Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ma'en Obeidat is active.

Publication


Featured researches published by Ma'en Obeidat.


The Lancet Respiratory Medicine | 2015

Novel insights into the genetics of smoking behaviour, lung function, and chronic obstructive pulmonary disease (UK BiLEVE): a genetic association study in UK Biobank.

Louise V. Wain; Nick Shrine; Suzanne Miller; Victoria E. Jackson; Ioanna Ntalla; María Soler Artigas; Charlotte K. Billington; Abdul Kader Kheirallah; Richard J. Allen; James P. Cook; Kelly Probert; Ma'en Obeidat; Yohan Bossé; Ke Hao; Dirkje S. Postma; Peter D. Paré; Adaikalavan Ramasamy; Reedik Mägi; Evelin Mihailov; Eva Reinmaa; Erik Melén; Jared O'Connell; Eleni Frangou; Olivier Delaneau; Colin Freeman; Desislava Petkova; Mark I. McCarthy; Ian Sayers; Panos Deloukas; Richard Hubbard

Summary Background Understanding the genetic basis of airflow obstruction and smoking behaviour is key to determining the pathophysiology of chronic obstructive pulmonary disease (COPD). We used UK Biobank data to study the genetic causes of smoking behaviour and lung health. Methods We sampled individuals of European ancestry from UK Biobank, from the middle and extremes of the forced expiratory volume in 1 s (FEV1) distribution among heavy smokers (mean 35 pack-years) and never smokers. We developed a custom array for UK Biobank to provide optimum genome-wide coverage of common and low-frequency variants, dense coverage of genomic regions already implicated in lung health and disease, and to assay rare coding variants relevant to the UK population. We investigated whether there were shared genetic causes between different phenotypes defined by extremes of FEV1. We also looked for novel variants associated with extremes of FEV1 and smoking behaviour and assessed regions of the genome that had already shown evidence for a role in lung health and disease. We set genome-wide significance at p<5 × 10−8. Findings UK Biobank participants were recruited from March 15, 2006, to July 7, 2010. Sample selection for the UK BiLEVE study started on Nov 22, 2012, and was completed on Dec 20, 2012. We selected 50 008 unique samples: 10 002 individuals with low FEV1, 10 000 with average FEV1, and 5002 with high FEV1 from each of the heavy smoker and never smoker groups. We noted a substantial sharing of genetic causes of low FEV1 between heavy smokers and never smokers (p=2·29 × 10−16) and between individuals with and without doctor-diagnosed asthma (p=6·06 × 10−11). We discovered six novel genome-wide significant signals of association with extremes of FEV1, including signals at four novel loci (KANSL1, TSEN54, TET2, and RBM19/TBX5) and independent signals at two previously reported loci (NPNT and HLA-DQB1/HLA-DQA2). These variants also showed association with COPD, including in individuals with no history of smoking. The number of copies of a 150 kb region containing the 5′ end of KANSL1, a gene that is important for epigenetic gene regulation, was associated with extremes of FEV1. We also discovered five new genome-wide significant signals for smoking behaviour, including a variant in NCAM1 (chromosome 11) and a variant on chromosome 2 (between TEX41 and PABPC1P2) that has a trans effect on expression of NCAM1 in brain tissue. Interpretation By sampling from the extremes of the lung function distribution in UK Biobank, we identified novel genetic causes of lung function and smoking behaviour. These results provide new insight into the specific mechanisms underlying airflow obstruction, COPD, and tobacco addiction, and show substantial shared genetic architecture underlying airflow obstruction across individuals, irrespective of smoking behaviour and other airway disease. Funding Medical Research Council.


