Manichaikul AW, Cho MH, Hersh CP, DeMeo DL, Banaei-Kashani F, Bowler RP, Lange LA, Kechris KJ. Proteomic Networks and Related Genetic Variants Associated with Smoking and Chronic Obstructive Pulmonary Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.26.24303069. [PMID: 38464285 PMCID: PMC10925350 DOI: 10.1101/2024.02.26.24303069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Studies have identified individual blood biomarkers associated with chronic obstructive pulmonary disease (COPD) and related phenotypes. However, complex diseases such as COPD typically involve changes in multiple molecules with interconnections that may not be captured when considering single molecular features. Methods Leveraging proteomic data from 3,173 COPDGene Non-Hispanic White (NHW) and African American (AA) participants, we applied sparse multiple canonical correlation network analysis (SmCCNet) to 4,776 proteins assayed on the SomaScan v4.0 platform to derive sparse networks of proteins associated with current vs. former smoking status, airflow obstruction, and emphysema quantitated from high-resolution computed tomography scans. We then used NetSHy, a dimension reduction technique leveraging network topology, to produce summary scores of each proteomic network, referred to as NetSHy scores. We next performed genome-wide association study (GWAS) to identify variants associated with the NetSHy scores, or network quantitative trait loci (nQTLs). Finally, we evaluated the replicability of the networks in an independent cohort, SPIROMICS. Results We identified networks of 13 to 104 proteins for each phenotype and exposure in NHW and AA, and the derived NetSHy scores significantly associated with the variable of interests. Networks included known (sRAGE, ALPP, MIP1) and novel molecules (CA10, CPB1, HIS3, PXDN) and interactions involved in COPD pathogenesis. We observed 7 nQTL loci associated with NetSHy scores, 4 of which remained after conditional analysis. Networks for smoking status and emphysema, but not airflow obstruction, demonstrated a high degree of replicability across race groups and cohorts. Conclusions In this work, we apply state-of-the-art molecular network generation and summarization approaches to proteomic data from COPDGene participants to uncover protein networks associated with COPD phenotypes. We further identify genetic associations with networks. This work discovers protein networks containing known and novel proteins and protein interactions associated with clinically relevant COPD phenotypes across race groups and cohorts.
Collapse
Affiliation(s)
- Iain R Konigsberg
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Thao Vu
- Department of Biostatistics and Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Weixuan Liu
- Department of Biostatistics and Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Elizabeth M Litkowski
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
- Department of Medicine, University of Michigan, Ann Arbor, MI
| | | | - Luciana B Vargas
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Niles Gilmore
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Mohamed Abdel-Hafiz
- Department of Computer Science and Engineering, University of Colorado - Denver, Denver, CO
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Craig P Hersh
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dawn L DeMeo
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | - Leslie A Lange
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Katerina J Kechris
- Department of Biostatistics and Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
9
|
Quintanilha JC, Sibley AB, Liu Y, Niedzwiecki D, Halabi S, Rogers L, O’Neil B, Kindler H, Kelly W, Venook A, McLeod HL, Ratain MJ, Nixon AB, Innocenti F, Owzar K. Common variation in a long non-coding RNA gene modulates variation of circulating TGF- β2 levels in metastatic colorectal cancer patients (Alliance). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23298815. [PMID: 38106038 PMCID: PMC10723514 DOI: 10.1101/2023.12.04.23298815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Herein, we report results from a genome-wide study conducted to identify protein quantitative trait loci (pQTL) for circulating angiogenic and inflammatory protein markers in patients with metastatic colorectal cancer (mCRC).The study was conducted using genotype, protein marker, and baseline clinical and demographic data from CALGB/SWOG 80405 (Alliance), a randomized phase III study designed to assess outcomes of adding VEGF or EGFR inhibitors to systemic chemotherapy in mCRC patients. Germline DNA derived from blood was genotyped on whole-genome array platforms. The abundance of protein markers was quantified using a multiplex enzyme-linked immunosorbent assay from plasma derived from peripheral venous blood collected at baseline. A robust rank-based method was used to assess the statistical significance of each variant and protein pair against a strict genome-wide level. A given pQTL was tested for validation in two external datasets of prostate (CALGB 90401) and pancreatic cancer (CALGB 80303) patients. Bioinformatics analyses were conducted to further establish biological bases for these findings. Results The final analysis was carried out based on data from 540,021 common typed genetic variants and 23 protein markers from 869 genetically estimated European patients with mCRC. Correcting for multiple testing, the analysis discovered a novel cis-pQTL in LINC02869, a long non-coding RNA gene, for circulating TGF-β2 levels (rs11118119; AAF = 0.11; P-value < 1.4e-14). This finding was validated in a cohort of 538 prostate cancer patients from CALGB 90401 (AAF = 0.10, P-value < 3.3e-25). The analysis also validated a cis-pQTL we had previously reported for VEGF-A in advanced pancreatic cancer, and additionally identified trans-pQTLs for VEGF-R3, and cis-pQTLs for CD73. Conclusions This study has provided evidence of a novel cis germline genetic variant that regulates circulating TGF-β2 levels in plasma of patients with advanced mCRC and prostate cancer. Moreover, the validation of previously identified pQTLs for VEGF-A, CD73, and VEGF-R3, potentiates the validity of these associations.
Collapse
Affiliation(s)
- Julia C.F. Quintanilha
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexander B. Sibley
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Donna Niedzwiecki
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Susan Halabi
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Layne Rogers
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Bert O’Neil
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, USA
| | - Hedy Kindler
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - William Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alan Venook
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
| | - Howard L. McLeod
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; and Utah Tech University, St George, UT, USA (current); and Intermountain Healthcare, St George, UT, USA (current)
| | - Mark J. Ratain
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Andrew B. Nixon
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| |
Collapse
|
10
|
Lanktree MB, Perrot N, Smyth A, Chong M, Narula S, Shanmuganathan M, Kroezen Z, Britz-Mckibbin P, Berger M, Krepinsky JC, Pigeyre M, Yusuf S, Paré G. A novel multi-ancestry proteome-wide Mendelian randomization study implicates extracellular proteins, tubular cells, and fibroblasts in estimated glomerular filtration rate regulation. Kidney Int 2023; 104:1170-1184. [PMID: 37774922 DOI: 10.1016/j.kint.2023.08.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 08/15/2023] [Accepted: 08/25/2023] [Indexed: 10/01/2023]
Abstract
Estimated glomerular filtration rate (eGFR) impacts the concentration of plasma biomarkers confounding biomarker association studies of eGFR with reverse causation. To identify biomarkers causally associated with eGFR, we performed a proteome-wide Mendelian randomization study. Genetic variants nearby biomarker coding genes were tested for association with plasma concentration of 1,161 biomarkers in a multi-ancestry sample of 12,066 participants from the Prospective Urban and Rural Epidemiological (PURE) study. Using two-sample Mendelian randomization, individual variants' effects on biomarker concentration were correlated with their effects on eGFR and kidney traits from published genome-wide association studies (GWAS). Genetically altered concentrations of 22 biomarkers were associated with eGFR above a Bonferroni-corrected significance threshold. Five biomarkers were previously identified by GWAS (UMOD, FGF5, LGALS7, NINJ1, COL18A1). Nine biomarkers were within 1 Mb of the lead GWAS variant but the gene for the biomarker was unidentified as the candidate for the GWAS signal (INHBC, TNFRSF11A, TCN2, PXN1, PRTN3, PSMD9, TFPI, ITGB6, CA3). Single-cell transcriptomic data indicated the 22 biomarkers are expressed in kidney tubules, collecting duct, fibroblasts, and immune cells. Pathway analysis showed significant enrichment of identified biomarkers in the extracellular kidney parenchyma. Thus, using genetic regulators of biomarker concentration via proteome-wide Mendelian randomization, we identified 22 biomarkers that appear to causally impact eGFR in either a beneficial or adverse manner. The current study provides rationale for novel therapeutic targets for eGFR and emphasized a role for extracellular proteins produced by tubular cells and fibroblasts for impacting eGFR.
Collapse
Affiliation(s)
- Matthew B Lanktree
- Population Health Research Institute, Hamilton, Ontario, Canada; Division of Nephrology, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Nicolas Perrot
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Andrew Smyth
- Population Health Research Institute, Hamilton, Ontario, Canada; HRB Clinical Research Facility Galway, University of Galway, Galway, Ireland
| | - Michael Chong
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sukrit Narula
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Zachary Kroezen
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Philip Britz-Mckibbin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Mario Berger
- Bayer AG, Pharmaceuticals Research & Development, Pharma Research Center, Wuppertal, Germany
| | - Joan C Krepinsky
- Division of Nephrology, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Marie Pigeyre
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Salim Yusuf
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Zhu Z, Chen X, Zhang S, Yu R, Qi C, Cheng L, Zhang X. Leveraging molecular quantitative trait loci to comprehend complex diseases/traits from the omics perspective. Hum Genet 2023; 142:1543-1560. [PMID: 37755483 DOI: 10.1007/s00439-023-02602-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
Comprehending the molecular basis of quantitative genetic variation is a principal goal for complex diseases or traits. Molecular quantitative trait loci (molQTLs) have made it possible to investigate the effects of genetic variants hiding behind large-scale omics data. A deeper understanding of molQTL is urgently required in light of the multi-dimensionalization of omics data to more fully elucidate the pertinent biological mechanisms. Herein, we reviewed molQTLs with the corresponding resource from the omics perspective and further discussed the integrative strategy of GWAS-molQTL to infer their causal effects. Subsequently, we described the opportunities and challenges encountered by molQTL. The case studies showed that molQTL is essential for complex diseases and traits, whether single- or multi-omics QTLs. Overall, we highlighted the functional significance of genetic variants to employ the discovery of molQTL in complex diseases and traits.
Collapse
Affiliation(s)
- Zijun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xinyu Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Sainan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Rui Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Changlu Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150028, Heilongjiang, China.
| | - Xue Zhang
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150028, Heilongjiang, China
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
12
|
Jiang MZ, Gaynor SM, Li X, Van Buren E, Stilp A, Buth E, Wang FF, Manansala R, Gogarten SM, Li Z, Polfus LM, Salimi S, Bis JC, Pankratz N, Yanek LR, Durda P, Tracy RP, Rich SS, Rotter JI, Mitchell BD, Lewis JP, Psaty BM, Pratte KA, Silverman EK, Kaplan RC, Avery C, North K, Mathias RA, Faraday N, Lin H, Wang B, Carson AP, Norwood AF, Gibbs RA, Kooperberg C, Lundin J, Peters U, Dupuis J, Hou L, Fornage M, Benjamin EJ, Reiner AP, Bowler RP, Lin X, Auer PL, Raffield LM, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, TOPMed Inflammation Working Group. Whole Genome Sequencing Based Analysis of Inflammation Biomarkers in the Trans-Omics for Precision Medicine (TOPMed) Consortium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.555215. [PMID: 37745480 PMCID: PMC10515765 DOI: 10.1101/2023.09.10.555215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Inflammation biomarkers can provide valuable insight into the role of inflammatory processes in many diseases and conditions. Sequencing based analyses of such biomarkers can also serve as an exemplar of the genetic architecture of quantitative traits. To evaluate the biological insight, which can be provided by a multi-ancestry, whole-genome based association study, we performed a comprehensive analysis of 21 inflammation biomarkers from up to 38,465 individuals with whole-genome sequencing from the Trans-Omics for Precision Medicine (TOPMed) program. We identified 22 distinct single-variant associations across 6 traits - E-selectin, intercellular adhesion molecule 1, interleukin-6, lipoprotein-associated phospholipase A2 activity and mass, and P-selectin - that remained significant after conditioning on previously identified associations for these inflammatory biomarkers. We further expanded upon known biomarker associations by pairing the single-variant analysis with a rare variant set-based analysis that further identified 19 significant rare variant set-based associations with 5 traits. These signals were distinct from both significant single variant association signals within TOPMed and genetic signals observed in prior studies, demonstrating the complementary value of performing both single and rare variant analyses when analyzing quantitative traits. We also confirm several previously reported signals from semi-quantitative proteomics platforms. Many of these signals demonstrate the extensive allelic heterogeneity and ancestry-differentiated variant-trait associations common for inflammation biomarkers, a characteristic we hypothesize will be increasingly observed with well-powered, large-scale analyses of complex traits.
