148 results on '"David C. Nickle"'
Search Results
2. Protein-altering germline mutations implicate novel genes related to lung cancer development
- Author
-
Xuemei Ji, Semanti Mukherjee, Maria Teresa Landi, Yohan Bosse, Philippe Joubert, Dakai Zhu, Ivan Gorlov, Xiangjun Xiao, Younghun Han, Olga Gorlova, Rayjean J. Hung, Yonathan Brhane, Robert Carreras-Torres, David C. Christiani, Neil Caporaso, Mattias Johansson, Geoffrey Liu, Stig E. Bojesen, Loic Le Marchand, Demetrios Albanes, Heike Bickeböller, Melinda C. Aldrich, William S. Bush, Adonina Tardon, Gad Rennert, Chu Chen, Jinyoung Byun, Konstantin H. Dragnev, John K. Field, Lambertus FA. Kiemeney, Philip Lazarus, Shan Zienolddiny, Stephen Lam, Matthew B. Schabath, Angeline S. Andrew, Pier A. Bertazzi, Angela C. Pesatori, Nancy Diao, Li Su, Lei Song, Ruyang Zhang, Natasha Leighl, Jakob S. Johansen, Anders Mellemgaard, Walid Saliba, Christopher Haiman, Lynne Wilkens, Ana Fernandez-Somoano, Guillermo Fernandez-Tardon, Erik H. F. M. van der Heijden, Jin Hee Kim, Michael P. A. Davies, Michael W. Marcus, Hans Brunnström, Jonas Manjer, Olle Melander, David C. Muller, Kim Overvad, Antonia Trichopoulou, Rosario Tumino, Gary E. Goodman, Angela Cox, Fiona Taylor, Penella Woll, Erich Wichmann, Thomas Muley, Angela Risch, Albert Rosenberger, Kjell Grankvist, Mikael Johansson, Frances Shepherd, Ming-Sound Tsao, Susanne M. Arnold, Eric B. Haura, Ciprian Bolca, Ivana Holcatova, Vladimir Janout, Milica Kontic, Jolanta Lissowska, Anush Mukeria, Simona Ognjanovic, Tadeusz M. Orlowski, Ghislaine Scelo, Beata Swiatkowska, David Zaridze, Per Bakke, Vidar Skaug, Lesley M. Butler, Kenneth Offit, Preethi Srinivasan, Chaitanya Bandlamudi, Matthew D. Hellmann, David B. Solit, Mark E. Robson, Charles M. Rudin, Zsofia K. Stadler, Barry S. Taylor, Michael F. Berger, Richard Houlston, John McLaughlin, Victoria Stevens, David C. Nickle, Ma’en Obeidat, Wim Timens, María Soler Artigas, Sanjay Shete, Hermann Brenner, Stephen Chanock, Paul Brennan, James D. McKay, and Christopher I. Amos
- Subjects
Science - Abstract
In lung cancer, relatively few germline mutations are known to impact risk. Here the authors looked at rare variants in 39,146 individuals and find novel germline mutations associated with risk, as well as implicating ATM and a new candidate gene for lung cancer risk.
- Published
- 2020
- Full Text
- View/download PDF
3. Identification of susceptibility pathways for the role of chromosome 15q25.1 in modifying lung cancer risk
- Author
-
Xuemei Ji, Yohan Bossé, Maria Teresa Landi, Jiang Gui, Xiangjun Xiao, David Qian, Philippe Joubert, Maxime Lamontagne, Yafang Li, Ivan Gorlov, Mariella de Biasi, Younghun Han, Olga Gorlova, Rayjean J. Hung, Xifeng Wu, James McKay, Xuchen Zong, Robert Carreras-Torres, David C. Christiani, Neil Caporaso, Mattias Johansson, Geoffrey Liu, Stig E. Bojesen, Loic Le Marchand, Demetrios Albanes, Heike Bickeböller, Melinda C. Aldrich, William S. Bush, Adonina Tardon, Gad Rennert, Chu Chen, M. Dawn Teare, John K. Field, Lambertus A. Kiemeney, Philip Lazarus, Aage Haugen, Stephen Lam, Matthew B. Schabath, Angeline S. Andrew, Hongbing Shen, Yun-Chul Hong, Jian-Min Yuan, Pier A. Bertazzi, Angela C. Pesatori, Yuanqing Ye, Nancy Diao, Li Su, Ruyang Zhang, Yonathan Brhane, Natasha Leighl, Jakob S. Johansen, Anders Mellemgaard, Walid Saliba, Christopher Haiman, Lynne Wilkens, Ana Fernandez-Somoano, Guillermo Fernandez-Tardon, Erik H. F. M. van der Heijden, Jin Hee Kim, Juncheng Dai, Zhibin Hu, Michael P. A. Davies, Michael W. Marcus, Hans Brunnström, Jonas Manjer, Olle Melander, David C. Muller, Kim Overvad, Antonia Trichopoulou, Rosario Tumino, Jennifer Doherty, Gary E. Goodman, Angela Cox, Fiona Taylor, Penella Woll, Irene Brüske, Judith Manz, Thomas Muley, Angela Risch, Albert Rosenberger, Kjell Grankvist, Mikael Johansson, Frances Shepherd, Ming-Sound Tsao, Susanne M. Arnold, Eric B. Haura, Ciprian Bolca, Ivana Holcatova, Vladimir Janout, Milica Kontic, Jolanta Lissowska, Anush Mukeria, Simona Ognjanovic, Tadeusz M. Orlowski, Ghislaine Scelo, Beata Swiatkowska, David Zaridze, Per Bakke, Vidar Skaug, Shanbeh Zienolddiny, Eric J. Duell, Lesley M. Butler, Woon-Puay Koh, Yu-Tang Gao, Richard Houlston, John McLaughlin, Victoria Stevens, David C. Nickle, Ma’en Obeidat, Wim Timens, Bin Zhu, Lei Song, María Soler Artigas, Martin D. Tobin, Louise V. Wain, Fangyi Gu, Jinyoung Byun, Ahsan Kamal, Dakai Zhu, Rachel F. Tyndale, Wei-Qi Wei, Stephen Chanock, Paul Brennan, and Christopher I. Amos
- Subjects
Science - Abstract
The chromosome 15q25.1 locus is a leading susceptibility region for lung cancer. Here, the authors interrogate three GWAS cohorts with 42,901 individuals to investigate potential pathological pathways such as gated channel activity and neuroactive ligand receptor interaction in lung cancer etiology.
- Published
- 2018
- Full Text
- View/download PDF
4. Multiethnic meta-analysis identifies ancestry-specific and cross-ancestry loci for pulmonary function
- Author
-
Annah B. Wyss, Tamar Sofer, Mi Kyeong Lee, Natalie Terzikhan, Jennifer N. Nguyen, Lies Lahousse, Jeanne C. Latourelle, Albert Vernon Smith, Traci M. Bartz, Mary F. Feitosa, Wei Gao, Tarunveer S. Ahluwalia, Wenbo Tang, Christopher Oldmeadow, Qing Duan, Kim de Jong, Mary K. Wojczynski, Xin-Qun Wang, Raymond Noordam, Fernando Pires Hartwig, Victoria E. Jackson, Tianyuan Wang, Ma’en Obeidat, Brian D. Hobbs, Tianxiao Huan, Hongsheng Gui, Margaret M. Parker, Donglei Hu, Lauren S. Mogil, Gleb Kichaev, Jianping Jin, Mariaelisa Graff, Tamara B. Harris, Ravi Kalhan, Susan R. Heckbert, Lavinia Paternoster, Kristin M. Burkart, Yongmei Liu, Elizabeth G. Holliday, James G. Wilson, Judith M. Vonk, Jason L. Sanders, R. Graham Barr, Renée de Mutsert, Ana Maria Baptista Menezes, Hieab H. H. Adams, Maarten van den Berge, Roby Joehanes, Albert M. Levin, Jennifer Liberto, Lenore J. Launer, Alanna C. Morrison, Colleen M. Sitlani, Juan C. Celedón, Stephen B. Kritchevsky, Rodney J. Scott, Kaare Christensen, Jerome I. Rotter, Tobias N. Bonten, Fernando César Wehrmeister, Yohan Bossé, Shujie Xiao, Sam Oh, Nora Franceschini, Jennifer A. Brody, Robert C. Kaplan, Kurt Lohman, Mark McEvoy, Michael A. Province, Frits R. Rosendaal, Kent D. Taylor, David C. Nickle, L. Keoki Williams, Esteban G. Burchard, Heather E. Wheeler, Don D. Sin, Vilmundur Gudnason, Kari E. North, Myriam Fornage, Bruce M. Psaty, Richard H. Myers, George O’Connor, Torben Hansen, Cathy C. Laurie, Patricia A. Cassano, Joohon Sung, Woo Jin Kim, John R. Attia, Leslie Lange, H. Marike Boezen, Bharat Thyagarajan, Stephen S. Rich, Dennis O. Mook-Kanamori, Bernardo Lessa Horta, André G. Uitterlinden, Hae Kyung Im, Michael H. Cho, Guy G. Brusselle, Sina A. Gharib, Josée Dupuis, Ani Manichaikul, and Stephanie J. London
- Subjects
Science - Abstract
Pulmonary function is influenced by environmental factors, lifestyle, and genetics. Here, in a multiethnic GWAS meta-analysis for pulmonary function traits, the authors identify over 50 additional genetic loci, a subset of which are specific for European, African, Asian, or Hispanic/Latino ancestry.
- Published
- 2018
- Full Text
- View/download PDF
5. Responsiveness to Ipratropium Bromide in Male and Female Patients with Mild to Moderate Chronic Obstructive Pulmonary Disease
- Author
-
Xuan Li, Ma'en Obeidat, Guohai Zhou, Janice M. Leung, Donald Tashkin, Robert Wise, John Connett, Philippe Joubert, Yohan Bossé, Maarten van den Berge, Corry-Anke Brandsma, David C. Nickle, Ke Hao, Peter D. Paré, and Don D. Sin
- Subjects
Sex ,FEV1 ,Ipratropium ,COPD ,Gene expression ,Lung ,Medicine ,Medicine (General) ,R5-920 - Abstract
Introduction: Although the prevalence of chronic obstructive pulmonary disease (COPD) is similar between men and women, current evidence used to support bronchodilator therapy has been generated in therapeutic trials that have predominately enrolled male patients. Here, we determined whether there is any significant sex-related differences in FEV1 responses to ipratropium bromide. Methods: Data from the Lung Health Study (n = 5887; 37% females) were used to determine changes in FEV1 with ipratropium or placebo in male and female subjects with mild to moderate COPD over 5 years. Lung Expression Quantitative Trait Loci (eQTL) dataset was used to determine whether there were any sex-related differences in gene expression for muscarinic (M2 and M3) receptors in lungs of male and female patients. Results: After 4 months, ipratropium therapy increased FEV1 by 6.0% in female and 2.9% in male subjects from baseline values (p = 2.42 × 10−16). This effect was modified by body mass index (BMI) such that the biggest improvements in FEV1 with ipratropium were observed in thin female subjects (p for BMI ∗ sex interaction = 0.044). The sex-related changes in FEV1 related to ipratropium persisted for 2 years (p = 0.0134). Female compared with male lungs had greater gene expression for M3 relative to M2 receptors (p = 6.86 × 10−8). Conclusion: Ipratropium induces a larger bronchodilator response in female than in male patients and the benefits are particularly notable in non-obese females. Female lungs have greater gene expression for the M3 muscarinic receptor relative to M2 receptors than male lungs. Female patients are thus more likely to benefit from ipratropium than male COPD patients.
- Published
- 2017
- Full Text
- View/download PDF
6. DIVEIN: a web server to analyze phylogenies, sequence divergence, diversity, and informative sites
- Author
-
Wenjie Deng, Brandon S. Maust, David C. Nickle, Gerald H. Learn, Yi Liu, Laura Heath, Sergei L Kosakovsky Pond, and James I. Mullins
- Subjects
phylogeny ,divergence ,diversity ,informative sites ,center of tree ,maximum likelihood ,Biology (General) ,QH301-705.5 - Abstract
DIVEIN is a web interface that performs automated phylogenetic and other analyses of nucleotide and amino acid sequences. Starting with a set of aligned sequences, DIVEIN estimates evolutionary parameters and phylogenetic trees while allowing the user to choose from a variety of evolutionary models; it then reconstructs the consensus (CON), most recent common ancestor (MRCA), and center of tree (COT) sequences. DIVEIN also provides tools for further analyses, including condensing sequence alignments to show only informative sites or private mutations; computing phylogenetic or pairwise divergence from any user-specified sequence (CON, MRCA, COT, or existing sequence from the alignment); computing and outputting all genetic distances in column format; calculating summary statistics of diversity and divergence from pairwise distances; and graphically representing the inferred tree and plots of divergence, diversity, and distance distribution histograms. DIVEIN is available at http://indra.mullins.microbiol.washington.edu/DIVEIN.
- Published
- 2010
- Full Text
- View/download PDF
7. Sources of variation in ancestral sequence reconstruction for HIV-1 envelope genes
- Author
-
James I. Mullins, Allen G. Rodrigo, Mark A. Jensen, Laura Heath, Yi Liu, David C. Nickle, and Howard A. Ross
- Subjects
ancestral sequence reconstruction ,center of tree ,taxon sampling ,rational vaccine design ,HIV-1 ,Evolution ,QH359-425 - Abstract
We characterized the variation in the reconstructed ancestor of 118 HIV-1 envelope gene sequences arising from the methods used for (a) estimating and (b) rooting the phylogenetic tree, and (c) reconstructing the ancestor on that tree, from (d) the sequence format, and from (e) the number of input sequences. The method of rooting the tree was responsible for most of the sequence variation both among the reconstructed ancestral sequences and between the ancestral and observed sequences. Variation in predicted 3-D structural properties of the ancestors mirrored their sequence variation. The observed sequence consensus and ancestral sequences from center-rooted trees were most similar in all predicted attributes. Only for the predicted number of N-glycosylation sites was there a difference between MP and ML methods of reconstruction. Taxon sampling effects were observed only for outgrouprooted trees, not center-rooted, reflecting the occurrence of several divergent basal sequences. Thus, for sequences exhibiting a radial phylogenetic tree, as does HIV-1, most of the variation in the estimated ancestor arises from the method of rooting the phylogenetic tree. Those investigating the ancestors of genes exhibiting such a radial tree should pay particular attention to alternate rooting methods in order to obtain a representative sample of ancestors.
- Published
- 2006
8. Correction: Lung eQTLs to Help Reveal the Molecular Underpinnings of Asthma.
- Author
-
Ke Hao, Yohan Bossé, David C. Nickle, Peter D. Paré, Dirkje S. Postma, Michel Laviolette, Andrew Sandford, Tillie L. Hackett, Denise Daley, James C. Hogg, W. Mark Elliott, Christian Couture, Maxime Lamontagne, Corry-Anke Brandsma, Maarten van den Berge, Gerard Koppelman, Alise S. Reicin, Donald W. Nicholson, Vladislav Malkov, Jonathan M. Derry, Christine Suver, Jeffrey A. Tsou, Amit Kulkarni, Chunsheng Zhang, Rupert Vessey, Greg J. Opiteck, Sean P. Curtis, Wim Timens, and Don D. Sin
- Subjects
Genetics ,QH426-470 - Published
- 2012
- Full Text
- View/download PDF
9. Sources of Variation in Ancestral Sequence Reconstruction for HIV-1 Envelope Genes
- Author
-
Howard A. Ross, David C. Nickle, Yi Liu, Laura Heath, Mark A. Jensen, Allen G. Rodrigo, and James I. Mullins
- Subjects
Evolution ,QH359-425 - Abstract
We characterized the variation in the reconstructed ancestor of 118 HIV-1 envelope gene sequences arising from the methods used for (a) estimating and (b) rooting the phylogenetic tree, and (c) reconstructing the ancestor on that tree, from (d) the sequence format, and from (e) the number of input sequences. The method of rooting the tree was responsible for most of the sequence variation both among the reconstructed ancestral sequences and between the ancestral and observed sequences. Variation in predicted 3-D structural properties of the ancestors mirrored their sequence variation. The observed sequence consensus and ancestral sequences from center-rooted trees were most similar in all predicted attributes. Only for the predicted number of N-glycosylation sites was there a difference between MP and ML methods of reconstruction. Taxon sampling effects were observed only for outgroup-rooted trees, not center-rooted, reflecting the occurrence of several divergent basal sequences. Thus, for sequences exhibiting a radial phylogenetic tree, as does HIV-1, most of the variation in the estimated ancestor arises from the method of rooting the phylogenetic tree. Those investigating the ancestors of genes exhibiting such a radial tree should pay particular attention to alternate rooting methods in order to obtain a representative sample of ancestors.
- Published
- 2006
- Full Text
- View/download PDF
10. Detection of respiratory syncytial virus defective genomes in nasal secretions is associated with distinct clinical outcomes
- Author
-
Kedir N. Turi, Allan Paras, Carolina B. López, Ana María Cárdenas, Agnieszka Jozwik, Kristen A. Feemster, Yan Sun, Sébastien A. Felt, Emna Achouri, Christopher Chiu, Larry J. Anderson, David C. Nickle, Maximillian S. Habibi, Shaon Sengupta, Tina V. Hartert, and Meiping Chang
- Subjects
Male ,Microbiology (medical) ,viruses ,Immunology ,Mucous membrane of nose ,Genome, Viral ,Respiratory Syncytial Virus Infections ,Applied Microbiology and Biotechnology ,Microbiology ,Genome ,Article ,Virus ,Defective virus ,Cohort Studies ,03 medical and health sciences ,Disease severity ,1108 Medical Microbiology ,Genetics ,Humans ,Medicine ,Respiratory system ,030304 developmental biology ,0303 health sciences ,Science & Technology ,030306 microbiology ,business.industry ,Infant, Newborn ,Defective Viruses ,Infant ,Cell Biology ,3. Good health ,Nasal Mucosa ,Viral replication ,Respiratory Syncytial Virus, Human ,Female ,business ,Life Sciences & Biomedicine ,Viral load ,0605 Microbiology - Abstract
Respiratory syncytial virus (RSV) causes respiratory illness in children, immunosuppressed individuals and the elderly. However, the viral factors influencing the clinical outcome of RSV infections remain poorly defined. Defective viral genomes (DVGs) can suppress virus replication by competing for viral proteins and by stimulating antiviral immunity. We studied the association between detection of DVGs of the copy-back type and disease severity in three RSV A-confirmed cohorts. In hospitalized children, detection of DVGs in respiratory samples at or around the time of admission associated strongly with more severe disease, higher viral load and a stronger pro-inflammatory response. Interestingly, in experimentally infected adults, the presence of DVGs in respiratory secretions differentially associated with RSV disease severity depending on when DVGs were detected. Detection of DVGs early after infection associated with low viral loads and mild disease, whereas detection of DVGs late after infection, especially if DVGs were present for prolonged periods, associated with high viral loads and severe disease. Taken together, we demonstrate that the kinetics of DVG accumulation and duration could predict clinical outcome of RSV A infection in humans, and thus could be used as a prognostic tool to identify patients at risk of worse clinical disease.