American Journal of Respiratory and Critical Care Medicine | 2011

Effect of five genetic variants associated with lung function on the risk of chronic obstructive lung disease, and their joint effects on lung function

M. Soler Artigas; Louise V. Wain; Emmanouela Repapi; Ma'en Obeidat; Ian Sayers; Paul R. Burton; Toby Johnson; Jiao Zhao; Eva Albrecht; Anna F. Dominiczak; Sm Kerr; Blair H. Smith; Gemma Cadby; Jennie Hui; Lyle J. Palmer; Aroon D. Hingorani; Sg Wannamethee; P H Whincup; S Ebrahim; George Davey Smith; Inês Barroso; Remco Loos; Nicholas J. Wareham; C Cooper; E Dennison; Seif O. Shaheen; Jimmy Z. Liu; Jonathan Marchini; Santosh Dahgam; Åsa Torinsson Naluai

RATIONALE Genomic loci are associated with FEV1 or the ratio of FEV1 to FVC in population samples, but their association with chronic obstructive pulmonary disease (COPD) has not yet been proven, nor have their combined effects on lung function and COPD been studied. OBJECTIVES To test association with COPD of variants at five loci (TNS1, GSTCD, HTR4, AGER, and THSD4) and to evaluate joint effects on lung function and COPD of these single-nucleotide polymorphisms (SNPs), and variants at the previously reported locus near HHIP. METHODS By sampling from 12 population-based studies (n = 31,422), we obtained genotype data on 3,284 COPD case subjects and 17,538 control subjects for sentinel SNPs in TNS1, GSTCD, HTR4, AGER, and THSD4. In 24,648 individuals (including 2,890 COPD case subjects and 13,862 control subjects), we additionally obtained genotypes for rs12504628 near HHIP. Each allele associated with lung function decline at these six SNPs contributed to a risk score. We studied the association of the risk score to lung function and COPD. MEASUREMENTS AND MAIN RESULTS Association with COPD was significant for three loci (TNS1, GSTCD, and HTR4) and the previously reported HHIP locus, and suggestive and directionally consistent for AGER and TSHD4. Compared with the baseline group (7 risk alleles), carrying 10-12 risk alleles was associated with a reduction in FEV1 (β = -72.21 ml, P = 3.90 × 10(-4)) and FEV1/FVC (β = -1.53%, P = 6.35 × 10(-6)), and with COPD (odds ratio = 1.63, P = 1.46 × 10(-5)). CONCLUSIONS Variants in TNS1, GSTCD, and HTR4 are associated with COPD. Our highest risk score category was associated with a 1.6-fold higher COPD risk than the population average score.


The Lancet Respiratory Medicine | 2015

Molecular mechanisms underlying variations in lung function: a systems genetics analysis

Ma'en Obeidat; Ke Hao; Yohan Bossé; David C. Nickle; Yunlong Nie; Dirkje S. Postma; Michel Laviolette; Andrew J. Sandford; Denise Daley; James C. Hogg; W. Mark Elliott; Nick Fishbane; Wim Timens; Pirro G. Hysi; Jaakko Kaprio; James F. Wilson; Jennie Hui; Rajesh Rawal; Holger Schulz; Beate Stubbe; Caroline Hayward; Ozren Polasek; Marjo-Riitta Järvelin; Jing Hua Zhao; Deborah Jarvis; Mika Kähönen; Nora Franceschini; Kari E. North; Daan W. Loth; Guy Brusselle