Collapse
Affiliation(s)
- Min-Zhi Jiang
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - Sheila M. Gaynor
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
- Regeneron Genetics Center, Tarrytown, NY, 10591, USA
| | - Xihao Li
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Eric Van Buren
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
| | - Adrienne Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, 98105, USA
| | - Erin Buth
- Department of Biostatistics, University of Washington, Seattle, WA, 98105, USA
| | - Fei Fei Wang
- Department of Biostatistics, University of Washington, Seattle, WA, 98105, USA
| | - Regina Manansala
- Centre for Health Economics Research & Modelling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO) WHO Collaborating Centre, University of Antwerp, Antwerp, BE
| | | | - Zilin Li
- School of Mathematics and Statistics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Linda M. Polfus
- Department of Preventive Medicine, Center for Genetic Epidemiology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shabnam Salimi
- Department of Epidemiology and Public Health, Division of Gerontology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave NE, Box 359458, Seattle, WA, 98195, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Lisa R. Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, 1830 E Monument St Rm 8024, Baltimore, MD, 21287, USA
| | - Peter Durda
- Department of Pathology & Laboratory Medicine, University of Vermont Larner College of Medicine, 360 South Park Drive, Colchester, VT, 05446, USA
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine, University of Vermont Larner College of Medicine, 360 South Park Drive, Colchester, VT, 05446, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA, 90502, USA
| | - Braxton D. Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, 670 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Joshua P. Lewis
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, 670 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 4333 Brooklyn Ave NE, Box 359458, Seattle, WA, 98195, USA
- Departments of Epidemiology and Health Systems and Population Health, University of Washington, 4333 Brooklyn Ave NE, Seattle, WA, 98101, USA
| | - Katherine A. Pratte
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Edwin K. Silverman
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Christy Avery
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kari North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rasika A. Mathias
- Department of Medicine, Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Cir JHAAC Room 3B53, Baltimore, MD, 21287, USA
| | - Nauder Faraday
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD, 21287, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, 55 Lake Ave North, Worcester, MA, 01655, USA
| | - Biqi Wang
- Department of Medicine, University of Massachusetts Chan Medical School, 55 Lake Ave North, Worcester, MA, 01655, USA
| | - April P. Carson
- Department of Medicine, University of Mississippi Medical Center, 350 W. Woodrow Wilson Avenue, Suite 701, Jackson, MS, 39213, USA
| | - Arnita F. Norwood
- Department of Medicine, University of Mississippi Medical Center, 350 W. Woodrow Wilson Avenue, Suite 701, Jackson, MS, 39213, USA
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jessica Lundin
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Josée Dupuis
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Québec, H3A 1G1, Canada
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Emelia J. Benjamin
- Department of Medicine, Cardiovascular Medicine, Boston Medical Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, 02118, USA
- Boston University and National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, 01702, USA
| | - Alexander P. Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, 98105, USA
| | - Russell P. Bowler
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
| | - Paul L. Auer
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | | |
Collapse
|
13
|
Hill AC, Guo C, Litkowski EM, Manichaikul AW, Yu B, Konigsberg IR, Gorbet BA, Lange LA, Pratte KA, Kechris KJ, DeCamp M, Coors M, Ortega VE, Rich SS, Rotter JI, Gerzsten RE, Clish CB, Curtis JL, Hu X, Obeidat ME, Morris M, Loureiro J, Ngo D, O'Neal WK, Meyers DA, Bleecker ER, Hobbs BD, Cho MH, Banaei-Kashani F, Bowler RP. Large scale proteomic studies create novel privacy considerations. Sci Rep 2023; 13:9254. [PMID: 37286633 PMCID: PMC10247808 DOI: 10.1038/s41598-023-34866-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Privacy protection is a core principle of genomic but not proteomic research. We identified independent single nucleotide polymorphism (SNP) quantitative trait loci (pQTL) from COPDGene and Jackson Heart Study (JHS), calculated continuous protein level genotype probabilities, and then applied a naïve Bayesian approach to link SomaScan 1.3K proteomes to genomes for 2812 independent subjects from COPDGene, JHS, SubPopulations and InteRmediate Outcome Measures In COPD Study (SPIROMICS) and Multi-Ethnic Study of Atherosclerosis (MESA). We correctly linked 90-95% of proteomes to their correct genome and for 95-99% we identify the 1% most likely links. The linking accuracy in subjects with African ancestry was lower (~ 60%) unless training included diverse subjects. With larger profiling (SomaScan 5K) in the Atherosclerosis Risk Communities (ARIC) correct identification was > 99% even in mixed ancestry populations. We also linked proteomes-to-proteomes and used the proteome only to determine features such as sex, ancestry, and first-degree relatives. When serial proteomes are available, the linking algorithm can be used to identify and correct mislabeled samples. This work also demonstrates the importance of including diverse populations in omics research and that large proteomic datasets (> 1000 proteins) can be accurately linked to a specific genome through pQTL knowledge and should not be considered unidentifiable.
Collapse
Affiliation(s)
| | | | | | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Bing Yu
- Department of Epidemiology and Human Genetics Center, UTHealth School of Public Health, Houston, TX, USA
| | | | - Betty A Gorbet
- Department of Epidemiology and Human Genetics Center, UTHealth School of Public Health, Houston, TX, USA
| | - Leslie A Lange
- University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Matthew DeCamp
- University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Marilyn Coors
- University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Robert E Gerzsten
- Division of Cardiovascular Medicine, Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | | | - Xiaowei Hu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | - Wanda K O'Neal
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Brian D Hobbs
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael H Cho
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | |
Collapse
|
14
|
Van Buren E, Radicioni G, Lester S, O’Neal WK, Dang H, Kasela S, Garudadri S, Curtis JL, Han MK, Krishnan JA, Wan ES, Silverman EK, Hastie A, Ortega VE, Lappalainen T, Nawijn MC, van den Berge M, Christenson SA, Li Y, Cho MH, Kesimer M, Kelada SNP. Genetic regulators of sputum mucin concentration and their associations with COPD phenotypes. PLoS Genet 2023; 19:e1010445. [PMID: 37352370 PMCID: PMC10325042 DOI: 10.1371/journal.pgen.1010445] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 07/06/2023] [Accepted: 04/26/2023] [Indexed: 06/25/2023] Open
Abstract
Hyper-secretion and/or hyper-concentration of mucus is a defining feature of multiple obstructive lung diseases, including chronic obstructive pulmonary disease (COPD). Mucus itself is composed of a mixture of water, ions, salt and proteins, of which the gel-forming mucins, MUC5AC and MUC5B, are the most abundant. Recent studies have linked the concentrations of these proteins in sputum to COPD phenotypes, including chronic bronchitis (CB) and acute exacerbations (AE). We sought to determine whether common genetic variants influence sputum mucin concentrations and whether these variants are also associated with COPD phenotypes, specifically CB and AE. We performed a GWAS to identify quantitative trait loci for sputum mucin protein concentration (pQTL) in the Sub-Populations and InteRmediate Outcome Measures in COPD Study (SPIROMICS, n = 708 for total mucin, n = 215 for MUC5AC, MUC5B). Subsequently, we tested for associations of mucin pQTL with CB and AE using regression modeling (n = 822-1300). Replication analysis was conducted using data from COPDGene (n = 5740) and by examining results from the UK Biobank. We identified one genome-wide significant pQTL for MUC5AC (rs75401036) and two for MUC5B (rs140324259, rs10001928). The strongest association for MUC5B, with rs140324259 on chromosome 11, explained 14% of variation in sputum MUC5B. Despite being associated with lower MUC5B, the C allele of rs140324259 conferred increased risk of CB (odds ratio (OR) = 1.42; 95% confidence interval (CI): 1.10-1.80) as well as AE ascertained over three years of follow up (OR = 1.41; 95% CI: 1.02-1.94). Associations between rs140324259 and CB or AE did not replicate in COPDGene. However, in the UK Biobank, rs140324259 was associated with phenotypes that define CB, namely chronic mucus production and cough, again with the C allele conferring increased risk. We conclude that sputum MUC5AC and MUC5B concentrations are associated with common genetic variants, and the top locus for MUC5B may influence COPD phenotypes, in particular CB.
Collapse
Affiliation(s)
- Eric Van Buren
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Giorgia Radicioni
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Sarah Lester
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Silva Kasela
- New York Genome Center, New York, New York, United States of America
- Department of Systems Biology, Columbia University, New York, New York, United States of America
| | - Suresh Garudadri
- Division of Pulmonary, Critical Care, Allergy, & Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Jeffrey L. Curtis
- Pulmonary & Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan, United States of America
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
| | - MeiLan K. Han
- Pulmonary & Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jerry A. Krishnan
- Breathe Chicago Center, University of Illinois, Chicago, Illinois, United States of America
| | - Emily S. Wan
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- VA Boston Healthcare System, Jamaica Plain, Massachusetts, United States of America
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Annette Hastie
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Victor E. Ortega
- Department of Internal Medicine, Division of Respiratory Medicine, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Tuuli Lappalainen
- New York Genome Center, New York, New York, United States of America
- Department of Systems Biology, Columbia University, New York, New York, United States of America
| | - Martijn C. Nawijn
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stephanie A. Christenson
- Division of Pulmonary, Critical Care, Allergy, & Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Yun Li
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mehmet Kesimer
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Samir N. P. Kelada
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
15
|
DiLillo KM, Norman KC, Freeman CM, Christenson SA, Alexis NE, Anderson WH, Barjaktarevic IZ, Barr RG, Comellas AP, Bleecker ER, Boucher RC, Couper DJ, Criner GJ, Doerschuk CM, Wells JM, Han MK, Hoffman EA, Hansel NN, Hastie AT, Kaner RJ, Krishnan JA, Labaki WW, Martinez FJ, Meyers DA, O'Neal WK, Ortega VE, Paine R, Peters SP, Woodruff PG, Cooper CB, Bowler RP, Curtis JL, Arnold KB. A blood and bronchoalveolar lavage protein signature of rapid FEV 1 decline in smoking-associated COPD. Sci Rep 2023; 13:8228. [PMID: 37217548 PMCID: PMC10203309 DOI: 10.1038/s41598-023-32216-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/24/2023] [Indexed: 05/24/2023] Open
Abstract
Accelerated progression of chronic obstructive pulmonary disease (COPD) is associated with increased risks of hospitalization and death. Prognostic insights into mechanisms and markers of progression could facilitate development of disease-modifying therapies. Although individual biomarkers exhibit some predictive value, performance is modest and their univariate nature limits network-level insights. To overcome these limitations and gain insights into early pathways associated with rapid progression, we measured 1305 peripheral blood and 48 bronchoalveolar lavage proteins in individuals with COPD [n = 45, mean initial forced expiratory volume in one second (FEV1) 75.6 ± 17.4% predicted]. We applied a data-driven analysis pipeline, which enabled identification of protein signatures that predicted individuals at-risk for accelerated lung function decline (FEV1 decline ≥ 70 mL/year) ~ 6 years later, with high accuracy. Progression signatures suggested that early dysregulation in elements of the complement cascade is associated with accelerated decline. Our results propose potential biomarkers and early aberrant signaling mechanisms driving rapid progression in COPD.
Collapse
Affiliation(s)
- Katarina M DiLillo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Katy C Norman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Christine M Freeman
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wayne H Anderson
- Marsico Lung Institute/Pulmonary and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Igor Z Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Alejandro P Comellas
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa, Iowa City, IA, USA
| | - Eugene R Bleecker
- Division of Genetics, Genomics and Precision Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Richard C Boucher
- Marsico Lung Institute/Cystic Fibrosis Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David J Couper
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, USA
| | - Claire M Doerschuk
- Marsico Lung Institute/Cystic Fibrosis Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J Michael Wells
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - MeiLan K Han
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Annette T Hastie
- Department of Internal Medicine, Wake Forest School of Medicine, Atrium Health, Wake Forest Baptist, Winston Salem, NC, USA
| | - Robert J Kaner
- Department of Medicine, Weill Cornell Medical Center, New York, NY, USA
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Wassim W Labaki
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Deborah A Meyers
- Division of Genetics, Genomics and Precision Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Wanda K O'Neal
- Marsico Lung Institute/Cystic Fibrosis Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victor E Ortega
- Department of Internal Medicine, Division of Respiratory Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA
| | - Stephen P Peters
- Department of Internal Medicine, Wake Forest School of Medicine, Atrium Health, Wake Forest Baptist, Winston Salem, NC, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christopher B Cooper
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Russell P Bowler
- Division of Pulmonary and Critical Care, National Jewish Health, Denver, CO, USA
| | - Jeffrey L Curtis
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Biological and Genetic Mechanisms of COPD, Its Diagnosis, Treatment, and Relationship with Lung Cancer. Biomedicines 2023; 11:biomedicines11020448. [PMID: 36830984 PMCID: PMC9953173 DOI: 10.3390/biomedicines11020448] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most prevalent chronic adult diseases, with significant worldwide morbidity and mortality. Although long-term tobacco smoking is a critical risk factor for this global health problem, its molecular mechanisms remain unclear. Several phenomena are thought to be involved in the evolution of emphysema, including airway inflammation, proteinase/anti-proteinase imbalance, oxidative stress, and genetic/epigenetic modifications. Furthermore, COPD is one main risk for lung cancer (LC), the deadliest form of human tumor; formation and chronic inflammation accompanying COPD can be a potential driver of malignancy maturation (0.8-1.7% of COPD cases develop cancer/per year). Recently, the development of more research based on COPD and lung cancer molecular analysis has provided new light for understanding their pathogenesis, improving the diagnosis and treatments, and elucidating many connections between these diseases. Our review emphasizes the biological factors involved in COPD and lung cancer, the advances in their molecular mechanisms' research, and the state of the art of diagnosis and treatments. This work combines many biological and genetic elements into a single whole and strongly links COPD with lung tumor features.
Collapse
|
17
|
Lee KK, Norris ET, Rishishwar L, Conley AB, Mariño-Ramírez L, McDonald JF, Jordan IK. Ethnic disparities in mortality and group-specific risk factors in the UK Biobank. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001560. [PMID: 36963080 PMCID: PMC10021328 DOI: 10.1371/journal.pgph.0001560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/09/2023] [Indexed: 02/25/2023]
Abstract
Despite a substantial overall decrease in mortality, disparities among ethnic minorities in developed countries persist. This study investigated mortality disparities and their associated risk factors for the three largest ethnic groups in the United Kingdom: Asian, Black, and White. Study participants were sampled from the UK Biobank (UKB), a prospective cohort enrolled between 2006 and 2010. Genetics, biological samples, and health information and outcomes data of UKB participants were downloaded and data-fields were prioritized based on participants with death registry records. Kaplan-Meier method was used to evaluate survival differences among ethnic groups; survival random forest feature selection followed by Cox proportional-hazard modeling was used to identify and estimate the effects of shared and ethnic group-specific mortality risk factors. The White ethnic group showed significantly worse survival probability than the Asian and Black groups. In all three ethnic groups, endoscopy and colonoscopy procedures showed significant protective effects on overall mortality. Asian and Black women show lower relative risk of mortality than men, whereas no significant effect of sex was seen for the White group. The strongest ethnic group-specific mortality associations were ischemic heart disease for Asians, COVID-19 for Blacks, and cancers of respiratory/intrathoracic organs for Whites. Mental health-related diagnoses, including substance abuse, anxiety, and depression, were a major risk factor for overall mortality in the Asian group. The effect of mental health on Asian mortality, particularly for digestive cancers, was exacerbated by an observed hesitance to answer mental health questions, possibly related to cultural stigma. C-reactive protein (CRP) serum levels were associated with both overall and cause-specific mortality due to COVID-19 and digestive cancers in the Black group, where elevated CRP has previously been linked to psychosocial stress due to discrimination. Our results point to mortality risk factors that are group-specific and modifiable, supporting targeted interventions towards greater health equity.