- Published
- 2021
11. Global Landscape of Clostridioides Difficile Phylogeography, Antibiotic Susceptibility, and Toxin Polymorphisms by Post-Hoc Whole-Genome Sequencing from the MODIFY I/II Studies
- Author
-
Rebecca L. Blanchard, Ziqing Deng, Zhen Zeng, Shida Zhu, Judong Shen, Lan Chen, Ye Peng, Peter M. Shaw, Junhua Li, Jie Meng, Pierra Y T Law, Mary Beth Dorr, Xun Xu, Huanzi Zhong, David C. Nickle, Mark H. Wilcox, Hailong Zhao, and Andrew Albright
- Subjects
0301 basic medicine ,Microbiology (medical) ,Antibiotic susceptibility ,Evolution ,Moxifloxacin ,030106 microbiology ,Genome ,Rifaximin ,03 medical and health sciences ,0302 clinical medicine ,Clostridioides (Clostridium) difficile ,Vancomycin ,Phylogenetics ,Metronidazole ,medicine ,SNP ,030212 general & internal medicine ,Typing ,Clade ,Gene ,Genetics ,Whole genome sequencing ,business.industry ,Phylogeography ,Infectious Diseases ,CDI ,Toxin ,business ,medicine.drug - Abstract
Introduction Clostridioides (Clostridium) difficile infection, the leading cause of healthcare-associated diarrhea, represents a significant burden on global healthcare systems. Despite being a global issue, information on C. difficile from a global perspective is lacking. The aim of this study is to model the global phylogeography of clinical C. difficile. Methods Using samples collected from the MODIFY I and II studies (NCT01241552, NCT01513239), we performed whole-genome sequencing of 1501 clinical isolates including 37 novel sequence types (STs), representing the largest worldwide collection to date. Results Our data showed ribotypes, multi-locus sequence typing clades, and whole-genome phylogeny were in good accordance. The clinical C. difficile genome was found to be more conserved than previously reported (61% core genes), and modest recombination rates of 1.4-5.0 were observed across clades. We observed a significant continent distribution preference among five C. difficile clades (Benjamini-Hochberg corrected Fisher's exact test P
- Published
- 2021
12. Phenotypic and functional translation of IL33 genetics in asthma
- Author
-
David O. Bates, Vincent Pang, Judith M. Vonk, Charlote K. Billington, John W. Holloway, Sangita Bhaker, Cornelis J. Vermeulen, Don D. Sin, Ian Sayers, Martin D. Tobin, Peter H. Howarth, F. Nicole Dijk, David C. Nickle, Ian P. Hall, Yohan Bossé, Louise V. Wain, Dominick E. Shaw, John D Blakey, Andrew M. Fogarty, Amisha Singapuri, Michael A. Portelli, Martijn C. Nawijn, Cheng J. Xu, Andrew V. Benest, Ma'en Obeidat, Liam G Heaney, Jenny Hankinson, Maarten van den Berge, Rekha Chaudhuri, Angela Simpson, Tricia M. McKeever, Simon R. Johnson, Adel H. Mansur, Robert Niven, Zara Pogson, Gabrielle A. Lockett, Christopher E. Brightling, Amanda P. Henry, Neil C. Thomson, Nick Shrine, Gerard H. Koppelman, Maria Ketelaar, Alen Faiz, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Adult ,Male ,0301 basic medicine ,Immunology ,asthma phenotypes ,Genome-wide association study ,Single-nucleotide polymorphism ,eQTL ,IL33 SNPs ,functional translation ,Polymorphism, Single Nucleotide ,Atopy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Immunology and Allergy ,Eosinophilia ,Genetic Predisposition to Disease ,Asthma ,bronchial epithelium ,Genetics ,House dust mite ,biology ,business.industry ,Middle Aged ,respiratory system ,Interleukin-33 ,medicine.disease ,biology.organism_classification ,respiratory tract diseases ,030104 developmental biology ,Gene Expression Regulation ,030228 respiratory system ,Expression quantitative trait loci ,Sputum ,Female ,medicine.symptom ,business ,Genome-Wide Association Study - Abstract
Background: Asthma is a complex disease with multiple phenotypes that may differ in disease pathobiology and treatment response. IL33 single nucleotide polymorphisms (SNPs) have been reproducibly associated with asthma. IL33 levels are elevated in sputum and bronchial biopsies of patients with asthma. The functional consequences of IL33 asthma SNPs remain unknown. Objective: This study sought to determine whether IL33 SNPs associate with asthma-related phenotypes and with IL33 expression in lung or bronchial epithelium. This study investigated the effect of increased IL33 expression on human bronchial epithelial cell (HBEC) function. Methods: Association between IL33 SNPs (Chr9: 5,815,786-6,657,983) and asthma phenotypes (Lifelines/DAG [Dutch Asthma GWAS]/GASP [Genetics of Asthma Severity & Phenotypes] cohorts) and between SNPs and expression (lung tissue, bronchial brushes, HBECs) was done using regression modeling. Lentiviral overexpression was used to study IL33 effects on HBECs. Results: We found that 161 SNPs spanning the IL33 region associated with 1 or more asthma phenotypes after correction for multiple testing. We report a main independent signal tagged by rs992969 associating with blood eosinophil levels, asthma, and eosinophilic asthma. A second, independent signal tagged by rs4008366 presented modest association with eosinophilic asthma. Neither signal associated with FEV 1, FEV 1/forced vital capacity, atopy, and age of asthma onset. The 2 IL33 signals are expression quantitative loci in bronchial brushes and cultured HBECs, but not in lung tissue. IL33 overexpression in vitro resulted in reduced viability and reactive oxygen species–capturing of HBECs, without influencing epithelial cell count, metabolic activity, or barrier function. Conclusions: We identify IL33 as an epithelial susceptibility gene for eosinophilia and asthma, provide mechanistic insight, and implicate targeting of the IL33 pathway specifically in eosinophilic asthma.
- Published
- 2021
13. Late Breaking Abstract - Evidence of target engagement and pathway modulation: biomarker analysis of the phase 1b inhaled seralutinib study
- Author
-
Jean-Marie Bruey, Ramona Mccaffrey, Luke Howard, Charles D. Burger, John W. McConnell, Robert P. Frantz, Kristin B. Highland, Robert Roscigno, Robin Osterhout, David C. Nickle, Matt Cravets, and Lawrence S. Zisman
- Subjects
business.industry ,Modulation ,Target engagement ,Phase (waves) ,Medicine ,Biomarker Analysis ,business ,Neuroscience - Published
- 2021
14. New genetic signals for lung function highlight pathways and chronic obstructive pulmonary disease associations across multiple ancestries
- Author
-
Phuwanat Sakornsakolpat, James F. Wilson, María Soler Artigas, Beate Stubbe, Nicholas J. Wareham, James D. Crapo, Victoria E. Jackson, Eleftheria Zeggini, Olga G. Troyanskaya, Medea Imboden, A. Mesut Erzurumluoglu, Adam S. Butterworth, Carl A. Melbourne, Ian Sayers, Lars Lind, Stefan Weiss, Margaret M. Parker, Timmers Prhj, Edwin K. Silverman, John C. Whittaker, Ian P. Hall, Ozren Polasek, Mika Kähönen, Katherine A. Fawcett, Anna Hansell, Liming Li, David J. Porteous, Louise V. Wain, Robert A. Scott, Richard Packer, Richard J. Allen, Robin G Walters, Anna L. Guyatt, Terho Lehtimäki, Nicole Probst-Hensch, Shona M. Kerr, Christian Gieger, Matthias Wielscher, Rajesh Rawal, Jing Hua Zhao, John E. Hokanson, Holger Schulz, Nicola F. Reeve, Claudia Langenberg, Ke Hao, Corry-Anke Brandsma, Megan L. Paynton, Vitart, Dmitry Prokopenko, Jennie Hui, Eugene R. Bleecker, Ma'en Obeidat, John Danesh, Ralf Ewert, Benjamin B. Sun, Frank Dudbridge, Jian Zhou, Brian D. Hobbs, Blair H. Smith, Don D. Sin, Dawn L. DeMeo, Joseph C. Maranville, Ida Surakka, Alison Catherine Murray, Nicholas Locantore, Katherine A. Kentistou, Terri H. Beaty, Igor Rudan, Kuang Lin, Amund Gulsvik, Ian J. Deary, Ruth Tal-Singer, Kijoung Song, David C. Nickle, Michael H. Cho, Jian'an Luan, Joshua D. Hoffman, Georg Homuth, Zhengming Chen, Dandi Qiao, Heiko Runz, Per Bakke, Chiara Batini, Caroline Hayward, Stefan Karrasch, Alan James, Andrew P. Morris, Prescott G. Woodruff, Laura M Yerges Armstrong, David Sparrow, Ulf Gyllensten, James P. Cook, Jonathan Marten, Olli T. Raitakari, Nick Shrine, Deborah A. Meyers, Tobin, Ruth Boxall, Marjo-Riitta Järvelin, David P. Strachan, Sarah E. Harris, Philippe Joubert, Stefan Enroth, Xingnan Li, van den Berge M, Anubha Mahajan, Yohan Bossé, Peter K. Joshi, Shrine, Nick [0000-0003-3641-4371], Guyatt, Anna L [0000-0003-1860-6337], Hobbs, Brian D [0000-0001-9564-0745], Sakornsakolpat, Phuwanat [0000-0002-4308-0974], Obeidat, Ma'en [0000-0002-5443-2752], Weiss, Stefan [0000-0002-3553-4315], Kentistou, Katherine A [0000-0002-5816-664X], Sun, Benjamin B [0000-0001-6347-2281], Enroth, Stefan [0000-0002-5056-9137], Vitart, Veronique [0000-0002-4991-3797], Allen, Richard J [0000-0002-8450-3056], Beaty, Terri H [0000-0002-7644-1705], Bossé, Yohan [0000-0002-3067-3711], Joshi, Peter K [0000-0002-6361-5059], Mahajan, Anubha [0000-0001-5585-3420], Murray, Alison [0000-0003-4915-4847], Porteous, David J [0000-0003-1249-6106], Sayers, Ian [0000-0001-5601-5410], Smith, Blair H [0000-0002-5362-9430], Tal-Singer, Ruth [0000-0002-5275-8062], Timmers, Paul RHJ [0000-0002-5197-1267], Troyanskaya, Olga G [0000-0002-5676-5737], Rudan, Igor [0000-0001-6993-6884], Wilson, James F [0000-0001-5751-9178], Zeggini, Eleftheria [0000-0003-4238-659X], Hayward, Caroline [0000-0002-9405-9550], Morris, Andrew P [0000-0002-6805-6014], Butterworth, Adam S [0000-0002-6915-9015], Walters, Robin G [0000-0002-9179-0321], Cho, Michael H [0000-0002-4907-1657], Strachan, David P [0000-0001-7854-1366], Tobin, Martin D [0000-0002-3596-7874], Wain, Louise V [0000-0003-4951-1867], Apollo - University of Cambridge Repository, Institute for Molecular Medicine Finland, Groningen Research Institute for Asthma and COPD (GRIAC), UNIVERSITY OF OULU, and Commission of the European Communities
- Subjects
Male ,Smoking/genetics ,LOCI ,Genome-wide association study ,VARIANTS ,Bioinformatics ,Genome-wide association studies ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Polymorphism (computer science) ,Risk Factors ,GWAS ,GENOME-WIDE ASSOCIATION ,HERITABILITY ,COPD ,Lung ,11 Medical and Health Sciences ,Genetics & Heredity ,Aged, 80 and over ,0303 health sciences ,Genetic Predisposition to Disease/genetics ,Smoking ,1184 Genetics, developmental biology, physiology ,Middle Aged ,Polymorphism, Single Nucleotide/genetics ,3. Good health ,medicine.anatomical_structure ,Meta-analysis ,Genome-Wide Association Study/methods ,Medical genetics ,Female ,Life Sciences & Biomedicine ,medicine.medical_specialty ,Understanding Society Scientific Group ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,030304 developmental biology ,Genetic association ,Aged ,Respiratory tract diseases ,Science & Technology ,Case-control study ,Lung/physiopathology ,06 Biological Sciences ,medicine.disease ,Pulmonary Disease, Chronic Obstructive/genetics ,respiratory tract diseases ,Case-Control Studies ,3111 Biomedicine ,030217 neurology & neurosurgery ,Developmental Biology ,Genome-Wide Association Study - Abstract
Reduced lung function predicts mortality and is key to the diagnosis of chronic obstructive pulmonary disease (COPD). In a genome-wide association study in 400,102 individuals of European ancestry, we define 279 lung function signals, 139 of which are new. In combination, these variants strongly predict COPD in independent patient populations. Furthermore, the combined effect of these variants showed generalizability across smokers and never-smokers, and across ancestral groups. We highlight biological pathways, known and potential drug targets for COPD and, in phenome-wide association studies, autoimmune-related and other pleiotropic effects of lung function associated variants. This new genetic evidence has potential to improve future preventive and therapeutic strategies for COPD., Editorial summary: A genome-wide association study in over 400,000 individuals identifies 139 new signals for lung function. These variants can predict chronic obstructive pulmonary disease in independent, trans-ethnic cohorts.
- Published
- 2019
15. The Genetics of HIV-1
- Author
-
Willscott E. Naugler, Allen G. Rodrigo, Jon P. Anderson, Gerald H. Learn, David C. Nickle, Yang Wang, Matthew Rain, James I. Mullins, and Daniel Shriner
- Subjects
Genetics ,business.industry ,Human immunodeficiency virus (HIV) ,medicine ,medicine.disease_cause ,business - Published
- 2021
16. Aryl hydrocarbon receptor deficiency causes the development of chronic obstructive pulmonary disease through the integration of multiple pathogenic mechanisms
- Author
-
Jean Bourbeau, Patricia J. Sime, Michela Zago, Wan C. Tan, M. Obeidat, Parameswaran Nair, Qutayba Hamid, Corry-Anke Brandsma, Annette Robichaud, Benjamin M. Smith, Thomas H. Thatcher, Angela Rico de Souza, Richard P. Phipps, David H. Eidelman, Andrea Benedetti, Don D. Sin, Yohan Bossé, Swati Pareek, Jun Ding, Leora Simon, Necola Guerrina, Carolyn J. Baglole, David C. Nickle, Hussein Traboulsi, Pei Z. Li, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
0301 basic medicine ,Male ,SOD2 ,Disease ,MMP9 ,Biochemistry ,Article ,Pathogenesis ,03 medical and health sciences ,Mice ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Forced Expiratory Volume ,Genetics ,Medicine ,Animals ,Humans ,Receptor ,Molecular Biology ,Lung ,Aged ,Aged, 80 and over ,Emphysema ,COPD ,biology ,business.industry ,Aryl Hydrocarbon Receptor Nuclear Translocator ,Smoking ,Middle Aged ,respiratory system ,medicine.disease ,Aryl hydrocarbon receptor ,3. Good health ,respiratory tract diseases ,030104 developmental biology ,Receptors, Aryl Hydrocarbon ,Immunology ,biology.protein ,business ,030217 neurology & neurosurgery ,Homeostasis ,Biotechnology - Abstract
Emphysema, a component of chronic obstructive pulmonary disease (COPD), is characterized by irreversible alveolar destruction that results in a progressive decline in lung function. This alveolar destruction is caused by cigarette smoke, the most important risk factor for COPD. Only 15%-20% of smokers develop COPD, suggesting that unknown factors contribute to disease pathogenesis. We postulate that the aryl hydrocarbon receptor (AHR), a receptor/transcription factor highly expressed in the lungs, may be a new susceptibility factor whose expression protects against COPD. Here, we report that Ahr-deficient mice chronically exposed to cigarette smoke develop airspace enlargement concomitant with a decline in lung function. Chronic cigarette smoke exposure also increased cleaved caspase-3, lowered SOD2 expression, and altered MMP9 and TIMP-1 levels in Ahr-deficient mice. We also show that people with COPD have reduced expression of pulmonary and systemic AHR, with systemic AHR mRNA levels positively correlating with lung function. Systemic AHR was also lower in never-smokers with COPD. Thus, AHR expression protects against the development of COPD by controlling interrelated mechanisms involved in the pathogenesis of this disease. This study identifies the AHR as a new, central player in the homeostatic maintenance of lung health, providing a foundation for the AHR as a novel therapeutic target and/or predictive biomarker in chronic lung disease.
- Published
- 2021
17. Genetic associations and architecture of asthma-chronic obstructive pulmonary disease overlap
- Author
-
Jennifer K Quint, S. Leng, I. Jonsdottir, Tarja Laitinen, Amund Gulsvik, Natalie Terzikhan, Yohan Bossé, Caroline Hayward, Steven A. Belinsky, Ben Michael Brumpton, Hans Petersen, Xingnan Li, Victor E. Ortega, Richard Packer, C. Langenberg, Eugene R. Bleecker, Per Bakke, Ma'en Obeidat, Ian Sayers, A. Cepelis, Josée Dupuis, G. Thorleifsson, Martin D. Tobin, Yohannes Tesfaigzi, Lies Lahousse, Samuli Ripatti, Lystra P. Hayden, A. I. Hernandez Cordero, Carlos Iribarren, Jukka Koskela, Ian P. Hall, S.H. Chu, Sina A. Gharib, T.A. Olafsdottir, Louise V. Wain, Deborah A. Meyers, Catherine John, Craig P. Hersh, Anna L. Guyatt, Michael H. Cho, Ann Chen Wu, Guy Brusselle, David C. Nickle, Don D. Sin, M. van den Berge, Arnulf Langhammer, Jiangyuan Liu, Lori C. Sakoda, Nick Shrine, Jessica Lasky-Su, Traci M. Bartz, Hanfei Xu, K. Stefansson, and Jian'an Luan
- Subjects
0303 health sciences ,medicine.medical_specialty ,COPD ,business.industry ,Pulmonary disease ,medicine.disease ,Biobank ,Signal on ,Genetic architecture ,respiratory tract diseases ,3. Good health ,Asthma chronic ,03 medical and health sciences ,0302 clinical medicine ,030228 respiratory system ,Internal medicine ,medicine ,business ,Lung function ,030304 developmental biology ,Asthma - Abstract
Some individuals have characteristics of both asthma and COPD (asthma-COPD overlap, ACO), and evidence suggests they experience worse outcomes than those with either condition alone. Improved knowledge of the genetic architecture would contribute to understanding whether determinants of risk in this group differ from those in COPD or asthma.We conducted a genome-wide association study in 8,068 cases and 40,360 controls of European ancestry from UK Biobank (stage 1). After excluding variants only associated with asthma or COPD we selected 31 variants for further investigation in 12 additional cohorts (stage 2), and discovered eight novel signals for ACO in a meta-analysis of stage 1 and 2 studies.Our signals include an intergenic signal on chromosome 5 not previously associated with asthma, COPD or lung function, and suggest a spectrum of shared and distinct genetic influences in asthma, COPD and ACO. A number of signals may represent loci that predispose to serious long-term consequences in people with asthma.