BACKGROUND Lung function measures reflect the physiological state of the lung, and are essential to the diagnosis of chronic obstructive pulmonary disease (COPD). The SpiroMeta-CHARGE consortium undertook the largest genome-wide association study (GWAS) so far (n=48,201) for forced expiratory volume in 1 s (FEV1) and the ratio of FEV1 to forced vital capacity (FEV1/FVC) in the general population. The lung expression quantitative trait loci (eQTLs) study mapped the genetic architecture of gene expression in lung tissue from 1111 individuals. We used a systems genetics approach to identify single nucleotide polymorphisms (SNPs) associated with lung function that act as eQTLs and change the level of expression of their target genes in lung tissue; termed eSNPs. METHODS The SpiroMeta-CHARGE GWAS results were integrated with lung eQTLs to map eSNPs and the genes and pathways underlying the associations in lung tissue. For comparison, a similar analysis was done in peripheral blood. The lung mRNA expression levels of the eSNP-regulated genes were tested for associations with lung function measures in 727 individuals. Additional analyses identified the pleiotropic effects of eSNPs from the published GWAS catalogue, and mapped enrichment in regulatory regions from the ENCODE project. Finally, the Connectivity Map database was used to identify potential therapeutics in silico that could reverse the COPD lung tissue gene signature. FINDINGS SNPs associated with lung function measures were more likely to be eQTLs and vice versa. The integration mapped the specific genes underlying the GWAS signals in lung tissue. The eSNP-regulated genes were enriched for developmental and inflammatory pathways; by comparison, SNPs associated with lung function that were eQTLs in blood, but not in lung, were only involved in inflammatory pathways. Lung function eSNPs were enriched for regulatory elements and were over-represented among genes showing differential expression during fetal lung development. An mRNA gene expression signature for COPD was identified in lung tissue and compared with the Connectivity Map. This in-silico drug repurposing approach suggested several compounds that reverse the COPD gene expression signature, including a nicotine receptor antagonist. These findings represent novel therapeutic pathways for COPD. INTERPRETATION The system genetics approach identified lung tissue genes driving the variation in lung function and susceptibility to COPD. The identification of these genes and the pathways in which they are enriched is essential to understand the pathophysiology of airway obstruction and to identify novel therapeutic targets and biomarkers for COPD, including drugs that reverse the COPD gene signature in silico. FUNDING The research reported in this article was not specifically funded by any agency. See Acknowledgments for a full list of funders of the lung eQTL study and the Spiro-Meta CHARGE GWAS.


The Journal of Infectious Diseases | 2015

Identification of TMPRSS2 as a Susceptibility Gene for Severe 2009 Pandemic A(H1N1) Influenza and A(H7N9) Influenza

Zhong-Shan Cheng; Jie Zhou; Kelvin K. W. To; Hin Chu; Cun Li; Dong Wang; Dong Yang; Shufa Zheng; Ke Hao; Yohan Bossé; Ma'en Obeidat; Corry-Anke Brandsma; You-Qiang Song; Yu Chen; Bo-Jian Zheng; Lanjuan Li; Kwok-Yung Yuen

Abstract The genetic predisposition to severe A(H1N1)2009 (A[H1N1]pdm09) influenza was evaluated in 409 patients, including 162 cases with severe infection and 247 controls with mild infection. We prioritized candidate variants based on the result of a pilot genome-wide association study and a lung expression quantitative trait locus data set. The GG genotype of rs2070788, a higher-expression variant of TMPRSS2, was a risk variant (odds ratio, 2.11; 95% confidence interval, 1.18–3.77; P = .01) to severe A(H1N1)pdm09 influenza. A potentially functional single-nucleotide polymorphism, rs383510, accommodated in a putative regulatory region was identified to tag rs2070788. Luciferase assay results showed the putative regulatory region was a functional element, in which rs383510 regulated TMPRSS2 expression in a genotype-specific manner. Notably, rs2070788 and rs383510 were significantly associated with the susceptibility to A(H7N9) influenza in 102 patients with A(H7N9) influenza and 106 healthy controls. Therefore, we demonstrate that genetic variants with higher TMPRSS2 expression confer higher risk to severe A(H1N1)pdm09 influenza. The same variants also increase susceptibility to human A(H7N9) influenza.


American Journal of Respiratory and Critical Care Medicine | 2017

Genome-wide Interaction Analysis of Air Pollution Exposure and Childhood Asthma with Functional Follow-up

Anna Gref; Simon Kebede Merid; Olena Gruzieva; Stephane Ballereau; Allan B. Becker; Tom Bellander; Anna Bergström; Yohan Bossé; Matteo Bottai; Moira Chan-Yeung; Elaine Fuertes; Despo Ierodiakonou; Ruiwei Jiang; Stéphane Joly; Meaghan J. Jones; Michael S. Kobor; Michal Korek; Anita L. Kozyrskyj; Ashish Kumar; Nathanaël Lemonnier; Elaina MacIntyre; Camille Ménard; David C. Nickle; Ma'en Obeidat; Johann Pellet; Marie Standl; Annika Sääf; Cilla Söderhäll; Carla M.T. Tiesler; Maarten van den Berge