Collapse
Affiliation(s)
- Kara Keun Lee
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Emily T Norris
- IHRC-Georgia Tech Applied Bioinformatics Laboratory, Atlanta, GA, United States of America
| | - Lavanya Rishishwar
- IHRC-Georgia Tech Applied Bioinformatics Laboratory, Atlanta, GA, United States of America
| | - Andrew B Conley
- IHRC-Georgia Tech Applied Bioinformatics Laboratory, Atlanta, GA, United States of America
| | - Leonardo Mariño-Ramírez
- National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, United States of America
| | - John F McDonald
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - I King Jordan
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
- IHRC-Georgia Tech Applied Bioinformatics Laboratory, Atlanta, GA, United States of America
| |
Collapse
|
18
|
Vu T, Litkowski EM, Liu W, Pratte KA, Lange L, Bowler RP, Banaei-Kashani F, Kechris KJ. NetSHy: network summarization via a hybrid approach leveraging topological properties. Bioinformatics 2023; 39:6957083. [PMID: 36548341 PMCID: PMC9831052 DOI: 10.1093/bioinformatics/btac818] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/30/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Biological networks can provide a system-level understanding of underlying processes. In many contexts, networks have a high degree of modularity, i.e. they consist of subsets of nodes, often known as subnetworks or modules, which are highly interconnected and may perform separate functions. In order to perform subsequent analyses to investigate the association between the identified module and a variable of interest, a module summarization, that best explains the module's information and reduces dimensionality is often needed. Conventional approaches for obtaining network representation typically rely only on the profiles of the nodes within the network while disregarding the inherent network topological information. RESULTS In this article, we propose NetSHy, a hybrid approach which is capable of reducing the dimension of a network while incorporating topological properties to aid the interpretation of the downstream analyses. In particular, NetSHy applies principal component analysis (PCA) on a combination of the node profiles and the well-known Laplacian matrix derived directly from the network similarity matrix to extract a summarization at a subject level. Simulation scenarios based on random and empirical networks at varying network sizes and sparsity levels show that NetSHy outperforms the conventional PCA approach applied directly on node profiles, in terms of recovering the true correlation with a phenotype of interest and maintaining a higher amount of explained variation in the data when networks are relatively sparse. The robustness of NetSHy is also demonstrated by a more consistent correlation with the observed phenotype as the sample size decreases. Lastly, a genome-wide association study is performed as an application of a downstream analysis, where NetSHy summarization scores on the biological networks identify more significant single nucleotide polymorphisms than the conventional network representation. AVAILABILITY AND IMPLEMENTATION R code implementation of NetSHy is available at https://github.com/thaovu1/NetSHy. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Thao Vu
- To whom correspondence should be addressed. or
| | - Elizabeth M Litkowski
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Biomedical Informatics & Personalized Medicine, School of Medicine, Colorado University Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Weixuan Liu
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katherine A Pratte
- Department of Biostatistics, National Jewish Health, Denver, CO 80206, USA
| | - Leslie Lange
- Division of Biomedical Informatics & Personalized Medicine, School of Medicine, Colorado University Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Russell P Bowler
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Farnoush Banaei-Kashani
- Department of Computer Science and Engineering, College of Engineering, Design and Computing, University of Colorado Denver, Denver, CO 80204, USA
| | | |
Collapse
|
19
|
Wang K, Cadzow M, Bixley M, Leask MP, Merriman ME, Yang Q, Li Z, Takei R, Phipps-Green A, Major TJ, Topless R, Dalbeth N, King F, Murphy R, Stamp LK, de Zoysa J, Wang Z, Shi Y, Merriman TR. A Polynesian-specific copy number variant encompassing the MICA gene associates with gout. Hum Mol Genet 2022; 31:3757-3768. [PMID: 35451026 PMCID: PMC9616569 DOI: 10.1093/hmg/ddac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/01/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Gout is of particularly high prevalence in the Māori and Pacific (Polynesian) populations of Aotearoa New Zealand (NZ). Here, we investigated the contribution of common population-specific copy number variation (CNV) to gout in the Aotearoa NZ Polynesian population. Microarray-generated genome-wide genotype data from Aotearoa NZ Polynesian individuals with (n = 1196) and without (n = 1249) gout were analyzed. Comparator population groups were 552 individuals of European ancestry and 1962 of Han Chinese ancestry. Levels of circulating major histocompatibility complex (MHC) class I polypeptide-related sequence A (MICA) were measured by enzyme-linked immunosorbent assay. Fifty-four CNV regions (CNVRs) appearing in at least 10 individuals were detected, of which seven common (>2%) CNVRs were specific to or amplified in Polynesian people. A burden test of these seven revealed associations of insertion/deletion with gout (odds ratio (OR) 95% confidence interval [CI] = 1.80 [1.01; 3.22], P = 0.046). Individually testing of the seven CNVRs for association with gout revealed nominal association of CNVR1 with gout in Western Polynesian (Chr6: 31.36-31.45 Mb, OR = 1.72 [1.03; 2.92], P = 0.04), CNVR6 in the meta-analyzed Polynesian sample sets (Chr1: 196.75-196.92 Mb, OR = 1.86 [1.16; 3.00], P = 0.01) and CNVR9 in Western Polynesian (Chr1: 189.35-189.54 Mb, OR = 2.75 [1.15; 7.13], P = 0.03). Analysis of European gout genetic association data demonstrated a signal of association at the CNVR1 locus that was an expression quantitative trait locus for MICA. The most common CNVR (CNVR1) includes deletion of the MICA gene, encoding an immunomodulatory protein. Expression of MICA was reduced in the serum of individuals with the deletion. In summary, we provide evidence for the association of CNVR1 containing MICA with gout in Polynesian people, implicating class I MHC-mediated antigen presentation in gout.
Collapse
Affiliation(s)
- Ke Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Matt Bixley
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Megan P Leask
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Marilyn E Merriman
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
| | - Zhiqiang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
- Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao 266003, China
| | - Riku Takei
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | - Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Ruth Topless
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland 1023, New Zealand
| | - Frances King
- Ngati Porou Hauora Charitable Trust, Te Puia Springs, New Zealand
| | - Rinki Murphy
- Department of Medicine, University of Auckland, Auckland 1023, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch 8013, New Zealand
| | - Janak de Zoysa
- Department of Medicine, University of Auckland, Auckland 1023, New Zealand
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
- Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao 266003, China
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
20
|
Kress S, Wigmann C, Zhao Q, Herder C, Abramson MJ, Schwender H, Schikowski T. Chronic air pollution-induced subclinical airway inflammation and polygenic susceptibility. Respir Res 2022; 23:265. [PMID: 36151579 PMCID: PMC9508765 DOI: 10.1186/s12931-022-02179-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Background Air pollutants can activate low-grade subclinical inflammation which further impairs respiratory health. We aimed to investigate the role of polygenic susceptibility to chronic air pollution-induced subclinical airway inflammation. Methods We used data from 296 women (69–79 years) enrolled in the population-based SALIA cohort (Study on the influence of Air pollution on Lung function, Inflammation and Aging). Biomarkers of airway inflammation were measured in induced-sputum samples at follow-up investigation in 2007–2010. Chronic air pollution exposures at residential addresses within 15 years prior to the biomarker assessments were used to estimate main environmental effects on subclinical airway inflammation. Furthermore, we calculated internally weighted polygenic risk scores based on genome-wide derived single nucleotide polymorphisms. Polygenic main and gene-environment interaction (GxE) effects were investigated by adjusted linear regression models. Results Higher exposures to nitrogen dioxide (NO2), nitrogen oxides (NOx), particulate matter with aerodynamic diameters of ≤ 2.5 μm, ≤ 10 μm, and 2.5–10 µm significantly increased the levels of leukotriene (LT)B4 by 19.7% (p-value = 0.005), 20.9% (p = 0.002), 22.1% (p = 0.004), 17.4% (p = 0.004), and 23.4% (p = 0.001), respectively. We found significant effects of NO2 (25.9%, p = 0.008) and NOx (25.9%, p-value = 0.004) on the total number of cells. No significant GxE effects were observed. The trends were mostly robust in sensitivity analyses. Conclusions While this study confirms that higher chronic exposures to air pollution increase the risk of subclinical airway inflammation in elderly women, we could not demonstrate a significant role of polygenic susceptibility on this pathway. Further studies are required to investigate the role of polygenic susceptibility. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02179-3.
Collapse
Affiliation(s)
- Sara Kress
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.,Medical Research School Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Claudia Wigmann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany
| | - Qi Zhao
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.,Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael J Abramson
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Holger Schwender
- Mathematical Institute, Heinrich Heine University, Düsseldorf, Germany
| | - Tamara Schikowski
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.
| |
Collapse
|
21
|
Pott J, Garcia T, Hauck SM, Petrera A, Wirkner K, Loeffler M, Kirsten H, Peters A, Scholz M. Genetically regulated gene expression and proteins revealed discordant effects. PLoS One 2022; 17:e0268815. [PMID: 35604899 PMCID: PMC9126407 DOI: 10.1371/journal.pone.0268815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 05/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background Although gene-expression (GE) and protein levels are typically strongly genetically regulated, their correlation is known to be low. Here we investigate this phenomenon by focusing on the genetic background of this correlation in order to understand the similarities and differences in the genetic regulation of these omics layers. Methods and results We performed locus-wide association studies of 92 protein levels measured in whole blood for 2,014 samples of European ancestry and found that 66 are genetically regulated. Three female- and one male-specific effects were detected. We estimated the genetically regulated GE for all significant genes in 49 GTEx v8 tissues. A total of 7 proteins showed negative correlations with their respective GE across multiple tissues. Finally, we tested for causal links of GE on protein expression via Mendelian Randomization, and confirmed a negative causal effect of GE on protein level for five of these genes in a total of 63 gene-tissue pairs: BLMH, CASP3, CXCL16, IL6R, and SFTPD. For IL6R, we replicated the negative causal effect on coronary-artery disease (CAD), while its GE was positively linked to CAD. Conclusion While total GE and protein levels are only weakly correlated, we found high correlations between their genetically regulated components across multiple tissues. Of note, strong negative causal effects of tissue-specific GE on five protein levels were detected. Causal network analyses revealed that GE effects on CAD risks was in general mediated by protein levels.
Collapse
Affiliation(s)
- Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
- * E-mail: (JP); (MS)
| | - Tarcyane Garcia
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Zentrum Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Zentrum Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Kerstin Wirkner
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Annette Peters
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Zentrum Munich - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
- * E-mail: (JP); (MS)
| |
Collapse
|
22
|
Reed D, Kumar D, Kumar S, Raina K, Punia R, Kant R, Saba L, Cruickshank-Quinn C, Tabakoff B, Reisdorph N, Edwards MG, Wempe M, Agarwal C, Agarwal R. Transcriptome and metabolome changes induced by bitter melon ( Momordica charantia)- intake in a high-fat diet induced obesity model. J Tradit Complement Med 2022; 12:287-301. [PMID: 35493312 PMCID: PMC9039170 DOI: 10.1016/j.jtcme.2021.08.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/22/2022] Open
Abstract
Background and aim Metabolic syndrome (MetS) is a complex disease of physiological imbalances interrelated to abnormal metabolic conditions, such as abdominal obesity, type II diabetes, dyslipidemia and hypertension. In the present pilot study, we investigated the nutraceutical bitter melon (Momordica charantia L) -intake induced transcriptome and metabolome changes and the converging metabolic signaling networks underpinning its inhibitory effects against MetS-associated risk factors. Experimental procedure Metabolic effects of lyophilized bitter melon juice (BMJ) extract (oral gavage 200 mg/kg/body weight-daily for 40 days) intake were evaluated in diet-induced obese C57BL/6J male mice [fed-high fat diet (HFD), 60 kcal% fat]. Changes in a) serum levels of biochemical parameters, b) gene expression in the hepatic transcriptome (microarray analysis using Affymetrix Mouse Exon 1.0 ST arrays), and c) metabolite abundance levels in lipid-phase plasma [liquid chromatography mass spectrometry (LC-MS)-based metabolomics] after BMJ intervention were assessed. Results and conclusion BMJ-mediated changes showed a positive trend towards enhanced glucose homeostasis, vitamin D metabolism and suppression of glycerophospholipid metabolism. In the liver, nuclear peroxisome proliferator-activated receptor (PPAR) and circadian rhythm signaling, as well as bile acid biosynthesis and glycogen metabolism targets were modulated by BMJ (p < 0.05). Thus, our in-depth transcriptomics and metabolomics analysis suggests that BMJ-intake lowers susceptibility to the onset of high-fat diet associated MetS risk factors partly through modulation of PPAR signaling and its downstream targets in circadian rhythm processes to prevent excessive lipogenesis, maintain glucose homeostasis and modify immune responses signaling.