- Published
- 2020
18. Rhinovirus and asthma: Challenges and opportunities
- Author
-
Laura L. Carter, David C. Nickle, and Hector Ortega
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Allergy ,Exacerbation ,030106 microbiology ,Population ,Reviews ,Common Cold ,Review ,medicine.disease_cause ,03 medical and health sciences ,Virology ,Internal medicine ,medicine ,Humans ,education ,Asthma ,education.field_of_study ,Picornaviridae Infections ,business.industry ,Genetic Variation ,Common cold ,medicine.disease ,SARS‐Cov‐2 ,respiratory tract diseases ,Pneumonia ,rhinovirus ,030104 developmental biology ,Infectious Diseases ,Bronchiolitis ,asthma exacerbations ,Rhinovirus ,influenza ,business - Abstract
Summary Human rhinoviruses (RVs) are the primary aetiological agent of the common cold. Generally, the associated infection is mild and self‐limiting, but may also be associated with bronchiolitis in infants, pneumonia in the immunocompromised and exacerbation in patients with pulmonary conditions such as asthma or chronic obstructive pulmonary disease. Viral infection accounts for as many as two thirds of asthma exacerbations in children and more than half in adults. Allergy and asthma are major risk factors for more frequent and severe RV‐related illnesses. The prevalence of RV‐induced wheezing will likely continue to increase given that asthma affects a significant proportion of the population, with allergic asthma accounting for the majority. Several new respiratory viruses and their subgroups have been discovered, with various degrees of relevance. This review will focus on RV infection in the context of the epidemiologic evidence, genetic variability, pathobiology, clinical studies in the context of asthma, differences with other viruses including COVID‐19 and current treatment interventions.
- Published
- 2020
19. 389 Combining transcriptomic- and tissue-based immune biomarkers to evaluate GB1275, a CD11b modulator, as a single agent or with pembrolizumab in patients with advanced solid tumors
- Author
-
Wungki Park, Jean-Marie Bruey, Johanna C. Bendell, Andrea Wang-Gillam, Laura L. Carter, Lei Zhou, David C. Nickle, Jakob Dupont, Drew W. Rasco, Anna Galkin, Marya F. Chaney, Wells A. Messersmith, Johann S. de Bono, and Beatrice Ferguson
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,Myeloid ,business.industry ,Tumor-infiltrating lymphocytes ,medicine.medical_treatment ,Cancer ,Pembrolizumab ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Immunophenotyping ,medicine.anatomical_structure ,Internal medicine ,medicine ,Biomarker (medicine) ,business ,Whole blood - Abstract
Background GB1275 is a first-in-class CD11b modulator in development as monotherapy and in combination with pembrolizumab or chemotherapy for the treatment of advanced solid tumors. Nonclinical data show that GB1275 reduced influx of tumor-associated myeloid-derived suppressor cells (MDSCs) and macrophages (TAMs), and repolarized M2 immuno-suppressive TAMs towards an M1 phenotype. We hypothesize that GB1275 administration can alleviate myeloid cell-mediated immunosuppressive effects and improve cancer treatment outcomes. A phase 1 trial evaluating GB1275 as monotherapy and in combination with pembrolizumab in specified advanced tumors in ongoing (NCT04060342). Methods Blood gene expression variations as well as core tissue biopsies pre- and post-treatment were assessed following GB1275 monotherapy and combination with pembrolizumab. After obtaining informed consent, peripheral blood for MDSCs was collected from 21 patients pre- and two weeks post-treatment; core tissue biopsies were collected from 13 patients pre- and post-treatment. The frequency of MDSCs in whole blood was measured using the Serametrix MDSC FACS Assay. Gene expression transcriptome profiles were generated using NovaSeq platform. CD8 staining was performed at Neogenomics, and tumor infiltrating lymphocyte (TIL) quantification was performed by an independent pathologist. Results Preliminary statistical analysis of MDSC immunophenotyping pre- and post- treatment is consistent with the proposed mechanism of GB1275, showing modulation of peripheral blood MDSCs in some patients. Preliminary gene expression analysis in the blood showed dose-dependent clusters following treatment with GB1275 alone. Moreover, the transcriptomic analysis revealed two unique expression patterns for patients treated with GB1275 monotherapy or in combination with pembrolizumab. Gene Set Enrichment Analysis showed that the CD11b pathway is downregulated in patients treated with GB1275. Analyses of TIL count revealed an increase in lymphocyte trafficking into the tumor after treatment with GB1275 alone or in combination with pembrolizumab. CD8 expression and transcriptomic analysis are underway and will be presented. Conclusions GB1275 alone or in combination with pembrolizumab demonstrates biological activity, which may be dose dependent. The observed increase in TILs after treatment is supportive of the mechanism of action of GB1275. Further biomarker analyses in blood and tissues are ongoing and will be correlated with clinical activity in a larger number of patients. Ethics Approval This ongoing study is being conducted in accordance with the the Declaration of Helsinki and Council for International Organizations of Medical Sciences (CIOMS) International Ethical Guidelines. The study was approved by the Ethics Boards of University of Colorado Hospital, Washington University School of Medicine - Siteman Cancer Center, Memorial Sloan Kettering Cancer Center, The Sarah Cannon Research Institute/Tennessee Oncology, South Texas Accelerated Research Therapeutics, and The Royal Marsden NHS Foundation Trust.
- Published
- 2020
20. Genetic regulation of gene expression of MIF family members in lung tissue
- Author
-
Corry-Anke Brandsma, Philippe Joubert, Gerrit J. Poelarends, Ma'en Obeidat, Maarten van den Berge, Laura Florez-Sampedro, Cornelis J. Vermeulen, Alen Faiz, Wim Timens, Rene Bults, David C. Nickle, Maaike de Vries, Barbro N. Melgert, Chemical and Pharmaceutical Biology, Groningen Research Institute for Asthma and COPD (GRIAC), Guided Treatment in Optimal Selected Cancer Patients (GUTS), Biopharmaceuticals, Discovery, Design and Delivery (BDDD), Medicinal Chemistry and Bioanalysis (MCB), and Molecular Pharmacology
- Subjects
0301 basic medicine ,Male ,Microarray ,lcsh:Medicine ,Single-nucleotide polymorphism ,Genome-wide association study ,chemical and pharmacologic phenomena ,Biology ,urologic and male genital diseases ,Polymorphism, Single Nucleotide ,Article ,Pathogenesis ,03 medical and health sciences ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Gene expression ,otorhinolaryngologic diseases ,SNP ,Humans ,lcsh:Science ,Lung ,Macrophage Migration-Inhibitory Factors ,Aged ,Regulation of gene expression ,Multidisciplinary ,Chronic obstructive pulmonary disease ,lcsh:R ,Functional genomics ,Middle Aged ,respiratory system ,biological factors ,Intramolecular Oxidoreductases ,030104 developmental biology ,030228 respiratory system ,Gene Expression Regulation ,Immunology ,Macrophage migration inhibitory factor ,lcsh:Q ,Female - Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine found to be associated with chronic obstructive pulmonary disease (COPD). However, there is no consensus on how MIF levels differ in COPD compared to control conditions and there are no reports on MIF expression in lung tissue. Here we studied gene expression of members of the MIF family MIF, D-Dopachrome Tautomerase (DDT) and DDT-like (DDTL) in a lung tissue dataset with 1087 subjects and identified single nucleotide polymorphisms (SNPs) regulating their gene expression. We found higher MIF and DDT expression in COPD patients compared to non-COPD subjects and found 71 SNPs significantly influencing gene expression of MIF and DDTL. Furthermore, the platform used to measure MIF (microarray or RNAseq) was found to influence the splice variants detected and subsequently the direction of the SNP effects on MIF expression. Among the SNPs found to regulate MIF expression, the major LD block identified was linked to rs5844572, a SNP previously found to be associated with lower diffusion capacity in COPD. This suggests that MIF may be contributing to the pathogenesis of COPD, as SNPs that influence MIF expression are also associated with symptoms of COPD. Our study shows that MIF levels are affected not only by disease but also by genetic diversity (i.e. SNPs). Since none of our significant eSNPs for MIF or DDTL have been described in GWAS for COPD or lung function, MIF expression in COPD patients is more likely a consequence of disease-related factors rather than a cause of the disease.
- Published
- 2020
21. Neutrophilic inflammation in the respiratory mucosa predisposes to RSV infection
- Author
-
Meiping Chang, Leah Cuthbertson, Trevor T. Hansel, Christopher Chiu, Tanushree Tunstall, Ryan S Thwaites, Amber Owen, Agnieszka Jozwik, Peter J. M. Openshaw, Augusto Varese, Phillip James, Miriam F. Moffatt, Cecilia Johansson, Allan Paras, Freja C M Kirsebom, Maximillian S. Habibi, David C. Nickle, Medical Research Council (MRC), Wellcome Trust, National Institute for Health Research, Commission of the European Communities, and GlaxoSmithKline Services Unlimited
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Neutrophils ,Chemokine CXCL1 ,medicine.medical_treatment ,CHILDREN ,CD8-Positive T-Lymphocytes ,Neutrophil Activation ,Mice ,0302 clinical medicine ,Respiratory system ,Multidisciplinary ,Interleukin-17 ,Middle Aged ,Respiratory Syncytial Viruses ,3. Good health ,Multidisciplinary Sciences ,Cytokine ,INFLUENZA ,DISEASE SEVERITY ,Science & Technology - Other Topics ,Tumor necrosis factor alpha ,Interleukin 17 ,medicine.symptom ,SYNCYTIAL VIRUS-INFECTION ,Viral load ,Adult ,Adolescent ,General Science & Technology ,Inflammation ,INNATE ,Respiratory Syncytial Virus Infections ,IMMUNITY ,Young Adult ,03 medical and health sciences ,Immune system ,Immunity ,medicine ,Animals ,Humans ,Science & Technology ,Tumor Necrosis Factor-alpha ,business.industry ,CD4 Lymphocyte Count ,SECUKINUMAB ,Mice, Inbred C57BL ,CYTOKINE ,Nasal Mucosa ,VIRAL LOAD ,030104 developmental biology ,Immunology ,T-CELLS ,business ,030215 immunology - Abstract
INTRODUCTION Even with intimate exposure to a virus, some people fail to become infected. Variable transmission partly depends on the dose and duration of exposure but is also governed by the immune status of the host, such as the presence of specific protective antibodies or T cells. However, for some infections, the reasons for erratic transmission are largely unknown. For example, respiratory syncytial virus (RSV) can repeatedly reinfect individuals throughout their lives despite the presence of specific immunity. Additionally, antibodies and T cells have limited efficacy against newly emergent pathogens with pandemic potential. However, the intrinsic and innate mechanisms underlying protection when people are exposed to these viruses are poorly understood. RATIONALE We reasoned that the prior state of the respiratory mucosa’s innate defenses may contribute to the variable outcome of RSV inoculation. By performing experimental challenge of adult volunteers, we were able to measure variations in the status of the nasal mucosa before inoculation and in mucosal responses during the presymptomatic phase of infection. Neither of these phases is easily observable during natural spontaneous transmission. Our observations could then be validated using specific interventional studies in a well-established mouse model of RSV infection. RESULTS After nasal administration of RSV, 57% of inoculated volunteers became infected. The uptake of infection was poorly explained by specific B or T cell immunity. However, transcriptomic profiling of the nasal tissue before inoculation demonstrated a neutrophilic inflammatory signal in those destined to develop symptomatic infection, and this was associated with suppression of an early interleukin-17 (IL-17)–dominated immune response during the presymptomatic period. This was followed by symptomatic infection associated with the expression of proinflammatory cytokines. By contrast, those who resisted infection showed a transient boost in mucosal markers of innate immune activation for several days after viral administration and no subsequent viral replication. In mice, chemokine-induced neutrophil recruitment to the airway before viral exposure transiently enhanced viral replication immediately after inoculation. As with human subjects, this was associated with decreased expression of some innate mediators in the respiratory tract, which was then followed by enhanced disease driven by CD8+ T cells. CONCLUSION Although reinfection with a respiratory virus can be partially explained by the waning of antibody titers and T cell numbers, neutrophilic inflammation in the airway at the time of pathogen exposure also predisposes individuals to symptomatic infection. After exposure, the response of the mucosa diverges: A mild and transient increase in nasal inflammatory mediators is accompanied by termination of viral infection, whereas failure to mount this response is followed by viral success. The state of innate immune preparedness of the respiratory mucosa is thus a major determinant of susceptibility to viral challenge. Our results show the importance of understanding the mucosal microenvironment in studies of respiratory infection and highlight targets for local intervention, which may enhance protection against a range of respiratory pathogens.
- Published
- 2020
22. Multi-omics highlights ABO plasma protein as a causal risk factor for COVID-19
- Author
-
Ana I. Hernández Cordero, Philippe Joubert, Chen Xi Yang, Ke Hao, Stephen Milne, David C. Nickle, Yohan Bossé, Maarten van den Berge, Don D. Sin, Wim Timens, Xuan Li, Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Candidate gene ,Quantitative Trait Loci ,Genome-wide association study ,Quantitative trait locus ,Biology ,Polymorphism, Single Nucleotide ,ABO Blood-Group System ,Cohort Studies ,03 medical and health sciences ,Risk Factors ,ABO blood group system ,Mendelian randomization ,Genetics ,Humans ,Genetic Predisposition to Disease ,Risk factor ,Lung ,Genetics (clinical) ,Original Investigation ,030304 developmental biology ,Blood type ,0303 health sciences ,SARS-CoV-2 ,030305 genetics & heredity ,COVID-19 ,Mendelian Randomization Analysis ,Blood Proteins ,Human genetics ,Coronavirus ,Expression quantitative trait loci ,Genome-Wide Association Study - Abstract
SARS-CoV-2 is responsible for the coronavirus disease 2019 (COVID-19) and the current health crisis. Despite intensive research efforts, the genes and pathways that contribute to COVID-19 remain poorly understood. We, therefore, used an integrative genomics (IG) approach to identify candidate genes responsible for COVID-19 and its severity. We used Bayesian colocalization (COLOC) and summary-based Mendelian randomization to combine gene expression quantitative trait loci (eQTLs) from the Lung eQTL (n = 1,038) and eQTLGen (n = 31,784) studies with published COVID-19 genome-wide association study (GWAS) data from the COVID-19 Host Genetics Initiative. Additionally, we used COLOC to integrate plasma protein quantitative trait loci (pQTL) from the INTERVAL study (n = 3,301) with COVID-19 loci. Finally, we determined any causal associations between plasma proteins and COVID-19 using multi-variable two-sample Mendelian randomization (MR). The expression of 18 genes in lung and/or blood co-localized with COVID-19 loci. Of these, 12 genes were in suggestive loci (PGWAS
- Published
- 2020
23. Prioritization of candidate causal genes in GWAS signals of asthma in UK Biobank
- Author
-
Jennifer Lamothe, Krystelle Godbout, Aida Eslami, Kim Valette, David C. Nickle, Arnaud Chignon, Andréanne Côté, Maarten van den Berge, Don D. Sin, Patrick Mathieu, Michel Laviolette, Zhonglin Li, Valentin Bon-Baret, Ke Hao, Yohan Bossé, Nathalie Gaudreault, Catherine Labbé, Jean-Christophe Bérubé, Philippe Joubert, Ma'en Obeidat, Sébastien Thériault, Louis-Philippe Boulet, and Wim Timens
- Subjects
Prioritization ,business.industry ,medicine ,Genome-wide association study ,Computational biology ,medicine.disease ,business ,Biobank ,Gene ,Asthma - Abstract
To identify susceptibility loci and candidate causal genes of asthma, we performed a genome-wide association study (GWAS) in UK Biobank on a broad asthma definition (n = 56,167 asthma cases and 352,255 controls). We then carried out functional mapping through transcriptome-wide association studies (TWAS) and Mendelian randomization in lung (n = 1,038) and blood (n = 31,684) tissues. The GWAS revealed 72 asthma-associated loci from 116 independent significant variants (PGWAS 20) including a stop-gain mutation in the filaggrin (FLG) gene. The top lung TWAS gene on 17q12-q21 was GSDMB (PTWAS=1.42E-54). Other TWAS genes of interest include TSLP on 5q22, RERE on 1p36, CLEC16A on 16p13, and IL4R on 16p12, which all replicated in GTEx lung (n = 515). A novel risk locus was also revealed by the lung asthma TWAS on 1q23.3 with the putative gene encoding the gamma chain of the high-affinity IgE receptor (FCER1G, PTWAS=2.13E-6), which was also replicated in GTEx lung (PTWAS=3.71E-7). By testing a comprehensive set of cells and tissues, we then demonstrated that the largest fold enrichment of regulatory and functional annotations among asthma-associated variants was in the blood. We mapped 485 eQTL-regulated genes associated with asthma in the blood and 50 of them were shown to be causally associated with asthma by Mendelian randomization. Prioritization of druggable genes revealed known (IL4R, TSLP, IL6, TNFSF4) and potentially new therapeutic targets for asthma.
- Published
- 2020
24. Prioritization of candidate causal genes for asthma in susceptibility loci derived from UK Biobank
- Author
-
Ma'en Obeidat, Krystelle Godbout, Kim Valette, Philippe Joubert, Louis-Philippe Boulet, Sébastien Thériault, Maarten van den Berge, Patrick Mathieu, Arnaud Chignon, Catherine Labbé, Valentin Bon-Baret, Jean-Christophe Bérubé, Jennifer Lamothe, Nathalie Gaudreault, Aida Eslami, David C. Nickle, Ke Hao, Don D. Sin, Zhonglin Li, Michel Laviolette, Wim Timens, Andréanne Côté, Yohan Bossé, Groningen Research Institute for Asthma and COPD (GRIAC), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
0301 basic medicine ,Adult ,Male ,QH301-705.5 ,Medicine (miscellaneous) ,Genome-wide association study ,CLEC16A ,VARIANTS ,Biology ,Genome-wide association studies ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Mendelian randomization ,medicine ,Humans ,Genetic Predisposition to Disease ,Biology (General) ,Gene ,EQTL ,Asthma ,Genetic association ,Aged ,Biological Specimen Banks ,RISK ,Genetics ,GENOME-WIDE ,ASSOCIATION ,Middle Aged ,medicine.disease ,Biobank ,United Kingdom ,Gene expression profiling ,respiratory tract diseases ,030104 developmental biology ,Genetic Loci ,Expression quantitative trait loci ,Female ,General Agricultural and Biological Sciences ,Transcriptome ,SET ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
To identify candidate causal genes of asthma, we performed a genome-wide association study (GWAS) in UK Biobank on a broad asthma definition (n = 56,167 asthma cases and 352,255 controls). We then carried out functional mapping through transcriptome-wide association studies (TWAS) and Mendelian randomization in lung (n = 1,038) and blood (n = 31,684) tissues. The GWAS reveals 72 asthma-associated loci from 116 independent significant variants (PGWAS, Kim Valette et al. perform a genomic study on asthma integrating genome-wide association study, functional mapping using lung and blood transcriptome-wide profiles, as well as Mendelian randomization. They show candidate causal genes expressed in lung and blood tissues that are putative therapeutic targets for asthma.