Rationale: The evidence supporting an association between traffic‐related air pollution exposure and incident childhood asthma is inconsistent and may depend on genetic factors. Objectives: To identify gene‐environment interaction effects on childhood asthma using genome‐wide single‐nucleotide polymorphism (SNP) data and air pollution exposure. Identified loci were further analyzed at epigenetic and transcriptomic levels. Methods: We used land use regression models to estimate individual air pollution exposure (represented by outdoor NO2 levels) at the birth address and performed a genome‐wide interaction study for doctors’ diagnoses of asthma up to 8 years in three European birth cohorts (n = 1,534) with look‐up for interaction in two separate North American cohorts, CHS (Childrens Health Study) and CAPPS/SAGE (Canadian Asthma Primary Prevention Study/Study of Asthma, Genetics and Environment) (n = 1,602 and 186 subjects, respectively). We assessed expression quantitative trait locus effects in human lung specimens and blood, as well as associations among air pollution exposure, methylation, and transcriptomic patterns. Measurements and Main Results: In the European cohorts, 186 SNPs had an interaction P < 1 × 10−4 and a look‐up evaluation of these disclosed 8 SNPs in 4 loci, with an interaction P < 0.05 in the large CHS study, but not in CAPPS/SAGE. Three SNPs within adenylate cyclase 2 (ADCY2) showed the same direction of the interaction effect and were found to influence ADCY2 gene expression in peripheral blood (P = 4.50 × 10−4). One other SNP with P < 0.05 for interaction in CHS, rs686237, strongly influenced UDP‐Gal:betaGlcNAc &bgr;‐1,4‐galactosyltransferase, polypeptide 5 (B4GALT5) expression in lung tissue (P = 1.18 × 10−17). Air pollution exposure was associated with differential discs, large homolog 2 (DLG2) methylation and expression. Conclusions: Our results indicated that gene‐environment interactions are important for asthma development and provided supportive evidence for interaction with air pollution for ADCY2, B4GALT5, and DLG2.


The Lancet Respiratory Medicine | 2017

Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: a genome-wide association study

Richard J. Allen; Joanne Porte; Rebecca Braybrooke; Carlos Flores; Tasha E. Fingerlin; Justin M. Oldham; Beatriz Guillen-Guio; Shwu Fan Ma; Tsukasa Okamoto; Alison E. John; Ma'en Obeidat; Ivana V. Yang; Amanda P. Henry; Richard Hubbard; Vidya Navaratnam; Gauri Saini; Norma Thompson; Helen Booth; Simon P. Hart; Michael Hill; Nik Hirani; Toby M. Maher; Robin J. McAnulty; Ab Millar; Philip L. Molyneaux; H Parfrey; Doris Rassl; Moira K. B. Whyte; William A. Fahy; Richard P. Marshall

Summary 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.


Human Molecular Genetics | 2015

Integrative Pathway Genomics of Lung Function and Airflow Obstruction

Sina A. Gharib; Daan W. Loth; María Soler Artigas; Timothy P. Birkland; Jemma B. Wilk; Louise V. Wain; Jennifer A. Brody; Ma'en Obeidat; Dana B. Hancock; Wenbo Tang; Rajesh Rawal; H. Marike Boezen; Medea Imboden; Jennifer E. Huffman; Lies Lahousse; Alexessander Couto Alves; Ani Manichaikul; Jennie Hui; Alanna C. Morrison; Adaikalavan Ramasamy; Albert V. Smith; Vilmundur Gudnason; Ida Surakka; Veronique Vitart; David Evans; David P. Strachan; Ian J. Deary; Albert Hofman; Sven Gläser; James F. Wilson