Collapse
Key Words
- AMPK, adenosine monophosphate-activated protein kinase
- BMJ, bitter melon juice
- Bitter melon
- DIO, diet-induced obese
- Diet intervention
- HDL, high density lipoprotein (cholesterol)
- HFD, high fat diet
- HMDB, Human Metabolome Database
- High fat diet-induced obesity
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LC-MS, liquid-chromatography mass spectrometry
- LDL, low density lipoprotein (cholesterol)
- MetS, Metabolic syndrome
- Metabolic syndrome
- Momordica charantia
- PC, phosphatidylcholine
- PE, phosphatidylethanolamine
- PPARs, Peroxisome proliferator-activated receptors
Collapse
Affiliation(s)
- Dominique Reed
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dileep Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sushil Kumar
- Division of Critical Care Medicine and Cardiovascular Pulmonary Research, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD, USA
| | - Reenu Punia
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Laura Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charmion Cruickshank-Quinn
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Michael Wempe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
23
|
Cho MH, Hobbs BD, Silverman EK. Genetics of chronic obstructive pulmonary disease: understanding the pathobiology and heterogeneity of a complex disorder. THE LANCET. RESPIRATORY MEDICINE 2022; 10:485-496. [PMID: 35427534 PMCID: PMC11197974 DOI: 10.1016/s2213-2600(21)00510-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a deadly and highly morbid disease. Susceptibility to and heterogeneity of COPD are incompletely explained by environmental factors such as cigarette smoking. Family-based and population-based studies have shown that a substantial proportion of COPD risk is related to genetic variation. Genetic association studies have identified hundreds of genetic variants that affect risk for COPD, decreased lung function, and other COPD-related traits. These genetic variants are associated with other pulmonary and non-pulmonary traits, demonstrate a genetic basis for at least part of COPD heterogeneity, have a substantial effect on COPD risk in aggregate, implicate early-life events in COPD pathogenesis, and often involve genes not previously suspected to have a role in COPD. Additional progress will require larger genetic studies with more ancestral diversity, improved profiling of rare variants, and better statistical methods. Through integration of genetic data with other omics data and comprehensive COPD phenotypes, as well as functional description of causal mechanisms for genetic risk variants, COPD genetics will continue to inform novel approaches to understanding the pathobiology of COPD and developing new strategies for management and treatment.
Collapse
Affiliation(s)
- Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Brian D Hobbs
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Zhang C, Hansen MEB, Tishkoff SA. Advances in integrative African genomics. Trends Genet 2022; 38:152-168. [PMID: 34740451 PMCID: PMC8752515 DOI: 10.1016/j.tig.2021.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
There has been a rapid increase in human genome sequencing in the past two decades, resulting in the identification of millions of previously unknown genetic variants. However, African populations are under-represented in sequencing efforts. Additional sequencing from diverse African populations and the construction of African-specific reference genomes is needed to better characterize the full spectrum of variation in humans. However, sequencing alone is insufficient to address the molecular and cellular mechanisms underlying variable phenotypes and disease risks. Determining functional consequences of genetic variation using multi-omics approaches is a fundamental post-genomic challenge. We discuss approaches to close the knowledge gaps about African genomic diversity and review advances in African integrative genomic studies and their implications for precision medicine.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew E B Hansen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah A Tishkoff
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Kühnapfel A, Horn K, Klotz U, Kiehntopf M, Rosolowski M, Loeffler M, Ahnert P, Suttorp N, Witzenrath M, Scholz M. Genetic Regulation of Cytokine Response in Patients with Acute Community-Acquired Pneumonia. Genes (Basel) 2022; 13:genes13010111. [PMID: 35052452 PMCID: PMC8774373 DOI: 10.3390/genes13010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 02/05/2023] Open
Abstract
Background: Community-acquired pneumonia (CAP) is an acute disease condition with a high risk of rapid deteriorations. We analysed the influence of genetics on cytokine regulation to obtain a better understanding of patient’s heterogeneity. Methods: For up to N = 389 genotyped participants of the PROGRESS study of hospitalised CAP patients, we performed a genome-wide association study of ten cytokines IL-1β, IL-6, IL-8, IL-10, IL-12, MCP-1 (MCAF), MIP-1α (CCL3), VEGF, VCAM-1, and ICAM-1. Consecutive secondary analyses were performed to identify independent hits and corresponding causal variants. Results: 102 SNPs from 14 loci showed genome-wide significant associations with five of the cytokines. The most interesting associations were found at 6p21.1 for VEGF (p = 1.58 × 10−20), at 17q21.32 (p = 1.51 × 10−9) and at 10p12.1 (p = 2.76 × 10−9) for IL-1β, at 10p13 for MIP-1α (CCL3) (p = 2.28 × 10−9), and at 9q34.12 for IL-10 (p = 4.52 × 10−8). Functionally plausible genes could be assigned to the majority of loci including genes involved in cytokine secretion, granulocyte function, and cilial kinetics. Conclusion: This is the first context-specific genetic association study of blood cytokine concentrations in CAP patients revealing numerous biologically plausible candidate genes. Two of the loci were also associated with atherosclerosis with probable common or consecutive pathomechanisms.
Collapse
Affiliation(s)
- Andreas Kühnapfel
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
- Correspondence:
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
| | - Ulrike Klotz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
| | - Michael Kiehntopf
- Institute for Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, 07740 Jena, Germany;
| | - Maciej Rosolowski
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
| | - Norbert Suttorp
- Division of Infectiology and Pneumonology, Medical Department, Charité—Berlin University Medicine, 13353 Berlin, Germany; (N.S.); (M.W.)
| | - Martin Witzenrath
- Division of Infectiology and Pneumonology, Medical Department, Charité—Berlin University Medicine, 13353 Berlin, Germany; (N.S.); (M.W.)
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (K.H.); (U.K.); (M.R.); (M.L.); (P.A.); (M.S.)
| |
Collapse
|
26
|
Keefe J, Yao C, Hwang SJ, Courchesne P, Lee GY, Dupuis J, Mizgerd JP, O’Connor G, Washko GR, Cho MH, Silverman EK, Levy D. An Integrative Genomic Strategy Identifies sRAGE as a Causal and Protective Biomarker of Lung Function. Chest 2022; 161:76-84. [PMID: 34237330 PMCID: PMC8783029 DOI: 10.1016/j.chest.2021.06.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND There are few clinically useful circulating biomarkers of lung function and lung disease. We hypothesized that genome-wide association studies (GWAS) of circulating proteins in conjunction with GWAS of pulmonary traits represents a clinically relevant approach to identifying causal proteins and therapeutically useful insights into mechanisms related to lung function and disease. STUDY QUESTION Can an integrative genomic strategy using GWAS of plasma soluble receptor for advanced glycation end-products (sRAGE) levels in conjunction with GWAS of lung function traits identify putatively causal relations of sRAGE to lung function? STUDY DESIGN AND METHODS Plasma sRAGE levels were measured in 6,861 Framingham Heart Study participants and GWAS of sRAGE was conducted to identify protein quantitative trait loci (pQTL), including cis-pQTL variants at the sRAGE protein-coding gene locus (AGER). We integrated sRAGE pQTL variants with variants from GWAS of lung traits. Colocalization of sRAGE pQTL variants with lung trait GWAS variants was conducted, and Mendelian randomization was performed using sRAGE cis-pQTL variants to infer causality of sRAGE for pulmonary traits. Cross-sectional and longitudinal protein-trait association analyses were conducted for sRAGE in relation to lung traits. RESULTS Colocalization identified shared genetic signals for sRAGE with lung traits. Mendelian randomization analyses suggested protective causal relations of sRAGE to several pulmonary traits. Protein-trait association analyses demonstrated higher sRAGE levels to be cross-sectionally and longitudinally associated with preserved lung function. INTERPRETATION sRAGE is produced by type I alveolar cells, and it acts as a decoy receptor to block the inflammatory cascade. Our integrative genomics approach provides evidence for sRAGE as a causal and protective biomarker of lung function, and the pattern of associations is suggestive of a protective role of sRAGE against restrictive lung physiology. We speculate that targeting the AGER/sRAGE axis may be therapeutically beneficial for the treatment and prevention of inflammation-related lung disease.
Collapse
Affiliation(s)
- Joshua Keefe
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Chen Yao
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Shih-Jen Hwang
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Paul Courchesne
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Gha Young Lee
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Joseph P. Mizgerd
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - George O’Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - George R. Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Michael H. Cho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Edwin K. Silverman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD,CORRESPONDENCE TO: Daniel Levy, MD
| |
Collapse
|
27
|
Genetic Risk and Chronic Obstructive Pulmonary Disease Independently Predict the Risk of Incident Severe COVID-19. Ann Am Thorac Soc 2022; 19:58-65. [PMID: 34242153 PMCID: PMC8787794 DOI: 10.1513/annalsats.202102-171oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rationale: Both genetic variants and chronic obstructive pulmonary disease (COPD) contribute to the risk of incident severe coronavirus disease (COVID-19). Whether genetic risk of incident severe COVID-19 is the same regardless of preexisting COPD is unknown. Objectives: In this study, we aimed to investigate the potential interaction between genetic risk and COPD in relation to severe COVID-19. Methods: We constructed a polygenic risk score for severe COVID-19 by using 112 single-nucleotide polymorphisms in 430,582 participants from the UK Biobank study. We examined the associations of genetic risk and COPD with severe COVID-19 by using logistic regression models. Results: Of 430,582 participants, 712 developed severe COVID-19 as of February 22, 2021, of whom 19.8% had preexisting COPD. Compared with participants at low genetic risk, those at intermediate genetic risk (odds ratio [OR], 1.34; 95% confidence interval [CI], 1.09-1.66) and high genetic risk (OR, 1.50; 95% CI, 1.18-1.92) had higher risk of severe COVID-19 (P for trend = 0.001), and the association was independent of COPD (P for interaction = 0.76). COPD was associated with a higher risk of incident severe COVID-19 (OR, 1.37; 95% CI, 1.12-1.67; P = 0.002). Participants at high genetic risk and with COPD had a higher risk of severe COVID-19 (OR, 2.05; 95% CI, 1.35-3.04; P < 0.001) than those at low genetic risk and without COPD. Conclusions: The polygenic risk score, which combines multiple risk alleles, can be effectively used in screening for high-risk populations of severe COVID-19. High genetic risk correlates with a higher risk of severe COVID-19, regardless of preexisting COPD.
Collapse
|
28
|
Strollo HC, Nouraie SM, Hoth KF, Riley CM, Karoleski C, Zhang Y, Hanania NA, Bowler RP, Bon J, Sciurba FC. Association of Systemic Inflammation with Depressive Symptoms in Individuals with COPD. Int J Chron Obstruct Pulmon Dis 2021; 16:2515-2522. [PMID: 34511896 PMCID: PMC8423410 DOI: 10.2147/copd.s322144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/16/2021] [Indexed: 01/02/2023] Open
Abstract
Rationale Depression is a prevalent comorbidity of chronic obstructive pulmonary disease (COPD) that, along with COPD, has been associated with inflammation. An association between inflammation and depression in COPD has not been validated in a large COPD cohort. Methods Individuals from the University of Pittsburgh SCCOR cohort and the COPDGene cohort with tobacco use history and airway obstruction (FEV1/FVC <0.7) were evaluated using the Beck Depression Inventory II (BDI-II) and the Hospital Anxiety and Depression Scale (HADS), respectively. Participants completed symptom-related questionnaires and plasma IL-6 measurements. T-test, Fisher’s Exact tests and logistic regression were used for statistical analysis. Results The SCCOR cohort included 220 obstructed participants: 44% female and 21.4% with elevated depressive symptoms. GOLD staging distribution was predominantly stage I and II. The COPDGene cohort included 745 obstructed participants: 44% female and 13.0% with elevated depressive symptoms. GOLD distribution was predominantly stage II and III. In the SCCOR cohort, correlation between IL-6 and depressive symptoms trended toward significance (p= 0.08). Multivariable modeling adjusted for FEV1, age, gender and medical comorbidities showed a significant association (OR = 1.70, 95% CI = 1.08–2.69). IL-6 was significantly associated with elevated depressive symptoms in COPDGene in both univariate (p=0.001) and multivariable modeling (OR = 1.52, 95% CI =1.13–2.04). Conclusion Elevated plasma IL-6 levels are associated with depressive symptoms in individuals with COPD independent of airflow limitation and comorbid risk factors for depression. Our results suggest that systemic inflammation may play a significant and possibly bidirectional role in depression associated with COPD.