- Published
- 2020
25. Gene Expression Network Analysis Provides Potential Targets Against SARS-CoV-2
- Author
-
Xuan Li, Chen Xi Yang, Stephen Milne, Don D. Sin, Maarten van den Berge, Ana I. Hernández Cordero, Ke Hao, Philippe Joubert, Wim Timens, David C. Nickle, Yohan Bossé, Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Adult ,Male ,Science ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Druggability ,ACE2 ,Pathogenesis ,Computational biology ,Biology ,Endocytosis ,medicine.disease_cause ,TMPRSS2 ,Article ,Cohort Studies ,Transcriptome ,Gene expression ,Genetics ,medicine ,Humans ,Lung ,Gene ,Aged ,Coronavirus ,Serine protease ,Multidisciplinary ,Serine Endopeptidases ,COVID-19 ,Middle Aged ,Transmembrane protein ,Risk factors ,biology.protein ,Medicine ,Female ,Angiotensin-Converting Enzyme 2 ,Databases, Chemical ,Receptors, Coronavirus - Abstract
BACKGROUNDCell entry of SARS-CoV-2, the novel coronavirus causing COVID-19, is facilitated by host cell angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). We aimed to identify and characterize genes that are co-expressed withACE2andTMPRSS2, and to further explore their biological functions and potential as druggable targets.METHODSUsing the gene expression profiles of 1,038 lung tissue samples, we performed a weighted gene correlation network analysis (WGCNA) to identify modules of co-expressed genes. We explored the biology of co-expressed genes using bioinformatics databases, and identified known drug-gene interactions.RESULTSACE2was in a module of 681 co-expressed genes; 12 genes with moderate-high correlation withACE2(r>0.3, FDRTMPRSS2was in a module of 1,086 co-expressed genes; 15 of these genes were enriched in the gene ontology biologic process ‘Entry into host cell’, and 53TMPRSS2-correlated genes had known interactions with drug compounds.CONCLUSIONDozens of genes are co-expressed withACE2andTMPRSS2, many of which have plausible links to COVID-19 pathophysiology. Many of the co-expressed genes are potentially targetable with existing drugs, which may help to fast-track the development of COVID-19 therapeutics.
- Published
- 2020
26. Genetic Association Reveals Protection against Recurrence of Clostridium difficile Infection with Bezlotoxumab Treatment
- Author
-
Emily R. Holzinger, Devan V. Mehrotra, Junhua Li, Judong Shen, Mark H. Wilcox, Rebecca L. Blanchard, Xun Xu, Aparna Chhibber, Peter M. Shaw, David C. Nickle, Zhen Zeng, and Mary Beth Dorr
- Subjects
0301 basic medicine ,lcsh:QR1-502 ,Single-nucleotide polymorphism ,Clostridium difficile toxin B ,Human leukocyte antigen ,030226 pharmacology & pharmacy ,Microbiology ,lcsh:Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Genotype ,genomics ,Medicine ,Genetic variability ,bezlotoxumab ,Molecular Biology ,Genetic association ,antibacterials ,business.industry ,Clostridium difficile ,clostridium difficile ,QR1-502 ,030104 developmental biology ,Bezlotoxumab ,Immunology ,business - Abstract
Bezlotoxumab is a human monoclonal antibody against Clostridium difficile toxin B, indicated to prevent recurrence of C. difficile infection (rCDI) in high-risk adults receiving antibacterial treatment for CDI. An exploratory genome-wide association study investigated whether human genetic variation influences bezlotoxumab response. DNA from 704 participants who achieved initial clinical cure in the phase 3 MODIFY I/II trials was genotyped. Single nucleotide polymorphisms (SNPs) and human leukocyte antigen (HLA) imputation were performed using IMPUTE2 and HIBAG, respectively. A joint test of genotype and genotype-by-treatment interaction in a logistic regression model was used to screen genetic variants associated with response to bezlotoxumab. The SNP rs2516513 and the HLA alleles HLA-DRB1*07:01 and HLA-DQA1*02:01, located in the extended major histocompatibility complex on chromosome 6, were associated with the reduction of rCDI in bezlotoxumab-treated participants. Carriage of a minor allele (homozygous or heterozygous) at any of the identified loci was related to a larger difference in the proportion of participants experiencing rCDI versus placebo; the effect was most prominent in the subgroup at high baseline risk for rCDI. Genotypes associated with an improved bezlotoxumab response showed no association with rCDI in the placebo cohort. These data suggest that a host-driven, immunological mechanism may impact bezlotoxumab response. Trial registration numbers are as follows: NCT01241552 (MODIFY I) and NCT01513239 (MODIFY II). IMPORTANCEClostridium difficile infection is associated with significant clinical morbidity and mortality; antibacterial treatments are effective, but recurrence of C. difficile infection is common. In this genome-wide association study, we explored whether host genetic variability affected treatment responses to bezlotoxumab, a human monoclonal antibody that binds C. difficile toxin B and is indicated for the prevention of recurrent C. difficile infection. Using data from the MODIFY I/II phase 3 clinical trials, we identified three genetic variants associated with reduced rates of C. difficile infection recurrence in bezlotoxumab-treated participants. The effects were most pronounced in participants at high risk of C. difficile infection recurrence. All three variants are located in the extended major histocompatibility complex on chromosome 6, suggesting the involvement of a host-driven immunological mechanism in the prevention of C. difficile infection recurrence.
- Published
- 2020
27. Phenotypic and functional translation of IL1RL1 locus polymorphisms in lung tissue and asthmatic airway epithelium
- Author
-
Charlotte K. Billington, Gerard H. Koppelman, Liam G Heaney, Adel H. Mansur, Jenny Hankinson, Martijn C. Nawijn, Corry-Anke Brandsma, Sangita Bhaker, Andrew M. Fogarty, Dominick E. Shaw, Michael A. Portelli, Cornelis J. Vermeulen, Judith M. Vonk, Nick Shrine, Amanda P. Henry, Ian Sayers, Yohan Bossé, Maria Ketelaar, Martin D. Tobin, Peter H. Howarth, David C. Nickle, Néomi S. Grotenboer, Ma'en Obeidat, Gabrielle A. Lockett, John W. Holloway, Christopher E. Brightling, Alen Faiz, Rekha Chaudhuri, Tricia M. McKeever, Simon R. Johnson, Paul de Vos, Dirkje S. Postma, Zara Pogson, Neil C. Thomson, Sharon Brouwer, Maarten van den Berge, Angela Simpson, Amisha Singapuri, Robert Niven, F. Nicole Dijk, Ian P. Hall, Louise V. Wain, John D Blakey, Groningen Research Institute for Asthma and COPD (GRIAC), Translational Immunology Groningen (TRIGR), and Man, Biomaterials and Microbes (MBM)
- Subjects
HAY-FEVER ,EXPRESSION ,0301 basic medicine ,medicine.medical_specialty ,Genotype ,IL1RL1 ,Single-nucleotide polymorphism ,Genome-wide association study ,Locus (genetics) ,Respiratory Mucosa ,VARIANTS ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,Molecular genetics ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic variation ,GENOME-WIDE ASSOCIATION ,Lung ,Asthma ,LARGE-SCALE ,Haplotype ,Cell Biology ,General Medicine ,ST2 ,medicine.disease ,Interleukin-1 Receptor-Like 1 Protein ,Phenotype ,respiratory tract diseases ,GENETIC RISK-FACTORS ,DIFFERENTIATION ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,IL-33 ,INTERLEUKIN-1 RECEPTOR ,Research Article - Abstract
The IL1RL1 (ST2) gene locus is robustly associated with asthma; however, the contribution of single nucleotide polymorphisms (SNPs) in this locus to specific asthma subtypes and the functional mechanisms underlying these associations remain to be defined. We tested for association between IL1RL1 region SNPs and characteristics of asthma as defined by clinical and immunological measures and addressed functional effects of these genetic variants in lung tissue and airway epithelium. Utilizing 4 independent cohorts (Lifelines, Dutch Asthma GWAS [DAG], Genetics of Asthma Severity and Phenotypes [GASP], and Manchester Asthma and Allergy Study [MAAS]) and resequencing data, we identified 3 key signals associated with asthma features. Investigations in lung tissue and primary bronchial epithelial cells identified context-dependent relationships between the signals and IL1RL1 mRNA and soluble protein expression. This was also observed for asthma-associated IL1RL1 nonsynonymous coding TIR domain SNPs. Bronchial epithelial cell cultures from asthma patients, exposed to exacerbation-relevant stimulations, revealed modulatory effects for all 4 signals on IL1RL1 mRNA and/ or protein expression, suggesting SNP-environment interactions. The IL1RL1 TIR signaling domain haplotype affected IL-33-driven NF-κB signaling, while not interfering with TLR signaling. In summary, we identify that IL1RL1 genetic signals potentially contribute to severe and eosinophilic phenotypes in asthma, as well as provide initial mechanistic insight, including genetic regulation of IL1RL1 isoform expression and receptor signaling.
- Published
- 2020
28. Protein-altering germline mutations implicate novel genes related to lung cancer development
- Author
-
Ma'en Obeidat, Richard S. Houlston, Pier Alberto Bertazzi, David C. Nickle, Angela Cecilia Pesatori, Penella J. Woll, Semanti Mukherjee, María Soler Artigas, Rosario Tumino, David C. Christiani, Ivan P. Gorlov, Chu Chen, Sanjay Shete, Anush Mukeria, Yohan Bossé, Lynne R. Wilkens, Hermann Brenner, Yonathan Brhane, Victoria L. Stevens, Matthew B. Schabath, Angeline S. Andrew, Dakai Zhu, Natasha B. Leighl, Nancy Diao, Thomas Muley, Christopher A. Haiman, Shan Zienolddiny, Xuemei Ji, Michael F. Berger, Loic Le Marchand, Adonina Tardón, Mikael Johansson, Frances A. Shepherd, Ghislaine Scelo, Angela Cox, Xiangjun Xiao, Olga Y. Gorlova, Michael W. Marcus, Susanne M. Arnold, Rayjean J. Hung, Stig E. Bojesen, Gad Rennert, Mark E. Robson, William S. Bush, Christopher I. Amos, Neil E. Caporaso, Mattias Johansson, David B. Solit, Simona Ognjanovic, Lesley M. Butler, Hans Brunnström, Jonas Manjer, Stephen J. Chanock, Gary E. Goodman, Jin Hee Kim, Erik H.F.M. van der Heijden, Tadeusz M Orlowski, Ana Fernández-Somoano, Albert Rosenberger, Kjell Grankvist, Kim Overvad, Fiona Taylor, Paul Brennan, Zsofia K. Stadler, Stephen Lam, Lei Song, James McKay, Charles M. Rudin, Anders Mellemgaard, Preethi Srinivasan, Guillermo Fernández-Tardón, Antonia Trichopoulou, Milica Kontic, Philip Lazarus, Younghun Han, Jolanta Lissowska, Wim Timens, Ruyang Zhang, Geoffrey Liu, Ming-Sound Tsao, Ivana Holcatova, John K. Field, John R. McLaughlin, Eric B. Haura, Erich Wichmann, Demetrios Albanes, Ciprian Bolca, Vidar Skaug, Angela Risch, Heike Bickeböller, Robert Carreras-Torres, Konstantin H. Dragnev, Kenneth Offit, Melinda C. Aldrich, Olle Melander, Jakob S Johansen, Li Su, Chaitanya Bandlamudi, David C. Muller, Beata Swiatkowska, David Zaridze, Vladimir Janout, Walid Saliba, Matthew D. Hellmann, Maria Teresa Landi, Philippe Joubert, Jinyoung Byun, Michael P.A. Davies, Lambertus Fa Kiemeney, Per Bakke, Barry S. Taylor, Guided Treatment in Optimal Selected Cancer Patients (GUTS), Groningen Research Institute for Asthma and COPD (GRIAC), Cancer Research UK, and National Institutes of Health
- Subjects
0301 basic medicine ,Male ,Candidate gene ,Lung Neoplasms ,Genotyping Techniques ,General Physics and Astronomy ,Ataxia Telangiectasia Mutated Proteins ,Germline ,Loss of heterozygosity ,0302 clinical medicine ,Risk Factors ,Databases, Genetic ,Odds Ratio ,RNA-Seq ,lcsh:Science ,Oligonucleotide Array Sequence Analysis ,Cancer ,Genetics ,Multidisciplinary ,respiratory system ,Middle Aged ,Pedigree ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,030220 oncology & carcinogenesis ,Female ,Medical Genetics ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Heterozygote ,Science ,Mutation, Missense ,Biology ,Adenocarcinoma ,General Biochemistry, Genetics and Molecular Biology ,White People ,Article ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Germline mutation ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Lung cancer ,Alleles ,Germ-Line Mutation ,Aged ,Medicinsk genetik ,General Chemistry ,medicine.disease ,Lung cancer susceptibility ,Computational biology and bioinformatics ,030104 developmental biology ,Jews ,lcsh:Q - Abstract
The CAPUA study was supported by FIS-FEDER/Spain grant numbers FIS-01/310, FIS-PI03-0365, and FIS-07-BI060604, FICYT/Asturias grant numbers FICYT PB02-67 and FICYT IB09-133, and the University Institute of Oncology (IUOPA), of the University of Oviedo and the Ciber de Epidemiologia y Salud Pública. CIBERESP, SPAIN (...), Ji, X., Mukherjee, S., Landi, M.T., Bosse, Y., Joubert, P., Zhu, D., Gorlov, I., Xiao, X., Han, Y., Gorlova, O., Hung, R.J., Brhane, Y., Carreras-Torres, R., Christiani, D.C., Caporaso, N., Johansson, M., Liu, G., Bojesen, S.E., Le Marchand, L., Albanes, D., Bickeböller, H., Aldrich, M.C., Bush, W.S., Tardon, A., Rennert, G., Chen, C., Byun, J., Dragnev, K.H., Field, J.K., Kiemeney, L.F., Lazarus, P., Zienolddiny, S., Lam, S., Schabath, M.B., Andrew, A.S., Bertazzi, P.A., Pesatori, A.C., Diao, N., Su, L., Song, L., Zhang, R., Leighl, N., Johansen, J.S., Mellemgaard, A., Saliba, W., Haiman, C., Wilkens, L., Fernandez-Somoano, A., Fernandez-Tardon, G., Heijden, E.H.F.M., Kim, J.H., Davies, M.P.A., Marcus, M.W., Brunnström, H., Manjer, J., Melander, O., Muller, D.C., Overvad, K., Trichopoulou, A., Tumino, R., Goodman, G.E., Cox, A., Taylor, F., Woll, P., Wichmann, E., Muley, T., Risch, A., Rosenberger, A., Grankvist, K., Johansson, M., Shepherd, F., Tsao, M.-S., Arnold, S.M., Haura, E.B., Bolca, C., Holcatova, I., Janout, V., Kontic, M., Lissowska, J., Mukeria, A., Ognjanovic, S., Orlowski, T.M., Scelo, G., Swiatkowska, B., Zaridze, D., Bakke, P., Skaug, V., Butler, L.M., Offit, K., Srinivasan, P., Bandlamudi, C., Hellmann, M.D., Solit, D.B., Robson, M.E., Rudin, C.M., Stadler, Z.K., Taylor, B.S., Berger, M.F., Houlston, R., McLaughlin, J., Stevens, V., Nickle, D.C., Obeidat, M., Timens, W., Artigas, M.S., Shete, S., Brenner, H., Chanock, S., Brennan, P., McKay, J.D., Amos, C.I.