Collapse
Affiliation(s)
- Hilary C Strollo
- University of Pittsburgh Medical Center, Department of Medicine, Department of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, PA, USA
| | - Seyed M Nouraie
- University of Pittsburgh Medical Center, Department of Medicine, Department of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, PA, USA
| | - Karin F Hoth
- University of Iowa Carver College of Medicine, Department of Psychiatry, Iowa City, IA, USA
| | - Craig M Riley
- Chester County Hospital, University of Pennsylvania Health System, West Chester, PA, USA
| | - Chad Karoleski
- University of Pittsburgh Medical Center, Department of Medicine, Department of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, PA, USA
| | - Yingze Zhang
- University of Pittsburgh Medical Center, Department of Medicine, Department of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, PA, USA
| | - Nicola A Hanania
- Baylor College of Medicine, Department of Pulmonary, Critical Care and Sleep Medicine, Houston, TX, USA
| | - Russell P Bowler
- National Jewish Health, Department of Medicine, Denver, CO, USA.,University of Colorado School of Medicine, Denver, CO, USA
| | - Jessica Bon
- University of Pittsburgh Medical Center, Department of Medicine, Department of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, PA, USA
| | - Frank C Sciurba
- University of Pittsburgh Medical Center, Department of Medicine, Department of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Li S, Zhao S, Wu Z, Wang F, Li W. Alteration of immune profiles is associated with pulmonary function and symptoms in patients with chronic obstructive pulmonary disease. Mol Med Rep 2021; 24:742. [PMID: 34435653 PMCID: PMC8430332 DOI: 10.3892/mmr.2021.12382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/10/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammation serves a key role in chronic obstructive pulmonary disease (COPD). However, changes in the immune profiles of patients with COPD remain unclear. The present prospective observational study aimed to determine the expression profiles of immune cells and inflammatory factors of patients with COPD and healthy controls, and to analyze the relationship between immune profiles and smoking history. A total of 140 subjects were enrolled in the present study between September 2018 and April 2019 at West China Hospital of Sichuan University (Chengdu, China). These included 87 patients with stable COPD and 24 patients with acute exacerbated COPD, as well as 29 healthy controls. Baseline characteristics were recorded during the screening period, and levels of immune cells were examined using flow cytometry. In addition, levels of inflammatory factors were measured using ELISAs. The results revealed increased expression of the CD64+/CD14+ mononuclear phagocyte system (MPS) and CD16+CD66+ neutrophils, and decreased expression of CD3+CD4+ T cells and CD3+ CD8+ T cells (all P<0.05) in the peripheral blood of patients with COPD and smokers relative to non-smoking controls. In addition, significant differences were observed in protein levels of IL-6, IL-1β, TNF-α, TGF-α, IFN-γ, IL-8, IL-17A and CRP among the three groups (all P<0.05). Furthermore, the IL-17A, TNF and NF-κB signaling pathways were found to serve a key role in the inflammatory network of COPD. Pearson's correlation analysis revealed a positive correlation between CD3+T lymphocyte counts and pulmonary function, and a negative correlation with smoking and exacerbations. Furthermore, neutrophils and MPS were negatively associated with pulmonary function, and IL-8 was positively associated with cough. There was also a negative association between CRP and IL-17A with pulmonary function but a positive correlation with symptoms and exacerbation. Club cell secretory protein was also negatively associated with emphysema parameters. In conclusion, the present findings revealed significant differences in profiles of immune factors among patients with COPD, smokers and non-smoking controls and their association with clinical characteristics. The clinical trial registration number of the present study is: ChiCTR1800015700 (registered with http://www.chictr.org.cn/index.aspx, 2018/04/16).
Collapse
Affiliation(s)
- Sixiang Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Zhao
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhenru Wu
- Pathology Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fangfang Wang
- Hematology Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
30
|
Yun JH, Lee C, Liu T, Liu S, Kim EY, Xu S, Curtis JL, Pinello L, Bowler RP, Silverman EK, Hersh CP, Zhou X. Hedgehog interacting protein-expressing lung fibroblasts suppress lymphocytic inflammation in mice. JCI Insight 2021; 6:e144575. [PMID: 34375314 PMCID: PMC8492352 DOI: 10.1172/jci.insight.144575] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 07/21/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly caused by cigarette smoking and characterized by chronic inflammation in vulnerable individuals. However, it is unknown how genetic factors may shape chronic inflammation in COPD. To understand how hedgehog interacting protein, encoded by HHIP gene identified in the genome-wide association study in COPD, plays a role in inflammation, we utilized Hhip+/– mice that present persistent inflammation and emphysema upon aging similar to that observed in human COPD. By performing single-cell RNA sequencing of the whole lung from mice at different ages, we found that Hhip+/– mice developed a cytotoxic immune response with a specific increase in killer cell lectin-like receptor G1–positive CD8+ T cells with upregulated Ifnγ expression recapitulating human COPD. Hhip expression was restricted to a lung fibroblast subpopulation that had increased interaction with CD8+ T lymphocytes in Hhip+/– compared with Hhip+/+ during aging. Hhip-expressing lung fibroblasts had upregulated IL-18 pathway genes in Hhip+/– lung fibroblasts, which was sufficient to drive increased levels of IFN-γ in CD8+ T cells ex vivo. Our finding provides insight into how a common genetic variation contributes to the amplified lymphocytic inflammation in COPD.
Collapse
Affiliation(s)
- Jeong H Yun
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - ChangHee Lee
- Department of Genetics, Harvard Medical School, Boston, United States of America
| | - Tao Liu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Siqi Liu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Edy Y Kim
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Shuang Xu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Jeffrey L Curtis
- VA Center, University of Michigan Medical School, Ann Arbor, United States of America
| | - Luca Pinello
- Department of Pathology, Massachusetts General Hospital, Boston, United States of America
| | - Russell P Bowler
- Department of Medicine, National Jewish Health, Denver, United States of America
| | - Edwin K Silverman
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Craig P Hersh
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Xiaobo Zhou
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| |
Collapse
|
31
|
Serban KA, Pratte KA, Bowler RP. Protein Biomarkers for COPD Outcomes. Chest 2021; 159:2244-2253. [PMID: 33434499 PMCID: PMC8213963 DOI: 10.1016/j.chest.2021.01.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/14/2020] [Accepted: 01/01/2021] [Indexed: 12/15/2022] Open
Abstract
COPD is a clinically heterogeneous syndrome characterized by injury to airways, airspaces, and lung vasculature and usually caused by tobacco smoke and/or air pollution exposure. COPD is also independently associated with nonpulmonary comorbidities (eg, cardiovascular disease) and malignancies (eg, GI, bladder), suggesting a role for systemic injury. Since not all those with exposure develop COPD, there has been a search for plasma and lung biomarkers that confer increased cross-sectional and longitudinal risk. This search typically focuses on clinically relevant COPD outcomes such as FEV1, FEV1 decline, CT measurements of emphysema, or exacerbation frequency. The rapid advances in omics technology and the molecular phenotyping of COPD cohorts now permit large-scale evaluation of genetic, transcriptomic, proteomic, and metabolic biomarkers. This review focuses on protein biomarkers associated with clinically relevant COPD outcomes. The prototypic COPD protein biomarker is alpha-1 antitrypsin; however, this biomarker only accounts for 1% to 5% of COPD. This article reviews and summarizes the evidence for other validated biomarkers for each COPD outcome, and discusses their advantages, weaknesses, and required regulatory steps to move the biomarker from the bench into clinic. Although we highlight the emergence of many novel biomarkers (eg, fibrinogen, soluble receptor for advanced glycation, surfactant protein D, club cell secretory protein), there is increasing evidence that individual biomarkers only explain a fraction of the increased COPD risk and that multiple biomarker panels are needed to completely explain clinical variation and risk in individuals and populations.
Collapse
Affiliation(s)
- Karina A Serban
- National Jewish Health, Denver; University of Colorado, Anschutz Medical Campus, Aurora, CO.
| | | | - Russell P Bowler
- National Jewish Health, Denver; University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
32
|
Zandstra J, Jongerius I, Kuijpers TW. Future Biomarkers for Infection and Inflammation in Febrile Children. Front Immunol 2021; 12:631308. [PMID: 34079538 PMCID: PMC8165271 DOI: 10.3389/fimmu.2021.631308] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
Febrile patients, suffering from an infection, inflammatory disease or autoimmunity may present with similar or overlapping clinical symptoms, which makes early diagnosis difficult. Therefore, biomarkers are needed to help physicians form a correct diagnosis and initiate the right treatment to improve patient outcomes following first presentation or admittance to hospital. Here, we review the landscape of novel biomarkers and approaches of biomarker discovery. We first discuss the use of current plasma parameters and whole blood biomarkers, including results obtained by RNA profiling and mass spectrometry, to discriminate between bacterial and viral infections. Next we expand upon the use of biomarkers to distinguish between infectious and non-infectious disease. Finally, we discuss the strengths as well as the potential pitfalls of current developments. We conclude that the use of combination tests, using either protein markers or transcriptomic analysis, have advanced considerably and should be further explored to improve current diagnostics regarding febrile infections and inflammation. If proven effective when combined, these biomarker signatures will greatly accelerate early and tailored treatment decisions.
Collapse
Affiliation(s)
- Judith Zandstra
- Division Research and Landsteiner Laboratory, Department of Immunopathology, Sanquin Blood Supply, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Ilse Jongerius
- Division Research and Landsteiner Laboratory, Department of Immunopathology, Sanquin Blood Supply, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
- Division Research and Landsteiner Laboratory, Department of Blood Cell Research, Sanquin Blood Supply, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
33
|
Luebbering N, Abdullah S, Lounder D, Lane A, Dole N, Rubinstein J, Hewison M, Gloude N, Jodele S, Perentesis KMR, Lake K, Litts B, Duell A, Dandoy CE, Davies SM. Endothelial injury, F-actin and vitamin-D binding protein after hematopoietic stem cell transplant and association with clinical outcomes. Haematologica 2021; 106:1321-1329. [PMID: 32241849 PMCID: PMC8094097 DOI: 10.3324/haematol.2019.233478] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Indexed: 01/22/2023] Open
Abstract
Endothelial injury after hematopoietic stem cell transplant is an important initiating factor for early transplant toxicities of thrombotic microangiopathy and acute graft versus host disease. We hypothesized that release of the angiopathic molecule filamentous actin (F-actin) from hematopoietic cells lysed during conditioning prior to stem cell transplant would be associated with clinical outcomes. We detected F-actin in the blood of 52% of stem cell transplant recipients in the first 14 days after transplant, and children with detectable F-actin had a significantly elevated risk of thrombotic microangiopathy (P=0.03) and non-relapse mortality (P=0.04). F-actin is cleared from the circulation by vitamin D binding protein (VDBP) so we expected that higher levels of VDBP would improve outcomes. In a cohort of 190 children receiving an allogeneic transplant, risk of thrombotic microangiopathy was reduced in those with serum concentrations of VDBP above the median at day 30 (10% vs. 31%, P=0.01), and graft versus host disease and non-relapse mortality were reduced in those with levels above the median at day 100 (3% vs. 18%, P=0.04 and 0% vs. 15%, P=0.002). Western blot analyses demonstrated actin-VDBP complexes in the blood, which cleared by day 21-28. Our data support modulation of cytokine secretion and macrophage phenotype by VDBP later after transplant. Taken together, our data identify an association between Factin, a mediator of endothelial damage, and VDBP, an actin scavenger, as modifiers of risk of clinical consequences of endothelial injury.
Collapse
Affiliation(s)
- Nathan Luebbering
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sheyar Abdullah
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dana Lounder
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam Lane
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nikhil Dole
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeremy Rubinstein
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Martin Hewison
- School of Clinical and Experimental Medicine, University of, University of Birmingham, UK
| | - Nicholas Gloude
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sonata Jodele
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kitty M R Perentesis
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kelly Lake
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bridget Litts
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexandra Duell
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christopher E Dandoy
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Stella M Davies
- Department of Pediatric, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
34
|
Hormozdiari F, Jung J, Eskin E, J. Joo JW. MARS: leveraging allelic heterogeneity to increase power of association testing. Genome Biol 2021; 22:128. [PMID: 33931127 PMCID: PMC8086090 DOI: 10.1186/s13059-021-02353-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 04/15/2021] [Indexed: 11/10/2022] Open
Abstract
In standard genome-wide association studies (GWAS), the standard association test is underpowered to detect associations between loci with multiple causal variants with small effect sizes. We propose a statistical method, Model-based Association test Reflecting causal Status (MARS), that finds associations between variants in risk loci and a phenotype, considering the causal status of variants, only requiring the existing summary statistics to detect associated risk loci. Utilizing extensive simulated data and real data, we show that MARS increases the power of detecting true associated risk loci compared to previous approaches that consider multiple variants, while controlling the type I error.
Collapse
Affiliation(s)
- Farhad Hormozdiari
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115 MA USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Junghyun Jung
- Department of Life Science, Dongguk University-Seoul, Seoul, 04620 South Korea
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, Los Angeles, 90095 CA USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, 90095 CA USA
| | - Jong Wha J. Joo
- Department of Computer Science and Engineering, Dongguk University-Seoul, Seoul, 04620 South Korea
| |
Collapse
|
35
|
Pratte KA, Curtis JL, Kechris K, Couper D, Cho MH, Silverman EK, DeMeo DL, Sciurba FC, Zhang Y, Ortega VE, O’Neal WK, Gillenwater LA, Lynch DA, Hoffman EA, Newell JD, Comellas AP, Castaldi PJ, Miller BE, Pouwels SD, Hacken NHTT, Bischoff R, Klont F, Woodruff PG, Paine R, Barr RG, Hoidal J, Doerschuk CM, Charbonnier JP, Sung R, Locantore N, Yonchuk JG, Jacobson S, Tal-singer R, Merrill D, Bowler RP. Soluble receptor for advanced glycation end products (sRAGE) as a biomarker of COPD. Respir Res 2021; 22:127. [PMID: 33906653 PMCID: PMC8076883 DOI: 10.1186/s12931-021-01686-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/16/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Soluble receptor for advanced glycation end products (sRAGE) is a proposed emphysema and airflow obstruction biomarker; however, previous publications have shown inconsistent associations and only one study has investigate the association between sRAGE and emphysema. No cohorts have examined the association between sRAGE and progressive decline of lung function. There have also been no evaluation of assay compatibility, receiver operating characteristics, and little examination of the effect of genetic variability in non-white population. This manuscript addresses these deficiencies and introduces novel data from Pittsburgh COPD SCCOR and as well as novel work on airflow obstruction. A meta-analysis is used to quantify sRAGE associations with clinical phenotypes. METHODS sRAGE was measured in four independent longitudinal cohorts on different analytic assays: COPDGene (n = 1443); SPIROMICS (n = 1623); ECLIPSE (n = 2349); Pittsburgh COPD SCCOR (n = 399). We constructed adjusted linear mixed models to determine associations of sRAGE with baseline and follow up forced expiratory volume at one second (FEV1) and emphysema by quantitative high-resolution CT lung density at the 15th percentile (adjusted for total lung capacity). RESULTS Lower plasma or serum sRAGE values were associated with a COPD diagnosis (P < 0.001), reduced FEV1 (P < 0.001), and emphysema severity (P < 0.001). In an inverse-variance weighted meta-analysis, one SD lower log10-transformed sRAGE was associated with 105 ± 22 mL lower FEV1 and 4.14 ± 0.55 g/L lower adjusted lung density. After adjusting for covariates, lower sRAGE at baseline was associated with greater FEV1 decline and emphysema progression only in the ECLIPSE cohort. Non-Hispanic white subjects carrying the rs2070600 minor allele (A) and non-Hispanic African Americans carrying the rs2071288 minor allele (A) had lower sRAGE measurements compare to those with the major allele, but their emphysema-sRAGE regression slopes were similar. CONCLUSIONS Lower blood sRAGE is associated with more severe airflow obstruction and emphysema, but associations with progression are inconsistent in the cohorts analyzed. In these cohorts, genotype influenced sRAGE measurements and strengthened variance modelling. Thus, genotype should be included in sRAGE evaluations.
Collapse
Affiliation(s)
| | - Jeffrey L. Curtis
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI USA
- Medical Service, Ann Arbor Healthcare System, Ann Arbor, MI USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, School of Public Health, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO USA
| | - David Couper
- Department of Biostatistics, Collaborative Studies Coordinating Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Dawn L. DeMeo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Frank C. Sciurba
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Victor E. Ortega
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Wanda K. O’Neal
- Marsico Lung Institute (CF Research Center), University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Lucas A. Gillenwater
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206 USA
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - David A. Lynch
- Department of Radiology, National Jewish Health, Denver, CO USA
| | - Eric A. Hoffman
- Department of Radiology and Biomedical Engineering, University of Iowa, Iowa City, IA USA
| | - John D. Newell
- Department of Radiology and Biomedical Engineering, University of Iowa, Iowa City, IA USA
| | - Alejandro P. Comellas
- Department of Internal Medicine, College of Medicine, University of Iowa Carver, Iowa City, IA USA
| | - Peter J. Castaldi
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | | | - Simon D. Pouwels
- Department of Pathology and Medical Biology, University of Groningen, Groningen, Netherlands
| | - Nick H. T. ten Hacken
- Department of Pathology and Medical Biology, University of Groningen, Groningen, Netherlands
| | - Rainer Bischoff
- Department of
Analytical Biochemistry, University of Groningen, Groningen, Netherlands
| | - Frank Klont
- Department of
Analytical Biochemistry, University of Groningen, Groningen, Netherlands
| | - Prescott G. Woodruff
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of California-San Francisco, San Francisco, CA USA
- Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA USA
| | - Robert Paine
- Division of Pulmonary and Critical Care, University of Utah, Salt Lake City, UT USA
| | - R. Graham Barr
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Columbia University, New York, NY USA
| | - John Hoidal
- Division of Pulmonary and Critical Care, University of Utah, Salt Lake City, UT USA
| | - Claire M. Doerschuk
- Marsico Lung Institute (CF Research Center), University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | | | - Ruby Sung
- Research and Development, GlaxoSmithKline, Collegeville, PA USA
| | | | - John G. Yonchuk
- Research and Development, GlaxoSmithKline, Collegeville, PA USA
| | - Sean Jacobson
- Department of Genetics, National Jewish Health, Denver, CO USA
| | | | | | - Russell P. Bowler
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, CO 80206 USA
| |
Collapse
|
36
|
Reilly JP, Meyer NJ, Shashaty MG, Anderson BJ, Ittner C, Dunn TG, Lim B, Forker C, Bonk MP, Kotloff E, Feng R, Cantu E, Mangalmurti NS, Calfee CS, Matthay MA, Mikacenic C, Walley KR, Russell J, Christiani DC, Wurfel MM, Lanken PN, Reilly MP, Christie JD. The ABO histo-blood group, endothelial activation, and acute respiratory distress syndrome risk in critical illness. J Clin Invest 2021; 131:139700. [PMID: 32931480 PMCID: PMC7773362 DOI: 10.1172/jci139700] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUNDThe ABO histo-blood group is defined by carbohydrate modifications and is associated with risk for multiple diseases, including acute respiratory distress syndrome (ARDS). We hypothesized that genetically determined blood subtype A1 is associated with increased risk of ARDS and markers of microvascular dysfunction and coagulation.METHODSWe conducted analyses in 3 cohorts of critically ill trauma and sepsis patients (n = 3710) genotyped on genome-wide platforms to determine the association of the A1 blood type genotype with ARDS risk. We subsequently determined whether associations were present in FUT2-defined nonsecretors who lack ABO antigens on epithelium, but not endothelium. In a patient subgroup, we determined the associations of blood type with plasma levels of endothelial glycoproteins and disseminated intravascular coagulation (DIC). Lastly, we tested whether blood type A was associated with less donor lung injury recovery during human ex vivo lung perfusion (EVLP).RESULTSThe A1 genotype was associated with a higher risk of moderate to severe ARDS relative to type O in all 3 populations. In sepsis, this relationship was strongest in nonpulmonary infections. The association persisted in nonsecretors, suggesting a vascular mechanism. The A1 genotype was also associated with higher DIC risk as well as concentrations of thrombomodulin and von Willebrand factor, which in turn were associated with ARDS risk. Blood type A was also associated with less lung injury recovery during EVLP.CONCLUSIONWe identified a replicable association between ABO blood type A1 and risk of ARDS among the critically ill, possibly mediated through microvascular dysfunction and coagulation.FUNDINGNIH HL122075, HL125723, HL137006, HL137915, DK097307, HL115354, HL101779, and the University of Pennsylvania McCabe Fund Fellowship Award.
Collapse
Affiliation(s)
- John P. Reilly
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
| | - Michael G.S. Shashaty
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
- Center for Clinical Epidemiology and Biostatics, and
| | - Brian J. Anderson
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
| | | | - Thomas G. Dunn
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
| | - Brian Lim
- Division of Pulmonary, Allergy, and Critical Care
| | | | | | | | - Rui Feng
- Center for Clinical Epidemiology and Biostatics, and
| | - Edward Cantu
- Center for Translational Lung Biology
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nilam S. Mangalmurti
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
| | - Carolyn S. Calfee
- Department of Medicine and
- Department of Anesthesia and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Michael A. Matthay
- Department of Medicine and
- Department of Anesthesia and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Carmen Mikacenic
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - Keith R. Walley
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - James Russell
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - David C. Christiani
- T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Mark M. Wurfel
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | | | - Muredach P. Reilly
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care
- Center for Translational Lung Biology
- Center for Clinical Epidemiology and Biostatics, and
| |
Collapse
|
37
|
Proteome-wide Systems Genetics to Identify Functional Regulators of Complex Traits. Cell Syst 2021; 12:5-22. [PMID: 33476553 DOI: 10.1016/j.cels.2020.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/15/2020] [Accepted: 10/07/2020] [Indexed: 02/08/2023]
Abstract
Proteomic technologies now enable the rapid quantification of thousands of proteins across genetically diverse samples. Integration of these data with systems-genetics analyses is a powerful approach to identify new regulators of economically important or disease-relevant phenotypes in various populations. In this review, we summarize the latest proteomic technologies and discuss technical challenges for their use in population studies. We demonstrate how the analysis of correlation structure and loci mapping can be used to identify genetic factors regulating functional protein networks and complex traits. Finally, we provide an extensive summary of the use of proteome-wide systems genetics throughout fungi, plant, and animal kingdoms and discuss the power of this approach to identify candidate regulators and drug targets in large human consortium studies.
Collapse
|
38
|
Integration of SNP Disease Association, eQTL, and Enrichment Analyses to Identify Risk SNPs and Susceptibility Genes in Chronic Obstructive Pulmonary Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3854196. [PMID: 33457407 PMCID: PMC7785362 DOI: 10.1155/2020/3854196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex disease caused by the disturbance of genetic and environmental factors. Single-nucleotide polymorphisms (SNPs) play a vital role in the genetic dissection of complex diseases. In-depth analysis of SNP-related information could recognize disease-associated biomarkers and further uncover the genetic mechanism of complex diseases. Risk-related variants might act on the disease by affecting gene expression and gene function. Through integrating SNP disease association study and expression quantitative trait loci (eQTL) analysis, as well as functional enrichment of containing known causal genes, four risk SNPs and four corresponding susceptibility genes were identified utilizing next-generation sequencing (NGS) data of COPD. Of the four risk SNPs, one could be found in the SNPedia database that stored disease-related SNPs and has been linked to a disease in the literature. Four genes showed significant differences from the perspective of normal/disease or variant/nonvariant samples, as well as the high performance of sample classification. It is speculated that the four susceptibility genes could be used as biomarkers of COPD. Furthermore, three of our susceptibility genes have been confirmed in the literature to be associated with COPD. Among them, two genes had an impact on the significance of expression correlation of known causal genes they interact with, respectively. Overall, this research may present novel insights into the diagnosis and pathogenesis of COPD and susceptibility gene identification of other complex diseases.
Collapse
|
39
|
Gillenwater LA, Pratte KA, Hobbs BD, Cho MH, Zhuang Y, Halper-Stromberg E, Cruickshank-Quinn C, Reisdorph N, Petrache I, Labaki WW, O'Neal WK, Ortega VE, Jones DP, Uppal K, Jacobson S, Michelotti G, Wendt CH, Kechris KJ, Bowler RP. Plasma Metabolomic Signatures of Chronic Obstructive Pulmonary Disease and the Impact of Genetic Variants on Phenotype-Driven Modules. NETWORK AND SYSTEMS MEDICINE 2020; 3:159-181. [PMID: 33987620 PMCID: PMC8109053 DOI: 10.1089/nsm.2020.0009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Small studies have recently suggested that there are specific plasma metabolic signatures in chronic obstructive pulmonary disease (COPD), but there have been no large comprehensive study of metabolomic signatures in COPD that also integrate genetic variants. Materials and Methods: Fresh frozen plasma from 957 non-Hispanic white subjects in COPDGene was used to quantify 995 metabolites with Metabolon's global metabolomics platform. Metabolite associations with five COPD phenotypes (chronic bronchitis, exacerbation frequency, percent emphysema, post-bronchodilator forced expiratory volume at one second [FEV1]/forced vital capacity [FVC], and FEV1 percent predicted) were assessed. A metabolome-wide association study was performed to find genetic associations with metabolite levels. Significantly associated single-nucleotide polymorphisms were tested for replication with independent metabolomic platforms and independent cohorts. COPD phenotype-driven modules were identified in network analysis integrated with genetic associations to assess gene-metabolite-phenotype interactions. Results: Of metabolites tested, 147 (14.8%) were significantly associated with at least 1 COPD phenotype. Associations with airflow obstruction were enriched for diacylglycerols and branched chain amino acids. Genetic associations were observed with 109 (11%) metabolites, 72 (66%) of which replicated in an independent cohort. For 20 metabolites, more than 20% of variance was explained by genetics. A sparse network of COPD phenotype-driven modules was identified, often containing metabolites missed in previous testing. Of the 26 COPD phenotype-driven modules, 6 contained metabolites with significant met-QTLs, although little module variance was explained by genetics. Conclusion: A dysregulation of systemic metabolism was predominantly found in COPD phenotypes characterized by airflow obstruction, where we identified robust heritable effects on individual metabolite abundances. However, network analysis, which increased the statistical power to detect associations missed previously in classic regression analyses, revealed that the genetic influence on COPD phenotype-driven metabolomic modules was modest when compared with clinical and environmental factors.
Collapse
Affiliation(s)
| | | | - Brian D. Hobbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yonghua Zhuang
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | | | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Irina Petrache
- National Jewish Health, Denver, Colorado, USA
- School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Wanda K. O'Neal
- Lung Institute/Cystic Fibrosis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victor E. Ortega
- Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Dean P. Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | | | | | - Christine H. Wendt
- Department of Medicine, University of Minnesota and the VAMC, Minneapolis, Minnesota, USA
| | - Katerina J. Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Russell P. Bowler
- National Jewish Health, Denver, Colorado, USA
- School of Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
40
|
Zhang W, Deng X, Tang R, Wang H. Receptor for advanced glycation end-product rs1800624 polymorphism contributes to increase breast cancer risk: Evidence from a meta-analysis. Medicine (Baltimore) 2020; 99:e22775. [PMID: 33126315 PMCID: PMC7598831 DOI: 10.1097/md.0000000000022775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although several studies have identified an association between the receptor for advanced glycation end-product (RAGE) rs1800624 polymorphism and breast cancer, the results have been conflicting. Therefore, we conducted a meta-analysis to assess the relationship between the RAGE rs1800624 polymorphism and breast cancer risk. METHODS Studies were searched in the PubMed, Web of Science, Embase, Wanfang Med Online, and China National Knowledge Infrastructure databases until September 20, 2019 to identify all potential literature on this association. Fixed-effect or random-effect models were used to calculate odds ratios (ORs) and their corresponding 95% confidence intervals (95% CIs). Subgroup and sensitivity analyses and tests for publication bias were also performed. RESULTS Five eligible studies involving 2823 subjects (1410 patients and 1413 healthy controls) were included in the current meta-analysis. The pooled analysis indicated a positive correlation between the RAGE rs1800624 polymorphism and the risk of breast cancer in a homozygous genetic model (OR = 1.423, 95% CI = 1.043-1.941, P = .026). Ethnicity-based subgroup analysis demonstrated that RAGE rs1800624 polymorphism may increase the risk of breast cancer in the Asian population in homozygous model (OR = 1.661, 95% CI = 1.178-2.342, P = .004). CONCLUSION The RAGE rs1800624 polymorphism may increase the risk of breast cancer in the homozygous genetic model, especially in Asian populations. Large-scale and well-designed studies are needed in different populations to further evaluate the role of the RAGE polymorphism in breast cancer.