- Published
- 2020
29. Genome-Wide Association Study of Susceptibility to Idiopathic Pulmonary Fibrosis
- Author
-
Simon P. Hart, Vilmundur Gudnason, Yingze Zhang, Hiroto Hatabu, Rebecca Braybrooke, R. Gisli Jenkins, Shwu-Fan Ma, Michael Ng, Carlos Flores, George T. O'Connor, Ma'en Obeidat, Nik Hirani, Brian D. Hobbs, Megan L. Paynton, Amy Dressen, Ayodeji Adegunsoye, Helen Booth, Dominic Furniss, Philippe Joubert, Eunice Oballa, Ian Sayers, Martin D. Tobin, Krina T. Zondervan, Richard Hubbard, Xuan Li, Yohan Bossé, John D. Newell, Beatriz Guillen-Guio, Ann B. Millar, Wim Timens, Mary E. Strek, Gunnar Gudmundsson, Philip L. Molyneaux, Gary M. Hunninghake, Ani Manichaikul, Tasha E. Fingerlin, Rachel K. Putman, Richard J. Allen, Vidyia Navaratnam, Maria Molina-Molina, Don D. Sin, Helen Parfrey, Luke M. Kraven, Moira K. B. Whyte, Xuting R. Sheng, Phuwanat Sakornsakolpat, David J. Lederer, David C. Nickle, Ian P. Hall, Brian L. Yaspan, Louise V. Wain, Toby M. Maher, Gauri Saini, Ke Hao, Michael Hill, Naftali Kaminski, Andrew P. Morris, Hanfei Xu, Justin M. Oldham, David A. Schwartz, Robin J. McAnulty, Michael H. Cho, Victor E. Ortega, Margaret Neighbors, Imre Noth, William A. Fahy, Action for Pulmonary Fibrosis, National Institute for Health Research, British Lung Foundation, Groningen Research Institute for Asthma and COPD (GRIAC), Guided Treatment in Optimal Selected Cancer Patients (GUTS), Læknadeild (HÍ), Faculty of Medicine (UI), Heilbrigðisvísindasvið (HÍ), School of Health Sciences (UI), Háskóli Íslands, and University of Iceland
- Subjects
Male ,Pathology ,Epidemiology ,Respiratory System ,Kinesins ,Gene Expression ,Genome-wide association study ,Cell Cycle Proteins ,TOR Serine-Threonine Kinases/metabolism ,VARIANTS ,Critical Care and Intensive Care Medicine ,Idiopathic pulmonary fibrosis ,0302 clinical medicine ,KIF15 ,TELOMERES ,genetics ,030212 general & internal medicine ,Respiratory system ,11 Medical and Health Sciences ,RISK ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,respiratory system ,Middle Aged ,MAD1L1 ,3. Good health ,medicine.anatomical_structure ,SURVIVAL ,epidemiology ,Female ,Life Sciences & Biomedicine ,Kinesin/genetics ,Signal Transduction ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Spindle Apparatus ,DIAGNOSIS ,Risk Assessment ,REGION ,03 medical and health sciences ,Critical Care Medicine ,General & Internal Medicine ,medicine ,Genetics ,Erfðafræði ,Humans ,Genetic Predisposition to Disease ,Aged ,Cell Cycle Proteins/genetics ,Lungnasjúkdómar ,Lung ,Science & Technology ,DEPTOR ,business.industry ,MUTATIONS ,Case-control study ,Editorials ,RTEL1 ,medicine.disease ,Epithelium ,Idiopathic Pulmonary Fibrosis ,respiratory tract diseases ,Genarannsóknir ,030228 respiratory system ,EVENT ,Case-Control Studies ,GAIN ,business ,Intracellular Signaling Peptides and Proteins/genetics ,Idiopathic Pulmonary Fibrosis/genetics ,Genome-Wide Association Study - Abstract
Publisher's version (útgefin grein), Rationale: Idiopathic pulmonary fibrosis (IPF) is a complex lung disease characterized by scarring of the lung that is believed to result from an atypical response to injury of the epithelium. Genome-wide association studies have reported signals of association implicating multiple pathways including host defense, telomere maintenance, signaling, and cell-cell adhesion. Objectives: To improve our understanding of factors that increase IPF susceptibility by identifying previously unreported genetic associations. Methods: We conducted genome-wide analyses across three independent studies and meta-analyzed these results to generate the largest genome-wide association study of IPF to date (2,668 IPF cases and 8,591 controls). We performed replication in two independent studies (1,456 IPF cases and 11,874 controls) and functional analyses (including statistical fine-mapping, investigations into gene expression, and testing for enrichment of IPF susceptibility signals in regulatory regions) to determine putatively causal genes. Polygenic risk scores were used to assess the collective effect of variants not reported as associated with IPF. Measurements and Main Results: We identified and replicated threenewgenome-wide significant (P, R.J.A. is an Action for Pulmonary Fibrosis Research Fellow. L.V.W. holds a GSK/British Lung Foundation Chair in Respiratory Research. R.G.J. is supported by a National Institute for Health Research (NIHR) Research Professorship (NIHR reference RP-2017-08-ST2-014). I.N. is supported by the NHLBI (R01HL130796). B.G.-G. is funded by Agencia Canaria de Investigación, Innovación y Sociedad de la Información (TESIS2015010057) cofunded by European Social Fund. J.M.O. is supported by the NHLBI (K23HL138190). C.F. is supported by the Spanish Ministry of Science, Innovation and Universities (grant RTC-2017-6471-1; Ministerio de Ciencia e Innovacion/Agencia Estatal de Investigación/Fondo Europeo de Desarrollo Regional, Unión Europea) cofinanced by the European Regional Development Funds “A way of making Europe” from the European Union and by agreement OA17/008 with Instituto Tecnológico y de Energías Renovables to strengthen scientific and technological education, training, research, development and innovation in Genomics, Personalized Medicine and Biotechnology. The Spain Biobank array genotyping service was performed at CEGEN-PRB3-ISCIII, which is supported by PT17/0019, of the PE I+D+i 2013–2016, funded by Instituto de Salud Carlos III, and cofinanced by the European Regional Development Funds. P.L.M. is an Action for Pulmonary Fibrosis Research Fellow. M.O. is a fellow of the Parker B. Francis Foundation and a Scholar of the Michael Smith Foundation for Health Research. B.D.H. is supported by NIH K08 HL136928, Parker B. Francis Research Opportunity Award. M.H.C. and G.M.H. are supported by NHLBI grants R01HL113264 (M.H.C.), R01HL137927 (M.H.C.), R01HL135142 (M.H.C. and G.M.H.), R01111024 (G.M.H.), and R01130974 (G.M.H.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The funding body has no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript. T.M.M. is supported by an NIHR Clinician Scientist Fellowship (NIHR Ref: CS-2013-13-017) and a British Lung Foundation Chair in Respiratory Research (C17-3). M.D.T. is supported by a Wellcome Trust Investigator Award (WT202849/Z/16/Z). The research was partially supported by the NIHR Leicester Biomedical Research Centre; the views expressed are those of the author(s) and not necessarily those of the National Health Service (NHS), the NIHR, or the Department of Health. I.P.H. was partially supported by the NIHR Nottingham Biomedical Research Centre; the views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR, or the Department of Health. I.S. is supported by Medical Research Council (G1000861) and Asthma UK (AUK-PG-2013-188). D.F. was supported by an Intermediate Fellowship from the Wellcome Trust (097152/Z/11/Z). This work was partially supported by the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre. V.N. is funded by an NIHR Clinical Lectureship. G.G. is supported by project grant 141513-051 from the Icelandic Research Fund and Landspitali Scientific Fund A-2016-023, A-2017-029, and A-2018-025. D.J.L. and A.M. are supported by Multi-Ethnic Study of Atherosclerosis (MESA) and the MESA SNP Health Association Resource (SHARe) project are conducted and supported by the NHLBI in collaboration with MESA investigators. Support for MESA is provided by contracts HHSN268201500003I, N01-HC-95159, N01-HC-95160, N01-HC-95161, N01-HC-95162, N01-HC-95163, N01-HC-95164, N01-HC-95165, N01-HC-95166, N01-HC-95167, N01-HC-95168, N01-HC-95169, UL1-TR-000040, UL1-TR-001079, UL1-TR-001420, UL1-TR-001881, and DK063491. Funding for SHARe genotyping was provided by NHLBI Contract N02-HL-64278. Genotyping was performed at Affymetrix (Santa Clara, California) and the Broad Institute of Harvard and Massachusetts Institute of Technology (Boston, Massachusetts) using the Affymetrix Genome-Wide Human SNP Array 6.0. This work was supported by NIH grants R01 HL131565 (A.M.), R01 HL103676 (D.J.L.), and R01 HL137234 (D.J.L.).
- Published
- 2020
30. Lung tissue gene-expression signature for the ageing lung in COPD
- Author
-
Roy R Woldhuis, Don D. Sin, Yohan Bossé, Corry-Anke Brandsma, Wim Timens, Tristan V. de Jong, Maarten van den Berge, Victor Guryev, David C. Nickle, Alen Faiz, Dirkje S. Postma, Maaike de Vries, Groningen Research Institute for Asthma and COPD (GRIAC), Stem Cell Aging Leukemia and Lymphoma (SALL), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Senescence ,Candidate gene ,HOMEOSTASIS ,Respiratory System ,PHENOTYPES ,OBSTRUCTIVE PULMONARY-DISEASE ,PATHWAY ,03 medical and health sciences ,Exon ,Gene expression ,Medicine ,CIRCULATING LEUKOCYTES ,XEDAR ,Gene ,HALLMARKS ,COPD ,Lung ,business.industry ,MTOR ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,medicine.anatomical_structure ,Ageing ,SENESCENCE ,Immunology ,JUNCTION ,Copd u Mechanisms ,business - Abstract
IntroductionCOPD is a chronic, progressive, inflammatory disease of the lungs and the third leading cause of death worldwide. The current knowledge of the pathophysiology of COPD is limited and novel insights in underlying disease mechanisms are urgently needed. Since there are clear parallels between ageing and COPD, we investigated genes underlying lung ageing in general and abnormal lung ageing in COPD.MethodsWhole genome mRNA profiling was performed on lung tissue samples (n=1197) and differential gene expression with increasing age was analysed using an adjusted linear regression model. Subsequent pathway analysis was performed using GeneNetwork and the gene-expression signature was compared with lung ageing in the Genotype-Tissue Expression (GTEx) project. In a subset of patients with COPD (n=311) and non-COPD controls (n=270), we performed an interaction analysis between age and COPD to identify genes differentially expressed with age in COPD compared with controls, followed by gene set enrichment pathway analysis.ResultsWe identified a strong gene-expression signature for lung ageing with 3509 differentially expressed genes, of which 33.5% were found nominal significant in the GTEx project. Interestingly, we foundEDA2Ras a strong candidate gene for lung ageing. The age*COPD interaction analysis revealed 69 genes significantly differentially expressed with age between COPD and controls.ConclusionsOur study indicates that processes related to lung development, cell-cell contacts, calcium signalling and immune responses are involved in lung ageing in general. Pathways related to extracellular matrix, mammalian target of rapamycin signalling, splicing of introns and exons and the ribosome complex are proposed to be involved in abnormal lung ageing in COPD.
- Published
- 2018
31. Responsiveness to Ipratropium Bromide in Male and Female Patients with Mild to Moderate Chronic Obstructive Pulmonary Disease
- Author
-
Robert A. Wise, Xuan Li, John E. Connett, M. Obeidat, Guohai Zhou, Ke Hao, Philippe Joubert, Donald P. Tashkin, Corry-Anke Brandsma, David C. Nickle, Yohan Bossé, Don D. Sin, Maarten van den Berge, Peter D. Paré, Janice M. Leung, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Male ,lcsh:Medicine ,EMPHYSEMA ,Ipratropium bromide ,Gastroenterology ,FEV1 ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Bronchodilator ,030212 general & internal medicine ,Precision Medicine ,Lung ,BRONCHODILATOR ,2. Zero hunger ,COPD ,lcsh:R5-920 ,Ipratropium ,General Medicine ,respiratory system ,Bronchodilator Agents ,medicine.anatomical_structure ,Treatment Outcome ,Sex ,Female ,lcsh:Medicine (General) ,SMOKERS ,medicine.drug ,Research Paper ,medicine.medical_specialty ,SEX-DIFFERENCES ,medicine.drug_class ,Muscarinic Antagonists ,LUNG HEALTH ,Placebo ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Asthma ,GENDER-DIFFERENCES ,business.industry ,lcsh:R ,AIRWAY HYPERRESPONSIVENESS ,DIFFERENCE ,medicine.disease ,respiratory tract diseases ,Endocrinology ,030228 respiratory system ,ASTHMA ,Gene expression ,business ,Body mass index - Abstract
Introduction Although the prevalence of chronic obstructive pulmonary disease (COPD) is similar between men and women, current evidence used to support bronchodilator therapy has been generated in therapeutic trials that have predominately enrolled male patients. Here, we determined whether there is any significant sex-related differences in FEV1 responses to ipratropium bromide. Methods Data from the Lung Health Study (n = 5887; 37% females) were used to determine changes in FEV1 with ipratropium or placebo in male and female subjects with mild to moderate COPD over 5 years. Lung Expression Quantitative Trait Loci (eQTL) dataset was used to determine whether there were any sex-related differences in gene expression for muscarinic (M2 and M3) receptors in lungs of male and female patients. Results After 4 months, ipratropium therapy increased FEV1 by 6.0% in female and 2.9% in male subjects from baseline values (p = 2.42 × 10− 16). This effect was modified by body mass index (BMI) such that the biggest improvements in FEV1 with ipratropium were observed in thin female subjects (p for BMI ∗ sex interaction = 0.044). The sex-related changes in FEV1 related to ipratropium persisted for 2 years (p = 0.0134). Female compared with male lungs had greater gene expression for M3 relative to M2 receptors (p = 6.86 × 10− 8). Conclusion Ipratropium induces a larger bronchodilator response in female than in male patients and the benefits are particularly notable in non-obese females. Female lungs have greater gene expression for the M3 muscarinic receptor relative to M2 receptors than male lungs. Female patients are thus more likely to benefit from ipratropium than male COPD patients., Highlights • Ipratropium; a muscarinic antagonist bronchodilator is more effective in female COPD patients compared to males. • The effect was modified by body mass index (BMI) such that thin female subjects respond better. • Female compared with male lungs had greater gene expression for the M3/M2 ratio of muscarinic receptors. Most evidence used to support bronchodilator therapy in COPD has been generated in therapeutic trials with predominately male patients. Here, we determined whether there are any significant sex-related differences in lung function responses to the bronchodilator ipratropium bromide. After 4 months, ipratropium therapy increased lung function in females twice as much as males. This effect was modified by body mass index (BMI) such that the biggest improvements in lung function with ipratropium were observed in thin female subjects. Female compared with male lungs had greater gene expression for ipratropium receptors. Female patients are likely to benefit more from ipratropium than male COPD patients.
- Published
- 2017
32. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets
- Author
-
Stefan Enroth, Robert J. Hall, James D. Crapo, Frederick E. Dewey, Victoria E. Jackson, Eleftheria Zeggini, Gudmar Thorleifsson, Terho Lehtimäki, David J. Porteous, Nicole Probst-Hensch, Ian J. Deary, A. Mesut Erzurumluoglu, Bram P. Prins, Maarten van den Berge, Craig E. Pennell, Catherine John, Terri H. Beaty, Claudia Schurmann, Michael H. Cho, Veronique Vitart, Erwin P. Bottinger, Carol A. Wang, H. Lester Kirchner, Medea Imboden, David J. Carey, Archie Campbell, Kari Stefansson, Matthias Wielscher, Ida Surakka, Igor Rudan, Shona M. Kerr, Holger Schulz, Michael A. Portelli, Thorarinn Gislason, Peter D. Paré, James F. Wilson, Boris Noyvert, Beate Stubbe, Zhengming Chen, Ruth J. F. Loos, Ma'en Obeidat, María Soler Artigas, Philippe Joubert, Kathleen C. Barnes, Christian Gieger, Andrew P. Morris, Rajesh Rawal, Yohan Bossé, Peter K. Joshi, Nadia N. Hansel, Emily S. Wan, David M. Evans, David C. Nickle, Caroline Hayward, Stefan Karrasch, Ke Hao, Tracy L. Rimington, Don D. Sin, Alan James, Stefan Jonsson, Shannon Bruse, Amanda P. Henry, Iona Y Millwood, Lara Bossini-Castillo, Ian P. Hall, David Sparrow, Ulf Gyllensten, Ian Sayers, Edwin K. Silverman, Robert Busch, David P. Strachan, Martin D. Tobin, Nick Shrine, Louise V. Wain, Robin G. Walters, Ingileif Jonsdottir, Peter D. Sly, Liming Li, Charlotte K. Billington, Anna Hansell, Omri Gottesman, Om P Kurmi, Ingo Ruczinski, Nicholas J. Wareham, Amund Gulsvik, Per Bakke, Jennie Hui, Corry-Anke Brandsma, Gosia Trynka, Anthony G. Fenech, Brian D. Hobbs, A. John Henderson, Jonathan Marten, Olli T. Raitakari, Richard J. Allen, Augusto A. Litonjua, Sarah E. Harris, Ozren Polasek, Mika Kähönen, Tineka Blake, Marjo-Riitta Järvelin, Rasika A. Mathias, Jing Hua Zhao, Julien Vaucher, Girish N. Nadkarni, Christopher E. Brightling, Groningen Research Institute for Asthma and COPD (GRIAC), Wareham, Nicholas [0000-0003-1422-2993], Zhao, Jing Hua [0000-0003-4930-3582], Marten, Jonathan [0000-0001-6916-2014], and Apollo - University of Cambridge Repository
- Subjects
Male ,0301 basic medicine ,Oncology ,Genome-wide association study ,heart disease ,VARIANTS ,SUSCEPTIBILITY ,Epigenesis, Genetic ,AIR-FLOW OBSTRUCTION ,Pulmonary Disease, Chronic Obstructive ,Risk Factors ,HISTORY ,GWAS ,EPIDEMIOLOGY ,Lung ,POPULATION ,Cause of death ,Genetics & Heredity ,Aged, 80 and over ,education.field_of_study ,COPD ,Framingham Risk Score ,Chronic obstructive pulmonary disease ,Heart ,11 Medical And Health Sciences ,Middle Aged ,3. Good health ,Female ,HEALTH ,Life Sciences & Biomedicine ,Adult ,medicine.medical_specialty ,Population ,genome-wide, lungs, target ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,education ,Alleles ,Aged ,Asthma ,Science & Technology ,ta1184 ,Odds ratio ,06 Biological Sciences ,ta3121 ,medicine.disease ,Lung function ,respiratory tract diseases ,030104 developmental biology ,Genetic Loci ,FAM13A ,Immunology ,Lungs ,Pulmonary disease ,Developmental Biology ,Genome-Wide Association Study - Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by reduced lung function and is the third leading cause of death globally. Through genome-wide association discovery in 48,943 individuals, selected from extremes of the lung function distribution in UK Biobank, and follow-up in 95,375 individuals, we increased the yield of independent signals for lung function from 54 to 97. A genetic risk score was associated with COPD susceptibility (odds ratio per 1 s.d. of the risk score (similar to 6 alleles) (95% confidence interval) = 1.24 (1.20-1.27), P = 5.05 x 10(-49)), and we observed a 3.7-fold difference in COPD risk between individuals in the highest and lowest genetic risk score deciles in UK Biobank. The 97 signals show enrichment in genes for development, elastic fibers and epigenetic regulation pathways. We highlight targets for drugs and compounds in development for COPD and asthma (genes in the inositol phosphate metabolism pathway and CHRM3) and describe targets for potential drug repositioning from other clinical indications.
- Published
- 2017
33. Genome-wide association study on the FEV1/FVC ratio in never-smokers identifies HHIP and FAM13A
- Author
-
Corry-Anke Brandsma, Don D. Sin, Alen Faiz, Diana A van der Plaat, Ivana Nedeljkovic, Dirkje S. Postma, Lies Lahousse, Judith M. Vonk, Kim de Jong, Cornelia M. van Duijn, David C. Nickle, Cleo C. van Diemen, H. Marike Boezen, Yohan Bossé, Guy Brusselle, Najaf Amin, Albert Hofman, Epidemiology, Pulmonary Medicine, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), and Life Course Epidemiology (LCE)
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Genome-wide association study ,GENETICS ,Immunology ,LEVEL ,Single-nucleotide polymorphism ,never-smokers ,Biology ,Bioinformatics ,OBSTRUCTIVE PULMONARY-DISEASE ,chronic obstructive pulmonary disease ,03 medical and health sciences ,FEV1/FVC ratio ,Internal medicine ,medicine ,Genetic predisposition ,Immunology and Allergy ,COPD ,Risk factor ,POLYMORPHISMS ,METAANALYSIS ,Genetic association ,GENERAL-POPULATION ,pulmonary function ,STANDARDIZATION ,ROTTERDAM ,respiratory tract diseases ,LUNG-FUNCTION ,030104 developmental biology ,Expression quantitative trait loci ,Hedgehog interacting protein - Abstract
Background: Although a striking proportion (25% to 45%) of patients with chronic obstructive pulmonary disease are never-smokers, most genetic susceptibility studies have not focused on this group exclusively.Objective: The aim of this study was to identify common genetic variants associated with FEV1 and its ratio to forced vital capacity (FVC) in never-smokers.Methods: Genome-wide association studies were performed in 5070 never-smokers of the identification cohort LifeLines, and results (P Results: We identified associations between the FEV1/ FVC ratio and 5 common genetic variants in the identification cohort, and 2 of these associations were replicated. The 2 variants annotated to the genes hedgehog interacting protein (HHIP) and family with sequence similarity 13 member A (FAM13A) were shown to have an additive effect on FEV1/ FVC levels in the genetic risk score analysis; were associated with gene expression of HHIP and FAM13A in lung tissue, respectively; and were genome-wide significant in a meta-analysis including both identification and 4 verification cohorts (P Conclusion: The genes HHIP and FAM13A confer a risk for airway obstruction in general that is not driven exclusively by cigarette smoking, which is the main risk factor for chronic obstructive pulmonary disease.