Collapse
Affiliation(s)
| | | | - Ruijun Tang
- Department of Pathology, Guilin TCM Hospital Affiliated to Guangxi University of Chinese Medicine, Guangxi, China
| | | |
Collapse
|
41
|
Balnis J, Vincent CE, Jones AJ, Drake LA, Coon JJ, Lee CG, Elias JA, Singer HA, Jaitovich A. Established Biomarkers of Chronic Obstructive Pulmonary Disease Reflect Skeletal Muscle Integrity's Response to Exercise in an Animal Model of Pulmonary Emphysema. Am J Respir Cell Mol Biol 2020; 63:266-269. [PMID: 32735164 DOI: 10.1165/rcmb.2019-0439le] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
42
|
Lin DY, Zeng D, Couper D. A general framework for integrative analysis of incomplete multiomics data. Genet Epidemiol 2020; 44:646-664. [PMID: 32691502 PMCID: PMC7951090 DOI: 10.1002/gepi.22328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/05/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022]
Abstract
There is a tremendous current interest in measuring multiple types of omics features (e.g., DNA sequences, RNA expressions, methylation profiles, metabolic profiles, protein expressions) on a large number of subjects. Although genotypes are typically available for all study subjects, other data types may be measured only on a subset of subjects due to cost or other constraints. In addition, quantitative omics measurements, such as metabolite levels and protein expressions, are subject to detection limits in that the measurements below (or above) certain thresholds are not detectable. In this article, we propose a rigorous and powerful approach to handle missing values and detection limits in integrative analysis of multiomics data. We relate quantitative omics variables to genetic variants and other variables through linear regression models and relate phenotypes to quantitative omics variables and other variables through generalized linear models. We derive the joint-likelihood for the two sets of models by allowing arbitrary patterns of missing values and detection limits for quantitative omics variables. We carry out maximum-likelihood estimation through computationally fast and stable algorithms. The resulting estimators are approximately unbiased and statistically efficient. An application to a major study on chronic obstructive lung disease yielded new biological insights.
Collapse
Affiliation(s)
- Dan-Yu Lin
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Donglin Zeng
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - David Couper
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
43
|
Folkersen L, Gustafsson S, Wang Q, Hansen DH, Hedman ÅK, Schork A, Page K, Zhernakova DV, Wu Y, Peters J, Eriksson N, Bergen SE, Boutin TS, Bretherick AD, Enroth S, Kalnapenkis A, Gådin JR, Suur BE, Chen Y, Matic L, Gale JD, Lee J, Zhang W, Quazi A, Ala-Korpela M, Choi SH, Claringbould A, Danesh J, Davey Smith G, de Masi F, Elmståhl S, Engström G, Fauman E, Fernandez C, Franke L, Franks PW, Giedraitis V, Haley C, Hamsten A, Ingason A, Johansson Å, Joshi PK, Lind L, Lindgren CM, Lubitz S, Palmer T, Macdonald-Dunlop E, Magnusson M, Melander O, Michaelsson K, Morris AP, Mägi R, Nagle MW, Nilsson PM, Nilsson J, Orho-Melander M, Polasek O, Prins B, Pålsson E, Qi T, Sjögren M, Sundström J, Surendran P, Võsa U, Werge T, Wernersson R, Westra HJ, Yang J, Zhernakova A, Ärnlöv J, Fu J, Smith JG, Esko T, Hayward C, Gyllensten U, Landen M, Siegbahn A, Wilson JF, Wallentin L, Butterworth AS, Holmes MV, Ingelsson E, Mälarstig A. Genomic and drug target evaluation of 90 cardiovascular proteins in 30,931 individuals. Nat Metab 2020; 2:1135-1148. [PMID: 33067605 PMCID: PMC7611474 DOI: 10.1038/s42255-020-00287-2] [Citation(s) in RCA: 398] [Impact Index Per Article: 79.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 09/02/2020] [Indexed: 02/02/2023]
Abstract
Circulating proteins are vital in human health and disease and are frequently used as biomarkers for clinical decision-making or as targets for pharmacological intervention. Here, we map and replicate protein quantitative trait loci (pQTL) for 90 cardiovascular proteins in over 30,000 individuals, resulting in 451 pQTLs for 85 proteins. For each protein, we further perform pathway mapping to obtain trans-pQTL gene and regulatory designations. We substantiate these regulatory findings with orthogonal evidence for trans-pQTLs using mouse knockdown experiments (ABCA1 and TRIB1) and clinical trial results (chemokine receptors CCR2 and CCR5), with consistent regulation. Finally, we evaluate known drug targets, and suggest new target candidates or repositioning opportunities using Mendelian randomization. This identifies 11 proteins with causal evidence of involvement in human disease that have not previously been targeted, including EGF, IL-16, PAPPA, SPON1, F3, ADM, CASP-8, CHI3L1, CXCL16, GDF15 and MMP-12. Taken together, these findings demonstrate the utility of large-scale mapping of the genetics of the proteome and provide a resource for future precision studies of circulating proteins in human health.
Collapse
Affiliation(s)
- Lasse Folkersen
- Department of Medicine, Karolinska Institute, Solna, Sweden
- Danish National Genome Center, Copenhagen, Denmark
- SCALLOP consortium
| | - Stefan Gustafsson
- SCALLOP consortium
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Qin Wang
- SCALLOP consortium
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | | | - Åsa K Hedman
- Department of Medicine, Karolinska Institute, Solna, Sweden
- SCALLOP consortium
- Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Andrew Schork
- SCALLOP consortium
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Capital Region, Roskilde, Denmark
- Neurogenomics Division, The Translational Genomics Research Institute (TGEN), Phoenix, AZ, USA
| | - Karen Page
- SCALLOP consortium
- Early Clinical Development, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Daria V Zhernakova
- SCALLOP consortium
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Yang Wu
- SCALLOP consortium
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - James Peters
- SCALLOP consortium
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| | - Niclas Eriksson
- SCALLOP consortium
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Sarah E Bergen
- SCALLOP consortium
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Thibaud S Boutin
- SCALLOP consortium
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland
| | - Andrew D Bretherick
- SCALLOP consortium
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland
| | - Stefan Enroth
- SCALLOP consortium
- Department of Immunology, Genetics, and Pathology, Biomedical Center, Science for Life Laboratory (SciLifeLab) Uppsala University, Uppsala, Sweden
| | - Anette Kalnapenkis
- SCALLOP consortium
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Jesper R Gådin
- Department of Medicine, Karolinska Institute, Solna, Sweden
- SCALLOP consortium
| | - Bianca E Suur
- SCALLOP consortium
- Department of Molecular Medicine and Surgery, Karolinska Institute, Solna, Sweden
| | - Yan Chen
- Department of Medicine, Karolinska Institute, Solna, Sweden
- SCALLOP consortium
| | - Ljubica Matic
- SCALLOP consortium
- Department of Molecular Medicine and Surgery, Karolinska Institute, Solna, Sweden
| | - Jeremy D Gale
- SCALLOP consortium
- Inflammation and Immunology Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Julie Lee
- SCALLOP consortium
- Early Clinical Development, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Weidong Zhang
- SCALLOP consortium
- Pfizer Global Product Development, Cambridge, MA, USA
| | - Amira Quazi
- SCALLOP consortium
- Early Clinical Development, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Mika Ala-Korpela
- SCALLOP consortium
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Seung Hoan Choi
- SCALLOP consortium
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Annique Claringbould
- SCALLOP consortium
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - John Danesh
- SCALLOP consortium
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - George Davey Smith
- SCALLOP consortium
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | | | - Sölve Elmståhl
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Gunnar Engström
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Eric Fauman
- SCALLOP consortium
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Celine Fernandez
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Lude Franke
- SCALLOP consortium
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Paul W Franks
- SCALLOP consortium
- Department of Clinical Sciences, Lund University Diabetes Center, Malmö, Sweden
| | - Vilmantas Giedraitis
- SCALLOP consortium
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Chris Haley
- SCALLOP consortium
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland
| | - Anders Hamsten
- Department of Medicine, Karolinska Institute, Solna, Sweden
- SCALLOP consortium
| | - Andres Ingason
- SCALLOP consortium
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Capital Region, Roskilde, Denmark
| | - Åsa Johansson
- SCALLOP consortium
- Department of Immunology, Genetics, and Pathology, Biomedical Center, Science for Life Laboratory (SciLifeLab) Uppsala University, Uppsala, Sweden
| | - Peter K Joshi
- SCALLOP consortium
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Lars Lind
- SCALLOP consortium
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cecilia M Lindgren
- SCALLOP consortium
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Steven Lubitz
- SCALLOP consortium
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Tom Palmer
- SCALLOP consortium
- Department of Mathematics and Statistics, University of Lancaster, Lancaster, UK
| | - Erin Macdonald-Dunlop
- SCALLOP consortium
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Martin Magnusson
- SCALLOP consortium
- Department of Cardiology, Skåne University Hospital Malmö, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- North-West University, Hypertension in Africa Research Team (HART), Potchefstroom, South Africa
| | - Olle Melander
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Karl Michaelsson
- SCALLOP consortium
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Andrew P Morris
- SCALLOP consortium
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Reedik Mägi
- SCALLOP consortium
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Michael W Nagle
- SCALLOP consortium
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Peter M Nilsson
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Jan Nilsson
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Marju Orho-Melander
- SCALLOP consortium
- Department of Clinical Sciences, Clinical Research Center, Lund University, Malmö, Sweden
| | - Ozren Polasek
- SCALLOP consortium
- Faculty of Medicine, University of Split, Split, Croatia
| | - Bram Prins
- SCALLOP consortium
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Erik Pålsson
- SCALLOP consortium
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ting Qi
- SCALLOP consortium
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Marketa Sjögren
- SCALLOP consortium
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Johan Sundström
- SCALLOP consortium
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Praveen Surendran
- SCALLOP consortium
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Urmo Võsa
- SCALLOP consortium
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Thomas Werge
- SCALLOP consortium
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Capital Region, Roskilde, Denmark
| | | | - Harm-Jan Westra
- SCALLOP consortium
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jian Yang
- SCALLOP consortium
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Alexandra Zhernakova
- SCALLOP consortium
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Johan Ärnlöv
- SCALLOP consortium
- Department of Neurobiology, Care Sciences and Society (NVS) Division of Family Medicine and Primary Care, Karolinska Institute, Solna, Sweden
| | - Jingyuan Fu
- SCALLOP consortium
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Paediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - J Gustav Smith
- SCALLOP consortium
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Tõnu Esko
- SCALLOP consortium
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Caroline Hayward
- SCALLOP consortium
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland
| | - Ulf Gyllensten
- SCALLOP consortium
- Department of Immunology, Genetics, and Pathology, Biomedical Center, Science for Life Laboratory (SciLifeLab) Uppsala University, Uppsala, Sweden
| | - Mikael Landen
- SCALLOP consortium
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Agneta Siegbahn
- SCALLOP consortium
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - James F Wilson
- SCALLOP consortium
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Lars Wallentin
- SCALLOP consortium
- Department of Medical Sciences, Cardiology and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Adam S Butterworth
- SCALLOP consortium
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Michael V Holmes
- SCALLOP consortium
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
| | - Erik Ingelsson
- SCALLOP consortium
- Department of Medicine, Division of Cardiovascular Medicine, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Anders Mälarstig
- Department of Medicine, Karolinska Institute, Solna, Sweden.
- SCALLOP consortium, .
- Emerging Science & Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA.
| |
Collapse
|
44
|
Genetics meets proteomics: perspectives for large population-based studies. Nat Rev Genet 2020; 22:19-37. [PMID: 32860016 DOI: 10.1038/s41576-020-0268-2] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 12/22/2022]
Abstract
Proteomic analysis of cells, tissues and body fluids has generated valuable insights into the complex processes influencing human biology. Proteins represent intermediate phenotypes for disease and provide insight into how genetic and non-genetic risk factors are mechanistically linked to clinical outcomes. Associations between protein levels and DNA sequence variants that colocalize with risk alleles for common diseases can expose disease-associated pathways, revealing novel drug targets and translational biomarkers. However, genome-wide, population-scale analyses of proteomic data are only now emerging. Here, we review current findings from studies of the plasma proteome and discuss their potential for advancing biomedical translation through the interpretation of genome-wide association analyses. We highlight the challenges faced by currently available technologies and provide perspectives relevant to their future application in large-scale biobank studies.
Collapse
|
45
|
He B, Shi J, Wang X, Jiang H, Zhu HJ. Genome-wide pQTL analysis of protein expression regulatory networks in the human liver. BMC Biol 2020; 18:97. [PMID: 32778093 PMCID: PMC7418398 DOI: 10.1186/s12915-020-00830-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 07/16/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Previous expression quantitative trait loci (eQTL) studies have identified thousands of genetic variants to be associated with gene expression at the mRNA level in the human liver. However, protein expression often correlates poorly with mRNA levels. Thus, protein quantitative trait loci (pQTL) study is required to identify genetic variants that regulate protein expression in human livers. RESULTS We conducted a genome-wide pQTL study in 287 normal human liver samples and identified 900 local pQTL variants and 4026 distant pQTL variants. We further discovered 53 genome hotspots of pQTL variants. Transcriptional region mapping analysis showed that 1133 pQTL variants are in transcriptional regulatory regions. Genomic region enrichment analysis of the identified pQTL variants revealed 804 potential regulatory interactions among 595 predicted regulators (e.g., non-coding RNAs) and 394 proteins. Moreover, pQTL variants and trait-variant integration analysis implied several novel mechanisms underlying the relationships between protein expression and liver diseases, such as alcohol dependence. Notably, over 2000 of the identified pQTL variants have not been reported in previous eQTL studies, suggesting extensive involvement of genetic polymorphisms in post-transcriptional regulation of protein expression in human livers. CONCLUSIONS We have partially established protein expression regulation networks in human livers and generated a wealth of pQTL data that could serve as a valuable resource for the scientific community.
Collapse
Affiliation(s)
- Bing He
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, MI, 48109-1065, USA
| | - Jian Shi
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, MI, 48109-1065, USA
| | - Xinwen Wang
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, MI, 48109-1065, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, MI, 48109-1065, USA.
| |
Collapse
|
46
|
Hillary RF, Trejo-Banos D, Kousathanas A, McCartney DL, Harris SE, Stevenson AJ, Patxot M, Ojavee SE, Zhang Q, Liewald DC, Ritchie CW, Evans KL, Tucker-Drob EM, Wray NR, McRae AF, Visscher PM, Deary IJ, Robinson MR, Marioni RE. Multi-method genome- and epigenome-wide studies of inflammatory protein levels in healthy older adults. Genome Med 2020; 12:60. [PMID: 32641083 PMCID: PMC7346642 DOI: 10.1186/s13073-020-00754-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The molecular factors which control circulating levels of inflammatory proteins are not well understood. Furthermore, association studies between molecular probes and human traits are often performed by linear model-based methods which may fail to account for complex structure and interrelationships within molecular datasets. METHODS In this study, we perform genome- and epigenome-wide association studies (GWAS/EWAS) on the levels of 70 plasma-derived inflammatory protein biomarkers in healthy older adults (Lothian Birth Cohort 1936; n = 876; Olink® inflammation panel). We employ a Bayesian framework (BayesR+) which can account for issues pertaining to data structure and unknown confounding variables (with sensitivity analyses using ordinary least squares- (OLS) and mixed model-based approaches). RESULTS We identified 13 SNPs associated with 13 proteins (n = 1 SNP each) concordant across OLS and Bayesian methods. We identified 3 CpG sites spread across 3 proteins (n = 1 CpG each) that were concordant across OLS, mixed-model and Bayesian analyses. Tagged genetic variants accounted for up to 45% of variance in protein levels (for MCP2, 36% of variance alone attributable to 1 polymorphism). Methylation data accounted for up to 46% of variation in protein levels (for CXCL10). Up to 66% of variation in protein levels (for VEGFA) was explained using genetic and epigenetic data combined. We demonstrated putative causal relationships between CD6 and IL18R1 with inflammatory bowel disease and between IL12B and Crohn's disease. CONCLUSIONS Our data may aid understanding of the molecular regulation of the circulating inflammatory proteome as well as causal relationships between inflammatory mediators and disease.
Collapse
Affiliation(s)
- Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Daniel Trejo-Banos
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Athanasios Kousathanas
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Sarah E Harris
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Anna J Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Marion Patxot
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Sven Erik Ojavee
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Qian Zhang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - David C Liewald
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Craig W Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Elliot M Tucker-Drob
- Department of Psychology, The University of Texas at Austin, Austin, TX, 78712, USA
- Population Research Center, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Ian J Deary
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Matthew R Robinson
- Institute of Science and Technology Austria, 3400, Klosterneuburg, Austria.
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
47
|
Zhong W, Spracklen CN, Mohlke KL, Zheng X, Fine J, Li Y. Multi-SNP mediation intersection-union test. Bioinformatics 2020; 35:4724-4729. [PMID: 31099385 DOI: 10.1093/bioinformatics/btz285] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/19/2019] [Accepted: 04/16/2019] [Indexed: 12/27/2022] Open
Abstract
SUMMARY Tens of thousands of reproducibly identified GWAS (Genome-Wide Association Studies) variants, with the vast majority falling in non-coding regions resulting in no eventual protein products, call urgently for mechanistic interpretations. Although numerous methods exist, there are few, if any methods, for simultaneously testing the mediation effects of multiple correlated SNPs via some mediator (e.g. the expression of a gene in the neighborhood) on phenotypic outcome. We propose multi-SNP mediation intersection-union test (SMUT) to fill in this methodological gap. Our extensive simulations demonstrate the validity of SMUT as well as substantial, up to 92%, power gains over alternative methods. In addition, SMUT confirmed known mediators in a real dataset of Finns for plasma adiponectin level, which were missed by many alternative methods. We believe SMUT will become a useful tool to generate mechanistic hypotheses underlying GWAS variants, facilitating functional follow-up. AVAILABILITY AND IMPLEMENTATION The R package SMUT is publicly available from CRAN at https://CRAN.R-project.org/package=SMUT. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Wujuan Zhong
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cassandra N Spracklen
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaojing Zheng
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason Fine
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Statistics and Operations Research, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
48
|
Dodig-Crnković T, Hong MG, Thomas CE, Häussler RS, Bendes A, Dale M, Edfors F, Forsström B, Magnusson PKE, Schuppe-Koistinen I, Odeberg J, Fagerberg L, Gummesson A, Bergström G, Uhlén M, Schwenk JM. Facets of individual-specific health signatures determined from longitudinal plasma proteome profiling. EBioMedicine 2020; 57:102854. [PMID: 32629387 PMCID: PMC7334812 DOI: 10.1016/j.ebiom.2020.102854] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Precision medicine approaches aim to tackle diseases on an individual level through molecular profiling. Despite the growing knowledge about diseases and the reported diversity of molecular phenotypes, the descriptions of human health on an individual level have been far less elaborate. METHODS To provide insights into the longitudinal protein signatures of well-being, we profiled blood plasma collected over one year from 101 clinically healthy individuals using multiplexed antibody assays. After applying an antibody validation scheme, we utilized > 700 protein profiles for in-depth analyses of the individuals' short-term health trajectories. FINDINGS We found signatures of circulating proteomes to be highly individual-specific. Considering technical and longitudinal variability, we observed that 49% of the protein profiles were stable over one year. We also identified eight networks of proteins in which 11-242 proteins covaried over time. For each participant, there were unique protein profiles of which some could be explained by associations to genetic variants. INTERPRETATION This observational and non-interventional study identifyed noticeable diversity among clinically healthy subjects, and facets of individual-specific signatures emerged by monitoring the variability of the circulating proteomes over time. To enable more personal hence precise assessments of health states, longitudinal profiling of circulating proteomes can provide a valuable component for precision medicine approaches. FUNDING This work was supported by the Erling Persson Foundation, the Swedish Heart and Lung Foundation, the Knut and Alice Wallenberg Foundation, Science for Life Laboratory, and the Swedish Research Council.
Collapse
Affiliation(s)
- Tea Dodig-Crnković
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Mun-Gwan Hong
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Cecilia Engel Thomas
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Ragna S Häussler
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Annika Bendes
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Matilda Dale
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Björn Forsström
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm 171 77, Sweden
| | - Ina Schuppe-Koistinen
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Center for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Jacob Odeberg
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT the Arctic University of Norway, Tromsø 9010, Norway; Coagulation unit, Department of Hematology, Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg 413 45, Sweden; Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg 413 45, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg 413 45, Sweden; Region Västra Götaland, Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg 413 45, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden.
| |
Collapse
|
49
|
Raffield LM, Dang H, Pratte KA, Jacobson S, Gillenwater L, Ampleford E, Barjaktarevic I, Basta P, Clish CB, Comellas AP, Cornell E, Curtis JL, Doerschuk C, Durda P, Emson C, Freeman C, Guo X, Hastie AT, Hawkins GA, Herrera J, Johnson WC, Labaki WW, Liu Y, Masters B, Miller M, Ortega VE, Papanicolaou G, Peters S, Taylor KD, Rich SS, Rotter JI, Auer P, Reiner AP, Tracy RP, Ngo D, Gerszten RE, O’Neal WK, Bowler RP, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium. Comparison of Proteomic Assessment Methods in Multiple Cohort Studies. Proteomics 2020; 20:e1900278. [PMID: 32386347 PMCID: PMC7425176 DOI: 10.1002/pmic.201900278] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 04/30/2020] [Indexed: 11/09/2022]
Abstract
Novel proteomics platforms, such as the aptamer-based SOMAscan platform, can quantify large numbers of proteins efficiently and cost-effectively and are rapidly growing in popularity. However, comparisons to conventional immunoassays remain underexplored, leaving investigators unsure when cross-assay comparisons are appropriate. The correlation of results from immunoassays with relative protein quantification is explored by SOMAscan. For 63 proteins assessed in two chronic obstructive pulmonary disease (COPD) cohorts, subpopulations and intermediate outcome measures in COPD Study (SPIROMICS), and COPDGene, using myriad rules based medicine multiplex immunoassays and SOMAscan, Spearman correlation coefficients range from -0.13 to 0.97, with a median correlation coefficient of ≈0.5 and consistent results across cohorts. A similar range is observed for immunoassays in the population-based Multi-Ethnic Study of Atherosclerosis and for other assays in COPDGene and SPIROMICS. Comparisons of relative quantification from the antibody-based Olink platform and SOMAscan in a small cohort of myocardial infarction patients also show a wide correlation range. Finally, cis pQTL data, mass spectrometry aptamer confirmation, and other publicly available data are integrated to assess relationships with observed correlations. Correlation between proteomics assays shows a wide range and should be carefully considered when comparing and meta-analyzing proteomics data across assays and studies.
Collapse
Affiliation(s)
- Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Katherine A. Pratte
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | - Sean Jacobson
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | - Lucas Gillenwater
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | - Elizabeth Ampleford
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Igor Barjaktarevic
- UCLA Division of Pulmonary and Critical Care, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Patricia Basta
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Clary B. Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Elaine Cornell
- Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Jeffrey L. Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI
- VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Claire Doerschuk
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Peter Durda
- Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Claire Emson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZenenca, Gaithersburg, MD
| | - Christine Freeman
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI
- VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Annette T. Hastie
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Gregory A. Hawkins
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI
| | - Yongmei Liu
- Department of Epidemiology & Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | | | | | - Victor E. Ortega
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - George Papanicolaou
- Epidemiology Branch, Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD
| | - Stephen Peters
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Paul Auer
- Zilber School of Public Health, University of Wisconsin Milwaukee, Milwaukee, WI
| | - Alex P. Reiner
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT
- Department of Biochemistry, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Debby Ngo
- Division of Pulmonary, Sleep and Critical Care Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Wanda Kay O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Russell P. Bowler
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | | |
Collapse
|
50
|
Darnell EP, Wroblewski KE, Pagel KL, Kern DW, McClintock MK, Pinto JM. IL-1Rahigh-IL-4low-IL-13low: A Novel Plasma Cytokine Signature Associated with Olfactory Dysfunction in Older US Adults. Chem Senses 2020; 45:407-414. [PMID: 32369568 PMCID: PMC7320218 DOI: 10.1093/chemse/bjaa029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inflammation has been implicated in physical frailty, but its role in sensory impairment is unclear. Given that olfactory impairment predicts dementia and mortality, determining the role of the immune system in olfactory dysfunction would provide insights mechanisms of neurosensory decline. We analyzed data from the National Social Life, Health and Aging Project, a representative sample of home-dwelling older US adults. Plasma levels of 18 cytokines were measured using standard protocols (Luminex xMAP). Olfactory function was assessed with validated tools (n-butanol sensitivity and odor identification, each via Sniffin' Sticks). We tested the association between cytokine profiles and olfactory function using multivariate ordinal logistic regression, adjusting for age, gender, race/ethnicity, education level, cognitive function, smoking status, and comorbidity. Older adults with the IL-1Rahigh-IL-4low-IL-13low cytokine profile had worse n-butanol odor sensitivity (odds ratio [OR] = 1.61, 95% confidence interval [CI] 1.19-2.17) and worse odor identification (OR = 1.42, 95% CI 1.11-1.80). Proinflammatory, Th1, or Th2 cytokine profiles were not associated with olfactory function. Moreover, accounting for physical frailty did not alter the main findings. In conclusion, we identified a plasma cytokine signature-IL-1Rahigh-IL-4low-IL-13low-that is associated with olfactory dysfunction in older US adults. These data implicate systemic inflammation in age-related olfactory dysfunction and support a role for immune mechanisms in this process, a concept that warrants additional scrutiny.
Collapse
Affiliation(s)
- Eli P Darnell
- Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
| | - Kristen E Wroblewski
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Kristina L Pagel
- Department of Comparative Human Development, The University of Chicago, Chicago, IL, USA
- Center on Demography and Aging, The University of Chicago, Chicago, IL, USA
- Institute for Mind and Biology, The University of Chicago, Chicago, IL, USA
| | - David W Kern
- Department of Comparative Human Development, The University of Chicago, Chicago, IL, USA
| | - Martha K McClintock
- Department of Comparative Human Development, The University of Chicago, Chicago, IL, USA
- Center on Demography and Aging, The University of Chicago, Chicago, IL, USA
- Institute for Mind and Biology, The University of Chicago, Chicago, IL, USA
| | - Jayant M Pinto
- Center on Demography and Aging, The University of Chicago, Chicago, IL, USA
- Section of Otolaryngology—Head and Neck Surgery, Department of Surgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|