- Published
- 2017
34. Genome-wide association study of susceptibility to idiopathic pulmonary fibrosis
- Author
-
George T. O'Connor, Gauri Saini, Helen Booth, Dominic Furniss, William A. Fahy, Ke Hao, Beatriz Guillen-Guio, Vilmundur Gudnason, Brian L. Yaspan, Imre Noth, Hanfei Xu, Hiroto Hatabu, Brian D. Hobbs, Ma'en Obeidat, Nik Hirani, Justin M. Oldham, Victor E. Ortega, David A. Schwartz, Simon P. Hart, Ani Manichaikul, Richard Hubbard, Xuan Li, Philippe Joubert, Michael Hill, Michael Ng, Ann B. Millar, Vidyia Navaratnam, Richard J. Allen, Rebecca Braybrooke, Ian Sayers, Ian P. Hall, Shwu-Fan Ma, Philip L. Molyneaux, Carlos Flores, Tasha E. Fingerlin, Martin D. Tobin, Gunnar Gudmundsson, Rachel K. Putman, Gary M. Hunninghake, Krina T. Zondervan, Moira K. B. Whyte, Robin J. McAnulty, Louise V. Wain, Andrew P. Morris, Michael H. Cho, David C. Nickle, Don D. Sin, Helen Parfrey, Toby M. Maher, Yohan Bossé, Maria Molina-Molina, John D. Newell, Eunice Oballa, Phuwanat Sakornsakolpat, David J. Lederer, R. Gisli Jenkins, Amy Dressen, Wim Timens, Luke M. Kraven, and Megan L. Paynton
- Subjects
0303 health sciences ,Lung ,business.industry ,Genome-wide association study ,Disease ,respiratory system ,medicine.disease ,DEPTOR ,MAD1L1 ,respiratory tract diseases ,3. Good health ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,0302 clinical medicine ,medicine.anatomical_structure ,030228 respiratory system ,Immunology ,Medicine ,business ,Gene ,030304 developmental biology ,Genetic association - Abstract
RationaleIdiopathic pulmonary fibrosis (IPF) is a complex lung disease characterised by scarring of the lung that is believed to result from an atypical response to injury of the epithelium. The mechanisms by which this arises are poorly understood and it is likely that multiple pathways are involved. The strongest genetic association with IPF is a variant in the promoter of MUC5B where each copy of the risk allele confers a five-fold risk of disease. However, genome-wide association studies have reported additional signals of association implicating multiple pathways including host defence, telomere maintenance, signalling and cell-cell adhesion.ObjectivesTo improve our understanding of mechanisms that increase IPF susceptibility by identifying previously unreported genetic associations.Methods and measurementsWe performed the largest genome-wide association study undertaken for IPF susceptibility with a discovery stage comprising up to 2,668 IPF cases and 8,591 controls with replication in an additional 1,467 IPF cases and 11,874 controls. Polygenic risk scores were used to assess the collective effect of variants not reported as associated with IPF.Main resultsWe identified and replicated three new genome-wide significant (P-8) signals of association with IPF susceptibility (near KIF15, MAD1L1 and DEPTOR) and confirm associations at 11 previously reported loci. Polygenic risk score analyses showed that the combined effect of many thousands of as-yet unreported IPF risk variants contribute to IPF susceptibility.ConclusionsNovel association signals support the importance of mTOR signalling in lung fibrosis and suggest a possible role of mitotic spindle-assembly genes in IPF susceptibility.
- Published
- 2019
35. Shared Single Nucleotide Polymorphisms Regulate Gene Expression of Macrophage Migration Inhibitory Factor and D-Dopachrome Tautomerase-Like Protein in Lung Tissue
- Author
-
L. Florez Sampedro, Alen Faiz, Ma'en Obeidat, M. de Vries, Corry-Anke Brandsma, Gerrit J. Poelarends, Philippe Joubert, Wim Timens, Barbro N. Melgert, and David C. Nickle
- Subjects
business.industry ,Gene expression ,Medicine ,Macrophage migration inhibitory factor ,Single-nucleotide polymorphism ,D-DOPACHROME TAUTOMERASE ,business ,Lung tissue ,Molecular biology - Published
- 2019
36. Hedgehog signaling in the airway epithelium of patients with chronic obstructive pulmonary disease
- Author
-
Cheng Wei Tony Yang, Gurpreet K. Singhera, Don D. Sin, M. Obeidat, Kelly M. McNagny, Tillie L. Hackett, A. Tam, Tawimas Shaipanich, Janice M. Leung, Peter D. Paré, Michael R. Hughes, David C. Nickle, D. R. Dorscheid, and James C. Hogg
- Subjects
Adult ,Male ,0301 basic medicine ,Patched ,endocrine system ,Pathology ,medicine.medical_specialty ,Chronic bronchitis ,lcsh:Medicine ,Bronchi ,Epithelium ,Article ,Mice ,Pulmonary Disease, Chronic Obstructive ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Medicine ,Gene Silencing ,lcsh:Science ,Aged ,Mice, Knockout ,COPD ,Gene knockdown ,Multidisciplinary ,Lung ,business.industry ,lcsh:R ,Middle Aged ,respiratory system ,medicine.disease ,respiratory tract diseases ,Patched-1 Receptor ,030104 developmental biology ,medicine.anatomical_structure ,PTCH1 ,Respiratory epithelium ,lcsh:Q ,Female ,business ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Genome-wide association studies have linked gene variants of the receptor patched homolog 1 (PTCH1) with chronic obstructive pulmonary disease (COPD). However, its biological role in the disease is unclear. Our objective was to determine the expression pattern and biological role of PTCH1 in the lungs of patients with COPD. Airway epithelial-specific PTCH1 protein expression and epithelial morphology were assessed in lung tissues of control and COPD patients. PTCH1 mRNA expression was measured in bronchial epithelial cells obtained from individuals with and without COPD. The effects of PTCH1 siRNA knockdown on epithelial repair and mucous expression were evaluated using human epithelial cell lines. Ptch1+/− mice were used to assess the effect of decreased PTCH1 on mucous expression and airway epithelial phenotypes. Airway epithelial-specific PTCH1 protein expression was significantly increased in subjects with COPD compared to controls, and its expression was associated with total airway epithelial cell count and thickness. PTCH1 knockdown attenuated wound closure and mucous expression in airway epithelial cell lines. Ptch1+/− mice had reduced mucous expression compared to wildtype mice following mucous induction. PTCH1 protein is up-regulated in COPD airway epithelium and may upregulate mucous expression. PTCH1 provides a novel target to reduce chronic bronchitis in COPD patients.
- Published
- 2019
37. Susceptibility genes for lung diseases in the major histocompatibility complex revealed by lung expression quantitative trait loci analysis
- Author
-
Don D. Sin, Ke Hao, Philippe Joubert, Michel Laviolette, Yohan Bossé, Wim Timens, Dirkje S. Postma, David C. Nickle, Maxime Lamontagne, Peter D. Paré, Groningen Research Institute for Asthma and COPD (GRIAC), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
Lung Diseases ,Male ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Quantitative Trait Loci ,Quantitative trait locus ,Major histocompatibility complex ,Polymorphism, Single Nucleotide ,Major Histocompatibility Complex ,03 medical and health sciences ,0302 clinical medicine ,Pulmonary fibrosis ,medicine ,Humans ,Genetic Predisposition to Disease ,Lung cancer ,Idiopathic interstitial pneumonia ,Asthma ,Lung ,biology ,business.industry ,PULMONARY-FIBROSIS ,respiratory system ,medicine.disease ,3. Good health ,respiratory tract diseases ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Expression quantitative trait loci ,Immunology ,biology.protein ,Female ,business - Abstract
The major histocompatibility complex (MHC) has been linked with hundreds of diseases [1]. The MHC is one of the most complex regions of the human genome, because of the high gene density, extended linkage disequilibrium (LD) and sequence diversity [2]. Recent genome-wide association studies (GWAS) have identified polymorphisms located in the MHC that are associated with lung diseases and related traits: asthma, cystic fibrosis, idiopathic interstitial pneumonia, lung cancer and lung function. However, due to the limitations of GWAS and tissue-specific characteristics of gene expression [3], the causal genes and genetic mechanisms mediating the heritable risk within this locus remain to be found.
- Published
- 2016
38. HCV evolutionary genetics of SVR versus virologic failure assessed from the vaniprevir phase III registration trials
- Author
-
Keisuke Nakamura, Wei Chang, Stuart Black, Steven W. Ludmerer, Anita Y. M. Howe, Tomona Hirano, Akiko Takase, David C. Nickle, Norio Hayashi, Yoshiyuki Tanaka, and Hiromitsu Kumada
- Subjects
Cyclopropanes ,0301 basic medicine ,Oncology ,Indoles ,Vaniprevir ,Hepacivirus ,Resistance ,Isoindoles ,medicine.disease_cause ,chemistry.chemical_compound ,0302 clinical medicine ,Pegylated interferon ,Genotype ,Treatment Failure ,Phylogeny ,Sulfonamides ,Mutation ,biology ,Hepatitis C ,Treatment Outcome ,HCV ,Drug Therapy, Combination ,030211 gastroenterology & hepatology ,medicine.drug ,medicine.medical_specialty ,Proline ,Lactams, Macrocyclic ,Antiviral Agents ,Evolution, Molecular ,03 medical and health sciences ,Leucine ,Virology ,Internal medicine ,Drug Resistance, Viral ,medicine ,Humans ,Codon ,Pharmacology ,Human evolutionary genetics ,business.industry ,Ribavirin ,Sequence Analysis, DNA ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,Amino Acid Substitution ,chemistry ,business - Abstract
In the phase III registration studies conducted in Japan, Japanese HCV gt1 patients administered vaniprevir 300 mg twice daily plus pegylated interferon/ribavirin for 12 or 24 weeks achieved SVR24 rates of 83.7–84.5% among treatment-naive patients, and 92.0–96.2% and 61.9% among breakthrough/relapsers or null-responders to prior interferon based therapy. As evidenced by direct sequencing, patients who did not achieve SVR24 principally failed due to treatment-emerging mutations at D168 or in a few cases R155. In this work, additional sequence analysis was conducted to address whether there were baseline polymorphisms associated with failure, evaluate the persistence of resistant virus among treatment failures, and assess for evidence of second site co-evolution with R155 or D168 mutations. To accomplish this, clonal sequencing (up to 40 clones per sample) was conducted on baseline, failure, and follow-up samples from all 38 patients among the vaniprevir treatment arms who met virologic failure criteria (37 gt1b, 1 gt1a, herein referred to as virologic failures) and baseline samples from 41 vaniprevir-treated SVR24 patients (all gt1b) selected among the three studies. SVR24 and virologic failure patients showed similar distributions of baseline polymorphisms previously associated with failure to one or more protease inhibitors. Furthermore, there was no evidence for baseline polymorphisms or a genetic signature across the NS3 protease domain specific to virologic failure patients, and which distinguishes them from baseline SVR24 sequences beyond a chance distribution. 24 of 32 virologic failures for whom baseline, failure, and follow-up samples were available showed reduced prevalence of the resistant virus first observed at the time of failure during the protocol-defined follow-up period of 24 weeks. Finally, pairwise analysis using either alignment or phylogenetic based methodologies provided no evidence for second site evolution with either the R155 or D168 mutations attributed to failure. This work supports and extends earlier findings based upon direct sequencing that attributed virologic failure to vaniprevir in the Phase III studies solely to the emergence of R155 or D168 mutations, with no apparent influence by other residues within the NS3 protease domain on treatment outcome. ClinicalTrials.govIdentifiers NCT01370642 , NCT01405937 , NCT01405560
- Published
- 2016
39. Abstract CT247: A phase 1/2 study of GB1275, a first-in-class CD11b modulator, as monotherapy and with an anti-PD-1 antibody in specified advanced solid tumors or with chemotherapy in metastatic pancreatic cancer (KEYNOTE-A36)
- Author
-
Eileen M. O'Reilly, Debbie Slee, Jack Shiansong Li, Beatrice Ferguson, Wungki Park, Laura L. Carter, Marya F. Chaney, Andrea Wang-Gillam, Jakob Dupont, Lei Zhou, David C. Nickle, Drew W. Rasco, Johanna C. Bendell, Rebecca Tran, Wells A. Messersmith, David G. DeNardo, Anna Galkin, Luisa Salter-Cid, and Vineet Gupta
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Tumor microenvironment ,Myeloid ,Colorectal cancer ,business.industry ,medicine.medical_treatment ,Immunosuppression ,Pembrolizumab ,medicine.disease ,Gemcitabine ,03 medical and health sciences ,Regimen ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Background: Tumor influx of CD11b-expressing myeloid-derived suppressor cells (MDSCs) and M2 tumor-associated macrophages (TAMs) creates an immunosuppressive tumor microenvironment associated with resistance to anti-PD-1 antibody therapy. GB1275 is a novel, first-in-class, CD11b modulator that, in vivo, reduced MDSCs and TAMs at the tumor site, repolarized M2 immunosuppressive TAMs to an M1 phenotype, and increased tumor infiltration of activated CD8+ T cells. In combination with an anti-PD-1 antibody or chemotherapy, these immunomodulatory effects translated into potent anti-tumor effects and prolonged survival in orthotopic PDAC models. We hypothesize that GB1275-based therapy can alleviate myeloid cell-mediated immunosuppression and improve cancer treatment outcomes. Methods: This is an open-label, first-in-human study comprising phase 1 dose escalation of GB1275 monotherapy (Regimen A) and GB1275 plus pembrolizumab (Regimen B) in patients with previously treated, locally advanced or metastatic PDA, esophageal, gastric/GEJ, triple negative breast, castration-resistant prostate, or microsatellite-stable colorectal cancer (MSS CRC), and GB1275 plus nab-paclitaxel and gemcitabine (Nab-P+Gem) (Regimen C) in mPDAC. This is followed by phase 2 expansion in three disease cohorts: 1) newly diagnosed stage IV mPDAC (GB1275+Nab-P+Gem), 2) MSS CRC (GB1275+pembrolizumab), and 3) PD-L1+ gastric/GEJ cancer (GB1275+pembrolizumab). Patients are enrolled in Regimen A initially, with Regimen B commencing after completion of the first few cohorts of Regimen A. Regimen C will initiate when Regimen A is completed. Eligible patients are ≥18 years of age, with histologically confirmed locally advanced/metastatic tumor specified and ECOG 0-1 PS; prior immunotherapy is permissible during Regimen A and B dose escalation, but not Regimen C or during the expansion phase. Patients are excluded for untreated or symptomatic CNS metastasis, prior myeloid targeting treatment or other prohibited medications, or a history of clinically significant cardiovascular disease. Patients with active autoimmune disease requiring systemic therapy will be excluded from Regimen B and from expansion cohorts 2 and 3. Primary objectives for phase 1 are to determine the maximum tolerated dose/recommended phase dose and pharmacokinetic profile of GB1275 alone and combination with pembrolizumab, and safety in combination with Nab-P+Gem, and for phase 2, to assess efficacy.Statistical Considerations: A 3+3 design is used for the dose escalation phase and Simon's 2-stage design for the expansion phase. Adverse events are graded per CTCAE v5.0, responses per RECIST v1.1. This study is open for recruitment (NCT04060342). Citation Format: Andrea Wang-Gillam, Drew W. Rasco, Wungki Park, Eileen O'Reilly, Wells Messersmith, David G. DeNardo, Vineet Gupta, Lei Zhou, Anna Galkin, Debbie Slee, Laura L. Carter, David Nickle, Rebecca Tran, Jack Li, Beatrice Ferguson, Marya F. Chaney, Luisa Salter-Cid, Jakob Dupont, Johanna C. Bendell. A phase 1/2 study of GB1275, a first-in-class CD11b modulator, as monotherapy and with an anti-PD-1 antibody in specified advanced solid tumors or with chemotherapy in metastatic pancreatic cancer (KEYNOTE-A36) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT247.
- Published
- 2020
40. Genome-wide interaction study of gene-by-occupational exposures on respiratory symptoms
- Author
-
Hans Kromhout, Corry-Anke Brandsma, David C. Nickle, Peter D. Paré, Xijin Xu, Judith M. Vonk, Alen Faiz, Roel Vermeulen, Kim de Jong, H. Marike Boezen, Diana A van der Plaat, Philippe Joubert, Xiang Zeng, Xia Huo, Groningen Research Institute for Asthma and COPD (GRIAC), Life Course Epidemiology (LCE), and One Health Chemisch
- Subjects
010504 meteorology & atmospheric sciences ,Interaction ,PROTEINS ,Population ,Respiratory Tract Diseases ,Environmental Sciences & Ecology ,Single-nucleotide polymorphism ,010501 environmental sciences ,01 natural sciences ,Polymorphism, Single Nucleotide ,Cohort Studies ,Genome wide ,IGE LEVELS ,Occupational Exposure ,MD Multidisciplinary ,medicine ,Genetics ,Humans ,Respiratory symptoms ,Respiratory system ,education ,lcsh:Environmental sciences ,POLYMORPHISMS ,POPULATION ,0105 earth and related environmental sciences ,General Environmental Science ,Asthma ,lcsh:GE1-350 ,education.field_of_study ,Science & Technology ,WORKERS ,AIR-POLLUTION ,ASSOCIATION ,Occupational exposure ,medicine.disease ,FCER1A ,LUNG-FUNCTION ,TOX3 ,Expression quantitative trait loci ,Immunology ,Cohort ,ASTHMA ,Life Sciences & Biomedicine ,IMMUNOGLOBULIN-E ,Environmental Sciences ,Genome-Wide Association Study - Abstract
Respiratory symptoms are important indicators of respiratory diseases. Both genetic and environmental factors contribute to respiratory symptoms development but less is known about gene-environment interactions. We aimed to assess interactions between single nucleotide polymorphisms (SNPs) and occupational exposures on respiratory symptoms cough, dyspnea and phlegm. As identification cohort LifeLines I (n = 7976 subjects) was used. Job-specific exposure was estimated using the ALOHA + job exposure matrix. SNP-by-occupational exposure interactions on respiratory symptoms were tested using logistic regression adjusted for gender, age, and current smoking. SNP-by-exposure interactions with a p-value
- Published
- 2018
41. The Overlap of Lung Tissue Transcriptome of Smoke Exposed Mice with Human Smoking and COPD
- Author
-
Peter D. Paré, Philip M. Hansbro, Martin R. Stämpfli, Ma'en Obeidat, Yohan Bossé, Corry-Anke Brandsma, Alvar Agusti, Don D. Sin, Anna Dvorkin-Gheva, David C. Nickle, Xuan Li, Rosa Faner, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
0301 basic medicine ,lcsh:Medicine ,Article ,Pathogenesis ,Transcriptome ,03 medical and health sciences ,Mice ,Pulmonary Disease, Chronic Obstructive ,Hàbit de fumar ,0302 clinical medicine ,Tabac ,Smoke ,Tobacco ,Gene expression ,medicine ,Animals ,Humans ,Chronic obstructive pulmonary diseases ,lcsh:Science ,Gene ,Lung ,Malalties pulmonars obstructives cròniques ,COPD ,Multidisciplinary ,business.industry ,lcsh:R ,Smoking ,Gene signature ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,Expression quantitative trait loci ,Immunology ,RNA ,lcsh:Q ,business - Abstract
Genome-wide mRNA profiling in lung tissue from human and animal models can provide novel insights into the pathogenesis of chronic obstructive pulmonary disease (COPD). While 6 months of smoke exposure are widely used, shorter durations were also reported. The overlap of short term and long-term smoke exposure in mice is currently not well understood, and their representation of the human condition is uncertain. Lung tissue gene expression profiles of six murine smoking experiments (n = 48) were obtained from the Gene Expression Omnibus (GEO) and analyzed to identify the murine smoking signature. The “human smoking” gene signature containing 386 genes was previously published in the lung eQTL study (n = 1,111). A signature of mild COPD containing 7 genes was also identified in the same study. The lung tissue gene signature of “severe COPD” (n = 70) contained 4,071 genes and was previously published. We detected 3,723 differentially expressed genes in the 6 month-exposure mice datasets (FDR −26) and a 1.4 fold in the severe COPD -related genes (P = 2.3 × 10−12). There was no significant enrichment of the mice and human smoking-related genes in mild COPD signature. These data suggest that murine smoke models are strongly representative of molecular processes of human smoking but less of COPD.
- Published
- 2018
42. Multiethnic meta-analysis identifies ancestry-specific and cross-ancestry loci for pulmonary function
- Author
-
Brian D. Hobbs, Myriam Fornage, Hae Kyung Im, Lenore J. Launer, Dennis O. Mook-Kanamori, Robert C. Kaplan, Hieab H.H. Adams, Maarten van den Berge, George T. O'Connor, Natalie Terzikhan, Rodney J. Scott, Ani Manichaikul, Ravi Kalhan, Guy Brusselle, Leslie A. Lange, Yohan Bossé, Qing Duan, Tamara B. Harris, Esteban G. Burchard, Albert M. Levin, Kent D. Taylor, Mi Kyeong Lee, Woo Jin Kim, Yongmei Liu, Vilmundur Gudnason, Nora Franceschini, Hongsheng Gui, James G. Wilson, Donglei Hu, Lavinia Paternoster, Thomas Hansen, Roby Joehanes, Tianyuan Wang, Jennifer Liberto, Victoria E. Jackson, Jianping Jin, Colleen M. Sitlani, Tianxiao Huan, Wei Gao, Michael A. Province, Christopher Oldmeadow, Tarunveer S. Ahluwalia, Alanna C. Morrison, Kari E. North, Stephen B. Kritchevsky, Michael H. Cho, Gleb Kichaev, Kaare Christensen, Renée de Mutsert, Lauren S. Mogil, Mark McEvoy, Xin-Qun Wang, Juan C. Celedón, Fernando Pires Hartwig, Wenbo Tang, Judith M. Vonk, Kristin M. Burkart, Heather E. Wheeler, Bharat Thyagarajan, John Attia, Kim de Jong, Bruce M. Psaty, Sina A. Gharib, Traci M. Bartz, Josée Dupuis, Stephanie J. London, Ma'en Obeidat, Jerome I. Rotter, Stephen S. Rich, Mary K. Wojczynski, Susan R. Heckbert, Albert V. Smith, Frits R. Rosendaal, Tobias Bonten, Tamar Sofer, Patricia A. Cassano, H. Marike Boezen, Sam S. Oh, Ana M. B. Menezes, Mary F. Feitosa, David C. Nickle, Jason L. Sanders, Bernardo L. Horta, Fernando C. Wehrmeister, Jennifer N. Nguyen, Annah B. Wyss, Jeanne C. Latourelle, Margaret M. Parker, R. Graham Barr, Elizabeth G. Holliday, Richard H. Myers, Cathy C. Laurie, Kurt Lohman, Lies Lahousse, Don D. Sin, André G. Uitterlinden, Shujie Xiao, Jennifer A. Brody, L. Keoki Williams, Joohon Sung, Raymond Noordam, Mariaelisa Graff, Erasmus MC other, Epidemiology, Radiology & Nuclear Medicine, Groningen Research Institute for Asthma and COPD (GRIAC), Life Course Epidemiology (LCE), Læknadeild (HÍ), Faculty of Medicine (UI), Heilbrigðisvísindasvið (HÍ), School of Health Sciences (UI), Háskóli Íslands, and University of Iceland
- Subjects
0301 basic medicine ,Lung Diseases ,Male ,Linkage disequilibrium ,Epidemiology ,Vital Capacity ,General Physics and Astronomy ,Genome-wide association study ,VARIANTS ,Genome-wide association studies ,DISEASE ,Linkage Disequilibrium ,AIR-FLOW OBSTRUCTION ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Forced Expiratory Volume ,Genetics research ,Medicine and Health Sciences ,lcsh:Science ,Lung ,Genetics ,education.field_of_study ,Multidisciplinary ,LARGE-SCALE ,Smoking ,SMOOTH-MUSCLE ,Öndunarfærasjúkdómar ,Genomics ,Hispanic or Latino ,3. Good health ,LUNG-FUNCTION ,Meta-analysis ,Regression Analysis ,Female ,Erfðarannsóknir ,Science ,Population ,Quantitative Trait Loci ,Black People ,Quantitative trait locus ,Biology ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Article ,White People ,03 medical and health sciences ,Humans ,SMOKING-BEHAVIOR ,Genetic Predisposition to Disease ,GENOME-WIDE ASSOCIATION ,1000 Genomes Project ,education ,Genetic association ,Respiratory tract diseases ,Asian ,Faraldsfræði ,Biology and Life Sciences ,General Chemistry ,030104 developmental biology ,030228 respiratory system ,Sample Size ,SEGREGATION ANALYSIS ,lcsh:Q ,FOLLOW-UP ,Genome-Wide Association Study - Abstract
Nearly 100 loci have been identified for pulmonary function, almost exclusively in studies of European ancestry populations. We extend previous research by meta-analyzing genome-wide association studies of 1000 Genomes imputed variants in relation to pulmonary function in a multiethnic population of 90,715 individuals of European (N = 60,552), African (N = 8429), Asian (N = 9959), and Hispanic/Latino (N = 11,775) ethnicities. We identify over 50 additional loci at genome-wide significance in ancestry-specific or multiethnic meta-analyses. Using recent fine-mapping methods incorporating functional annotation, gene expression, and differences in linkage disequilibrium between ethnicities, we further shed light on potential causal variants and genes at known and newly identified loci. Several of the novel genes encode proteins with predicted or established drug targets, including KCNK2 and CDK12. Our study highlights the utility of multiethnic and integrative genomics approaches to extend existing knowledge of the genetics of lung function and clinical relevance of implicated loci., Supported in part by the Intramural Research Program of the National Institutes of Health, NIEHS. Infrastructure for the CHARGE Consortium is supported in part by the National Heart, Lung, and Blood Institute Grant R01HL105756
- Published
- 2018
43. New genetic signals for lung function highlight pathways and pleiotropy, and chronic obstructive pulmonary disease associations across multiple ancestries
- Author
-
Nick Shrine, Anna L Guyatt, A Mesut Erzurumluoglu, Victoria E Jackson, Brian D Hobbs, Carl Melbourne, Chiara Batini, Katherine A Fawcett, Kijoung Song, Phuwanat Sakornsakolpat, Xingnan Li, Ruth Boxall, Nicola F Reeve, Ma’en Obeidat, Jing Hua Zhao, Matthias Wielscher, Understanding Society Scientific Group, Stefan Weiss, Katherine A Kentistou, James P Cook, Benjamin B Sun, Jian Zhou, Jennie Hui, Stefan Karrasch, Medea Imboden, Sarah E Harris, Jonathan Marten, Stefan Enroth, Shona M Kerr, Ida Surakka, Veronique Vitart, Terho Lehtimäki, Richard J Allen, Per S Bakke, Terri H Beaty, Eugene R Bleecker, Yohan Bossé, Corry-Anke Brandsma, Zhengming Chen, James D Crapo, John Danesh, Dawn L DeMeo, Frank Dudbridge, Ralf Ewert, Christian Gieger, Amund Gulsvik, Anna L Hansell, Ke Hao, Josh D Hoffman, John Hokanson, Georg Homuth, Peter K Joshi, Philippe Joubert, Claudia Langenberg, Xuan Li, Liming Li, Kuang Lin, Lars Lind, Nick Locantore, Jian’an Luan, Anubha Mahajan, Joseph C Maranville, Alison Murray, David C Nickle, Richard Packer, Margaret M Parker, Megan L Paynton, David Porteous, Dmitry Prokopenko, Dandi Qiao, Rajesh Rawal, Heiko Runz, Ian Sayers, Don D Sin, Blair H Smith, María Soler Artigas, David Sparrow, Ruth Tal-Singer, Paul RHJ Timmers, Maarten Van den Berge, John C Whittaker, Prescott Woodruff, Laura M Yerges Armstrong, Olga G Troyanskaya, Olli T Raitakari, Mika Kähönen, Ozren Polasek, Ulf Gyllensten, Igor Rudan, Ian J Deary, Nicole M Probst-Hensch, Holger Schulz, Alan L James, James F Wilson, Beate Stubbe, Eleftheria Zeggini, Marjo-Riitta Jarvelin, Nick Wareham, Edwin K Silverman, Caroline Hayward, Andrew P Morris, Adam S Butterworth, Robert A Scott, Robin G Walters, Deborah A Meyers, Michael H Cho, David P Strachan, Ian P Hall, Martin D Tobin, and Louise V Wain
- Subjects
0303 health sciences ,COPD ,business.industry ,Pulmonary disease ,Bioinformatics ,medicine.disease ,3. Good health ,respiratory tract diseases ,03 medical and health sciences ,0302 clinical medicine ,Pleiotropy (drugs) ,Medicine ,business ,030217 neurology & neurosurgery ,Lung function ,030304 developmental biology ,Genetic association - Abstract
Reduced lung function predicts mortality and is key to the diagnosis of COPD. In a genome-wide association study in 400,102 individuals of European ancestry, we define 279 lung function signals, one-half of which are new. In combination these variants strongly predict COPD in deeply-phenotyped patient populations. Furthermore, the combined effect of these variants showed generalisability across smokers and never-smokers, and across ancestral groups. We highlight biological pathways, known and potential drug targets for COPD and, in phenome-wide association studies, autoimmune-related and other pleiotropic effects of lung function associated variants. This new genetic evidence has potential to improve future preventive and therapeutic strategies for COPD.
- Published
- 2018
- Full Text
- View/download PDF
44. Understanding the role of the chromosome 15q25.1 in COPD through epigenetics and transcriptomics
- Author
-
Yohan Bossé, Cornelia M. van Duijn, Guy Brusselle, Kim de Jong, Diana A van der Plaat, Lies Lahousse, Ivana Nedeljkovic, Joyce J B van Meurs, André G. Uitterlinden, Alen Faiz, Judith M. Vonk, H. Marike Boezen, Dirkje S. Postma, David C. Nickle, Bruno H. Stricker, Ma'en Obeidat, Elena Carnero-Montoro, Najaf Amin, Cleo C. van Diemen, Maarten van den Berge, Groningen Research Institute for Asthma and COPD (GRIAC), Life Course Epidemiology (LCE), Epidemiology, Internal Medicine, Pulmonary Medicine, APH - Methodology, APH - Mental Health, Amsterdam Reproduction & Development, and Biological Psychology
- Subjects
0301 basic medicine ,Male ,Genome-wide association study ,Receptors, Nicotinic ,Pulmonary Disease, Chronic Obstructive ,Risk Factors ,Medicine ,SUSCEPTIBILITY LOCUS ,Genetics (clinical) ,DNA METHYLATION LEVELS ,Genetics & Heredity ,RISK ,COPD ,education.field_of_study ,LOCALIZATION ,Middle Aged ,DNA methylation ,Female ,Life Sciences & Biomedicine ,Biochemistry & Molecular Biology ,Proteasome Endopeptidase Complex ,GENES ,Population ,Quantitative Trait Loci ,Locus (genetics) ,OBSTRUCTIVE PULMONARY-DISEASE ,Article ,Cigarette Smoking ,03 medical and health sciences ,LUNG-CANCER ,SDG 3 - Good Health and Well-being ,Genetics ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,GENOME-WIDE ASSOCIATION ,education ,Iron Regulatory Protein 2 ,Genetic Association Studies ,Genetic association ,Aged ,0604 Genetics ,Chromosomes, Human, Pair 15 ,Science & Technology ,business.industry ,DNA Methylation ,medicine.disease ,NICOTINE DEPENDENCE ,respiratory tract diseases ,030104 developmental biology ,Gene Expression Regulation ,Immunology ,Expression quantitative trait loci ,CIGARETTE-SMOKING ,business - Abstract
Chronic obstructive pulmonary disease (COPD) is a major health burden in adults and cigarette smoking is considered the most important environmental risk factor of COPD. Chromosome 15q25.1 locus is associated with both COPD and smoking. Our study aims at understanding the mechanism underlying the association of chromosome 15q25.1 with COPD through epigenetic and transcriptional variation in a population-based setting. To assess if COPD-associated variants in 15q25.1 are methylation quantitative trait loci, epigenome-wide association analysis of four genetic variants, previously associated with COPD (P < 5 × 10-8) in the 15q25.1 locus (rs12914385:C>T-CHRNA3, rs8034191:T>C-HYKK, rs13180:C>T-IREB2 and rs8042238:C>T-IREB2), was performed in the Rotterdam study (n = 1489). All four variants were significantly associated (P < 1.4 × 10-6) with blood DNA methylation of IREB2, CHRNA3 and PSMA4, of which two, including IREB2 and PSMA4, were also differentially methylated in COPD cases and controls (P < 0.04). Further additive and multiplicative effects of smoking were evaluated and no significant effect was observed. To evaluate if these four genetic variants are expression quantitative trait loci, transcriptome-wide association analysis was performed in 1087 lung samples. All four variants were also significantly associated with differential expression of the IREB2 3'UTR in lung tissues (P < 5.4 × 10-95). We conclude that regulatory mechanisms affecting the expression of IREB2 gene, such as DNA methylation, may explain the association between genetic variants in chromosome 15q25.1 and COPD, largely independent of smoking.
- Published
- 2018
45. Leveraging lung tissue transcriptome to uncover candidate causal genes in COPD genetic associations
- Author
-
Jean-Christophe Bérubé, Maxime Lamontagne, Ke Hao, David C. Nickle, Maarten van den Berge, Peter D. Paré, Don D. Sin, Michel Laviolette, Kim de Jong, Yohan Bossé, Wim Timens, Ma'en Obeidat, Philippe Joubert, Phuwanat Sakornsakolpat, Brian D. Hobbs, Michael H. Cho, H. Marike Boezen, Groningen Research Institute for Asthma and COPD (GRIAC), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Life Course Epidemiology (LCE)
- Subjects
0301 basic medicine ,Candidate gene ,Quantitative Trait Loci ,Genomics ,Genome-wide association study ,Biology ,Genome ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Mendelian randomization ,Genetics ,Journal Article ,Humans ,Genetic Predisposition to Disease ,Molecular Biology ,Genotyping ,Lung ,Genetics (clinical) ,Genetic Association Studies ,Genetic association ,Association Studies Articles ,General Medicine ,3. Good health ,respiratory tract diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,Expression quantitative trait loci ,Transcriptome ,Genome-Wide Association Study - Abstract
Causal genes of chronic obstructive pulmonary disease (COPD) remain elusive. The current study aims at integrating genome-wide association studies (GWAS) and lung expression quantitative trait loci (eQTL) data to map COPD candidate causal genes and gain biological insights into the recently discovered COPD susceptibility loci. Two complementary genomic datasets on COPD were studied. First, the lung eQTL dataset which included whole-genome gene expression and genotyping data from 1038 individuals. Second, the largest COPD GWAS to date from the International COPD Genetics Consortium (ICGC) with 13 710 cases and 38 062 controls. Methods that integrated GWAS with eQTL signals including transcriptome-wide association study (TWAS), colocalization and Mendelian randomization-based (SMR) approaches were used to map causality genes, i.e. genes with the strongest evidence of being the functional effector at specific loci. These methods were applied at the genome-wide level and at COPD risk loci derived from the GWAS literature. Replication was performed using lung data from GTEx. We collated 129 non-overlapping risk loci for COPD from the GWAS literature. At the genome-wide scale, 12 new COPD candidate genes/loci were revealed and six replicated in GTEx including CAMK2A, DMPK, MYO15A, TNFRSF10A, BTN3A2 and TRBV30. In addition, we mapped candidate causal genes for 60 out of the 129 GWAS-nominated loci and 23 of them were replicated in GTEx. Mapping candidate causal genes in lung tissue represents an important contribution to the genetics of COPD, enriches our biological interpretation of GWAS findings, and brings us closer to clinical translation of genetic associations.
- Published
- 2018
46. Identification of susceptibility pathways for the role of chromosome 15q25.1 in modifying lung cancer risk
- Author
-
Natasha B. Leighl, Mattias Johansson, John McLaughlin, Gad Rennert, Juncheng Dai, Ana Fernández-Somoano, Jian-Min Yuan, David C. Qian, Kjell Grankvist, Angela Risch, Rayjean J. Hung, Lei Song, Xuchen Zong, David C. Muller, Philip Lazarus, Frances A. Shepherd, Lesley M. Butler, Heike Bickeböller, Robert Carreras-Torres, Loic Le Marchand, Hongbing Shen, Ming-Sound Tsao, Christopher I. Amos, Zhibin Hu, Tadeusz M Orlowski, Jin Hee Kim, Angeline S. Andrew, Mariella De Biasi, Paul Brennan, Fangyi Gu, Kim Overvad, Yu-Tang Gao, Anush Mukeria, Yohan Bossé, Shanbeh Zienolddiny, Jennifer A. Doherty, Thomas Muley, Neil E. Caporaso, David Zaridze, Maxime Lamontagne, Jiang Gui, Irene Brüske, Antonia Trichopoulou, Adonina Tardón, Guillermo Fernández-Tardón, Nancy Diao, Albert Rosenberger, Woon-Puay Koh, Eric B. Haura, David C. Christiani, Beata Swiatkowska, Christopher A. Haiman, Milica Kontic, Pier Alberto Bertazzi, Aage Haugen, Rosario Tumino, Rachel F. Tyndale, Eric J. Duell, Jolanta Lissowska, David C. Nickle, María Soler Artigas, Melinda C. Aldrich, Anders Mellemgaard, Yun-Chul Hong, Demetrios Albanes, Per Bakke, Penella J. Woll, Matthew B. Schabath, Judith Manz, Mikael Johansson, Yafang Li, Angela Cecilia Pesatori, Stig E. Bojesen, Stephen J. Chanock, Dakai Zhu, Louise V. Wain, Chu Chen, Simona Ognjanovic, Olle Melander, Gary E. Goodman, Stephen Lam, Xifeng Wu, Martin D. Tobin, Erik H.F.M. van der Heijden, Jakob S Johansen, Li Su, Ivana Holcatova, Xiangjun Xiao, Ivan P. Gorlov, Susanne M. Arnold, Hans Brunnström, Jonas Manjer, Walid Saliba, M. Dawn Teare, John K. Field, Bin Zhu, James D. McKay, Richard S. Houlston, Ghislaine Scelo, Younghun Han, Ahsan Kamal, Lynne R. Wilkens, Yonathan Brhane, Maria Teresa Landi, William S. Bush, Wei-Qi Wei, Fiona Taylor, Lambertus A. Kiemeney, Victoria L. Stevens, Yuanqing Ye, Jinyoung Byun, Michael P.A. Davies, Ruyang Zhang, Xuemei Ji, Ma'en Obeidat, Michael W. Marcus, Philippe Joubert, V. Janout, Angela Cox, Olga Y. Gorlova, Ciprian Bolca, Wim Timens, Vidar Skaug, Geoffrey Liu, Groningen Research Institute for Asthma and COPD (GRIAC), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Nofer Institute of Occupational Medicine
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,LOCI ,General Physics and Astronomy ,Genome-wide association study ,CA2+ TRANSPORT ,Cohort Studies ,Risk Factors ,GENETIC-VARIANTS ,Smoking/adverse effects ,Gene Regulatory Networks ,Child ,Genetics ,Gated channel activity ,AFRICAN-AMERICANS ,Chromosomes, Human, Pair 15/genetics ,Multidisciplinary ,Factors de risc en les malalties ,Smoking ,Genome-Wide ,Middle Aged ,3. Good health ,Multidisciplinary Sciences ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,Child, Preschool ,Science & Technology - Other Topics ,Female ,Lung cancer ,Medical Genetics ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,EXPRESSION ,Adult ,Lung Neoplasms/genetics ,Adolescent ,Risk factors in diseases ,Science ,Quantitative Trait Loci ,Single-nucleotide polymorphism ,Locus (genetics) ,Quantitative trait locus ,Biology ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Young Adult ,All institutes and research themes of the Radboud University Medical Center ,MD Multidisciplinary ,medicine ,Humans ,Genetic Predisposition to Disease ,KEGG ,GENOME-WIDE ASSOCIATION ,Aged ,Medicinsk genetik ,Chromosomes, Human, Pair 15 ,Science & Technology ,SET ENRICHMENT ANALYSIS ,Infant, Newborn ,Infant ,Reproducibility of Results ,ADENOCARCINOMA ,General Chemistry ,medicine.disease ,NICOTINE DEPENDENCE ,030104 developmental biology ,Gene Ontology ,Expression quantitative trait loci ,Càncer de pulmó ,Quantitative Trait Loci/genetics - Abstract
Supported by FIS-FEDER/Spain grant numbers FIS-01/310, FIS-PI03-0365, and FIS-07-BI060604, FICYT/Asturias grant numbers FICYT PB02-67 and FICYT IB09-133, and the University Institute of Oncology (IUOPA) of the University of Oviedo and the Ciber de Epidemiologia y Salud Pública., Ji, X., Bossé, Y., Landi, M.T., Gui, J., Xiao, X., Qian, D., Joubert, P., Lamontagne, M., Li, Y., Gorlov, I., de Biasi, M., Han, Y., Gorlova, O., Hung, R.J., Wu, X., McKay, J., Zong, X., Carreras-Torres, R., Christiani, D.C., Caporaso, N., Johansson, M., Liu, G., Bojesen, S.E., Le Marchand, L., Albanes, D., Bickeböller, H., Aldrich, M.C., Bush, W.S., Tardon, A., Rennert, G., Chen, C., Teare, M.D., Field, J.K., Kiemeney, L.A., Lazarus, P., Haugen, A., Lam, S., Schabath, M.B.,,rew, A.S., Shen, H., Hong, Y.-C., Yuan, J.-M., Bertazzi, P.A., Pesatori, A.C., Ye, Y., Diao, N., Su, L., Zhang, R., Brhane, Y., Leighl, N., Johansen, J.S., Mellemgaard, A., Saliba, W., Haiman, C., Wilkens, L., Fernandez-Somoano, A., Fernandez-Tardon, G., van der Heijden, E.H.F.M., Kim, J.H., Dai, J., Hu, Z., Davies, M.P.A., Marcus, M.W., Brunnström, H., Manjer, J., Melander, O., Muller, D.C., Overvad, K., Trichopoulou, A., Tumino, R., Doherty, J., Goodman, G.E., Cox, A., Taylor, F., Woll, P., Brüske, I., Manz, J., Muley, T., Risch, A., Rosenberger, A., Grankvist, K., Johansson, M., Shepherd, F., Tsao, M.-S., Arnold, S.M., Haura, E.B., Bolca, C., Holcatova, I., Janout, V., Kontic, M., Lissowska, J., Mukeria, A., Ognjanovic, S., Orlowski, T.M., Scelo, G., Swiatkowska, B., Zaridze, D., Bakke, P., Skaug, V., Zienolddiny, S., Duell, E.J., Butler, L.M., Koh, W.-P., Gao, Y.-T., Houlston, R., McLaughlin, J., Stevens, V., Nickle, D.C., Obeidat, M., Timens, W., Zhu, B., Song, L., Artigas, M.S., Tobin, M.D., Wain, L.V., Gu, F., Byun, J., Kamal, A., Zhu, D., Tyndale, R.F., Wei, W.-Q., Chanock, S., Brennan, P., Amos, C.I.
- Published
- 2018
47. A large lung gene expression study identifying fibulin-5 as a novel player in tissue repair in COPD
- Author
-
Don D. Sin, Maarten van den Berge, Rudolf S N Fehrmann, Sharon Brouwer, Juha Karjalainen, Peter D. Paré, Michel Laviolette, Corry-Anke Brandsma, Wim Timens, Marnix R. Jonker, Dirkje S. Postma, Yohan Bossé, Anita I.R. Spanjer, Ke Hao, Lude Franke, David C. Nickle, Molecular Pharmacology, Damage and Repair in Cancer Development and Cancer Treatment (DARE), Guided Treatment in Optimal Selected Cancer Patients (GUTS), Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Groningen Research Institute for Asthma and COPD (GRIAC), and Stem Cell Aging Leukemia and Lymphoma (SALL)
- Subjects
Male ,Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Blotting, Western ,EMPHYSEMA ,Polymerase Chain Reaction ,OBSTRUCTIVE PULMONARY-DISEASE ,Pulmonary Disease, Chronic Obstructive ,Fibrosis ,Gene expression ,medicine ,Humans ,PERIOSTIN ,FIBROSIS ,RNA, Messenger ,ELASTIN ,Lung ,IN-VIVO ,Aged ,Extracellular Matrix Proteins ,COPD ,biology ,business.industry ,Gene Expression Profiling ,TGF-BETA ,Middle Aged ,Gene signature ,medicine.disease ,Immunohistochemistry ,Elasticity ,respiratory tract diseases ,Gene expression profiling ,medicine.anatomical_structure ,Gene Expression Regulation ,FBLN5 ,biology.protein ,Female ,business ,Elastin ,Genome-Wide Association Study ,BINDS - Abstract
Background Chronic obstructive pulmonary disease (COPD) is a progressive, incurable lung disease characterised by abnormal tissue repair causing emphysema and small airways fibrosis. Since current therapy cannot modify this abnormal repair, it is crucial to unravel its underlying molecular mechanisms. Unbiased analysis of genome-wide gene expression profiles in lung tissue provides a powerful tool to investigate this. Methods We performed genome-wide gene expression profiling in 581 lung tissue samples from current and ex-smokers with (n=311) and without COPD (n=270). Subsequently, quantitative PCR, western blot and immunohistochemical analyses were performed to validate our main findings. Results 112 genes were found to be upregulated in patients with COPD compared with controls, whereas 61 genes were downregulated. Among the most upregulated genes were fibulin-5 ( FBLN5 ), elastin ( ELN ), latent transforming growth factor β binding protein 2 ( LTBP2 ) and microfibrillar associated protein 4 ( MFAP4 ), all implicated in elastogenesis. Our gene expression findings were validated at mRNA and protein level. We demonstrated higher ELN gene expression in COPD lung tissue and similar trends for FBLN5 and MFAP4 , and negative correlations with lung function. FBLN5 protein levels were increased in COPD lung tissue and cleaved, possibly non-functional FBLN5 protein was present. Strong coexpression of FBLN5 , ELN , LTBP2 and MFAP4 in lung tissue and in silico analysis indicated cofunctionality of these genes. Finally, colocalisation of FBLN5, MFAP4 and LTBP2 with elastic fibres was demonstrated in lung tissue. Conclusions We identified a clear gene signature for elastogenesis in COPD and propose FBLN5 as a novel player in tissue repair in COPD.
- Published
- 2015
48. Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: A genome-wide association study
- Author
-
Imre Noth, Doris M Rassl, Nick Shrine, Ann B. Millar, Richard P. Marshall, Philip L. Molyneaux, Ma'en Obeidat, Alison E. John, Justin M. Oldham, Shwu-Fan Ma, Michael Hill, Don D. Sin, Rebecca Braybrooke, Gauri Saini, Vidya Navaratnam, David A. Schwartz, Ian Sayers, Simon P. Hart, Martin D. Tobin, Moira K. B. Whyte, Nik Hirani, Carlos Flores, Amanda P. Henry, Richard Hubbard, Ivana V. Yang, Norma Thompson, Eunice Oballa, Yohan Bossé, Ian P. Hall, Wim Timens, Louise V. Wain, Toby M. Maher, Joanne Porte, David C. Nickle, William A. Fahy, R. Gisli Jenkins, Tsukasa Okamoto, Richard J. Allen, Helen Booth, Beatriz Guillen-Guio, Robin J. McAnulty, Tasha E. Fingerlin, Helen Parfrey, Groningen Research Institute for Asthma and COPD (GRIAC), Guided Treatment in Optimal Selected Cancer Patients (GUTS), and National Institute for Health Research
- Subjects
Male ,0301 basic medicine ,A Kinase Anchor Proteins ,Genome-wide association study ,Disease ,DISEASE ,Idiopathic pulmonary fibrosis ,0302 clinical medicine ,LUNG FIBROBLASTS ,IMPUTATION ,Medicine ,Middle Aged ,respiratory system ,3. Good health ,Europe ,medicine.anatomical_structure ,TGF-BETA ACTIVATION ,SURVIVAL ,Female ,Signal Transduction ,Pulmonary and Respiratory Medicine ,EXPRESSION ,White People ,Article ,Minor Histocompatibility Antigens ,CYCLOOXYGENASE-2 ,ALVEOLITIS ,03 medical and health sciences ,RHO ,Proto-Oncogene Proteins ,Journal Article ,TGF beta Activation ,Humans ,Genetic Predisposition to Disease ,RNA, Messenger ,Allele ,Aged ,Lung ,business.industry ,Case-control study ,Genetic Variation ,Odds ratio ,medicine.disease ,NUCLEOTIDE EXCHANGE FACTOR ,Idiopathic Pulmonary Fibrosis ,respiratory tract diseases ,Tertiary Lymphoid Structures ,030104 developmental biology ,030228 respiratory system ,Alveolar Epithelial Cells ,Case-Control Studies ,Immunology ,rhoA GTP-Binding Protein ,business ,Rho Guanine Nucleotide Exchange Factors ,Software ,Genome-Wide Association Study - Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high mortality, uncertain cause, and few treatment options. Studies have identified a significant genetic risk associated with the development of IPF; however, mechanisms by which genetic risk factors promote IPF remain unclear. We aimed to identify genetic variants associated with IPF susceptibility and provide mechanistic insight using gene and protein expression analyses.Methods: We used a two-stage approach: a genome-wide association study in patients with IPF of European ancestry recruited from nine different centres in the UK and controls selected from UK Biobank (stage 1) matched for age, sex, and smoking status; and a follow-up of associated genetic variants in independent datasets of patients with IPF and controls from two independent US samples from the Chicago consortium and the Colorado consortium (stage 2). We investigated the effect of novel signals on gene expression in large transcriptomic and genomic data resources, and examined expression using lung tissue samples from patients with IPF and controls.Findings: 602 patients with IPF and 3366 controls were selected for stage 1. For stage 2, 2158 patients with IPF and 5195 controls were selected. We identified a novel genome-wide significant signal of association with IPF susceptibility near A-kinase anchoring protein 13 (AKAP13; rs62025270, odds ratio [OR] 1·27 [95% CI 1·18–1·37], p=1·32 × 10−9) and confirmed previously reported signals, including in mucin 5B (MUC5B; rs35705950, OR 2·89 [2·56–3·26], p=1·12 × 10−66) and desmoplakin (DSP; rs2076295, OR 1·44 [1·35–1·54], p=7·81 × 10−28). For rs62025270, the allele A associated with increased susceptibility to IPF was also associated with increased expression of AKAP13 mRNA in lung tissue from patients who had lung resection procedures (n=1111). We showed that AKAP13 is expressed in the alveolar epithelium and lymphoid follicles from patients with IPF, and AKAP13 mRNA expression was 1·42-times higher in lung tissue from patients with IPF (n=46) than that in lung tissue from controls (n=51).Interpretation: AKAP13 is a Rho guanine nucleotide exchange factor regulating activation of RhoA, which is known to be involved in profibrotic signalling pathways. The identification of AKAP13 as a susceptibility gene for IPF increases the prospect of successfully targeting RhoA pathway inhibitors in patients with IPF.Funding: UK Medical Research Council, National Heart, Lung, and Blood Institute of the US National Institutes of Health, Agencia Canaria de Investigación, Innovación y Sociedad de la Información, Spain, UK National Institute for Health Research, and the British Lung Foundation.
- Published
- 2017
49. COPD GWAS variant at 19q13.2 in relation with DNA methylation and gene expression
- Author
-
Maarten van den Berge, Ma'en Obeidat, Ivana Nedeljkovic, Yohan Bossé, Kim de Jong, Bruno H. Stricker, Elena Carnero-Montoro, Najaf Amin, Cornelia M. van Duijn, André G. Uitterlinden, H. Marike Boezen, Guy Brusselle, Lies Lahousse, Joyce B. J. van Meurs, Judith M. Vonk, Alen Faiz, Dirkje S. Postma, Cleo C. van Diemen, Diana A van der Plaat, David C. Nickle, Epidemiology, Erasmus MC other, Internal Medicine, Pulmonary Medicine, APH - Methodology, APH - Mental Health, Amsterdam Reproduction & Development, Biological Psychology, Groningen Research Institute for Asthma and COPD (GRIAC), and Life Course Epidemiology (LCE)
- Subjects
0301 basic medicine ,Male ,LOCI ,Gene Expression ,Genome-wide association study ,Epigenesis, Genetic ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Medicine and Health Sciences ,Association Studies Article ,Genetics (clinical) ,Genetics ,Genetics & Heredity ,RISK ,COPD ,Smoking ,Chromosome Mapping ,General Medicine ,Methylation ,11 Medical And Health Sciences ,Middle Aged ,3. Good health ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,Life Sciences & Biomedicine ,Adult ,Biochemistry & Molecular Biology ,Quantitative Trait Loci ,Biology ,Methylation Site ,OBSTRUCTIVE PULMONARY-DISEASE ,Polymorphism, Single Nucleotide ,Hypoxia-Inducible Factor-Proline Dioxygenases ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,medicine ,Journal Article ,Humans ,SMOKING-BEHAVIOR ,Genetic Predisposition to Disease ,Epigenetics ,GENOME-WIDE ASSOCIATION ,Molecular Biology ,Genetic association ,Aged ,Science & Technology ,rab4 GTP-Binding Proteins ,Genetic Variation ,DNA Methylation ,06 Biological Sciences ,medicine.disease ,030104 developmental biology ,Expression quantitative trait loci ,Chromosomes, Human, Pair 19 ,Genome-Wide Association Study - Abstract
© The Author 2017. Chronic obstructive pulmonary disease (COPD) is among the major health burdens in adults. While cigarette smoking is the leading risk factor, a growing number of genetic variations have been discovered to influence disease susceptibility. Epigenetic modifications may mediate the response of the genome to smoking and regulate gene expression. Chromosome 19q13.2 region is associated with both smoking and COPD, yet its functional role is unclear. Our study aimed to determine whether rs7937 (RAB4B, EGLN2), a top genetic variant in 19q13.2 region identified in genome-wide association studies of COPD, is associated with differential DNA methylation in blood (N=1490) and gene expression in blood (N=721) and lungs (N=1087). We combined genetic and epigenetic data from the Rotterdam Study (RS) to perform the epigenome-wide association analysis of rs7937. Further, we used genetic and transcriptomic data from blood (RS) and from lung tissue (Lung expression quantitative trait loci mapping study), to perform the transcriptome-wide association study of rs7937. Rs7937 was significantly (FDR < 0.05) and consistently associated with differential DNA methylation in blood at 4 CpG sites in cis, independent of smoking. One methylation site (cg11298343-EGLN2) was also associated with COPD (P=0.001). Additionally, rs7937 was associated with gene expression levels in blood in cis (EGLN2), 42% mediated through cg11298343, and in lung tissue, in cis and trans (NUMBL, EGLN2, DNMT3A, LOC101929709 and PAK2). Our results suggest that changes of DNA methylation and gene expression may be intermediate steps between genetic variants and COPD, but further causal studies in lung tissue should confirm this hypothesis.
- Published
- 2017
50. Multiethnic meta-analysis identifies new loci for pulmonary function
- Author
-
Yongmei Liu, Dennis O. Mook-Kanamori, Robert C. Kaplan, Brian D. Hobbs, Josée Dupuis, Nora Franceschini, Tarunveer S. Ahluwalia, Hieab H.H. Adams, André G. Uitterlinden, Stephen B. Kritchevsky, Gleb Kichaev, Mary F. Feitosa, Ani Manichaikul, Tamar Sofer, Myriam Fornage, Annah B. Wyss, Jeanne C. Latourelle, Patricia A. Cassano, H. Marike Boezen, Mark McEvoy, Roby Joehanes, Xin-Qun Wang, Victoria E. Jackson, Hae Kyung Im, Lenore J. Launer, Don D. Sin, R. Graham Barr, Tianyuan Wang, George T. O'Connor, Natalie Terzikhan, Tamara B. Harris, Wei Gao, Jennifer A. Brody, Rd Mutsert, Lavinia Paternoster, Thomas Hansen, Tianxiao Huan, Fernando C. Wehrmeister, Christopher Oldmeadow, Rodney J. Scott, Joohon Sung, Jason L. Sanders, Kurt Lohman, Woo Jin Kim, Guy Brusselle, Raymond Noordam, Michael A. Province, Yohan Bossé, Kari E. North, Mary K. Wojczynski, Fernando Pires Hartwig, Wenbo Tang, Bharat Thyagarajan, Judith M. Vonk, Jennifer N. Nguyen, Sina A. Gharib, Jianping Jin, Mariaelisa Graff, Kent D. Taylor, Mi Kyeong Lee, James G. Wilson, Elizabeth G. Holliday, Lies Lahousse, Richard H. Myers, Tobias Bonten, Stephen S. Rich, Cathy C. Laurie, Michael H. Cho, Susan R. Heckbert, Ravi Kalhan, Mvd Berge, Bernardo L. Horta, John Attia, Bruce M. Psaty, Amb Menezes, Albert V. Smith, Juan C. Celedón, Kd Jong, Jerome I. Rotter, David C. Nickle, Traci M. Bartz, Frits R. Rosendaal, Stephanie J. London, Alanna C. Morrison, Kristin M. Burkart, Colleen M. Sitlani, Ma'en Obeidat, Leslie A. Lange, Qing Duan, and Kaare Christensen
- Subjects
Genetics ,0303 health sciences ,Linkage disequilibrium ,Biology ,Pulmonary function testing ,Novel gene ,03 medical and health sciences ,0302 clinical medicine ,Functional annotation ,030220 oncology & carcinogenesis ,Meta-analysis ,1000 Genomes Project ,Gene ,030304 developmental biology ,Genetic association - Abstract
Nearly 100 loci have been identified for pulmonary function, almost exclusively in studies of European ancestry populations. We extend previous research by meta-analyzing genome-wide association studies of 1000 Genomes imputed variants in relation to pulmonary function in a multiethnic population of 90,715 individuals of European (N=60,552), African (N=8,429), Asian (N=9,959), and Hispanic/Latino (N=11,775) ethnicities. We identified over 50 novel loci at genome-wide significance in ancestry-specific and/or multiethnic meta-analyses. Recent fine mapping methods incorporating functional annotation, gene expression, and/or differences in linkage disequilibrium between ethnicities identified potential causal variants and genes at known and newly identified loci. Sixteen of the novel genes encode proteins with predicted or established drug targets, including KCNK2 and CDK12.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.