Chronic respiratory disorders are important contributors to the global burden of disease. Genome-wide association studies (GWASs) of lung function measures have identified several trait-associated loci, but explain only a modest portion of the phenotypic variability. We postulated that integrating pathway-based methods with GWASs of pulmonary function and airflow obstruction would identify a broader repertoire of genes and processes influencing these traits. We performed two independent GWASs of lung function and applied gene set enrichment analysis to one of the studies and validated the results using the second GWAS. We identified 131 significantly enriched gene sets associated with lung function and clustered them into larger biological modules involved in diverse processes including development, immunity, cell signaling, proliferation and arachidonic acid. We found that enrichment of gene sets was not driven by GWAS-significant variants or loci, but instead by those with less stringent association P-values. Next, we applied pathway enrichment analysis to a meta-analyzed GWAS of airflow obstruction. We identified several biologic modules that functionally overlapped with those associated with pulmonary function. However, differences were also noted, including enrichment of extracellular matrix (ECM) processes specifically in the airflow obstruction study. Network analysis of the ECM module implicated a candidate gene, matrix metalloproteinase 10 (MMP10), as a putative disease target. We used a knockout mouse model to functionally validate MMP10s role in influencing lungs susceptibility to cigarette smoke-induced emphysema. By integrating pathway analysis with population-based genomics, we unraveled biologic processes underlying pulmonary function traits and identified a candidate gene for obstructive lung disease.


Scientific Reports | 2015

Functional variants regulating LGALS1 (Galectin 1) expression affect human susceptibility to influenza A(H7N9)

Yu Chen; Jie Zhou; Zhong-Shan Cheng; Shigui Yang; Hin Chu; Yanhui Fan; Cun Li; Bosco Ho-Yin Wong; Shufa Zheng; Yixin Zhu; Fei Yu; Yiyin Wang; Xiaoli Liu; Hainv Gao; Liang Yu; Linglin Tang; Dawei Cui; Ke Hao; Yohan Bossé; Ma'en Obeidat; Corry-Anke Brandsma; You Qiang Song; Kelvin K. W. To; Pak Sham; Kwok-Yung Yuen; Lanjuan Li

The fatality of avian influenza A(H7N9) infection in humans was over 30%. To identify human genetic susceptibility to A(H7N9) infection, we performed a genome-wide association study (GWAS) involving 102 A(H7N9) patients and 106 heavily-exposed healthy poultry workers, a sample size critically restricted by the small number of human A(H7N9) cases. To tackle the stringent significance cutoff of GWAS, we utilized an artificial imputation program SnipSnip to improve the association signals. In single-SNP analysis, one of the top SNPs was rs13057866 of LGALS1. The artificial imputation (AI) identified three non-genotyped causal variants, which can be represented by three anchor/partner SNP pairs rs13057866/rs9622682 (AI P = 1.81 × 10−7), rs4820294/rs2899292 (2.13 × 10−7) and rs62236673/rs2899292 (4.25 × 10−7) respectively. Haplotype analysis of rs4820294 and rs2899292 could simulate the signal of a causal variant. The rs4820294/rs2899292 haplotype GG, in association with protection from A(H7N9) infection (OR = 0.26, P = 5.92 × 10−7) correlated to significantly higher levels of LGALS1 mRNA (P = 0.050) and protein expression (P = 0.025) in lymphoblast cell lines. Additionally, rs4820294 was mapped as an eQTL in human primary monocytes and lung tissues. In conclusion, functional variants of LGALS1 causing the expression variations are contributable to the differential susceptibility to influenza A(H7N9).


EBioMedicine | 2017

Responsiveness to Ipratropium Bromide in Male and Female Patients with Mild to Moderate Chronic Obstructive Pulmonary Disease

Xuan Li; Ma'en Obeidat; Guohai Zhou; Janice M. Leung; Donald P. Tashkin; Robert A. Wise; John E. Connett; Philippe Joubert; Yohan Bossé; Maarten van den Berge; Corry-Anke Brandsma; David C. Nickle; Ke Hao; Peter D. Paré; Don D. Sin

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.


American Journal of Respiratory Cell and Molecular Biology | 2017

Integrative Genomics of Emphysema-Associated Genes Reveals Potential Disease Biomarkers

Ma'en Obeidat; Yunlong Nie; Nick Fishbane; Xuan Li; Yohan Bossé; Philippe Joubert; David C. Nickle; Ke Hao; Dirkje S. Postma; Wim Timens; Marc A. Sze; Casey P. Shannon; Zsuzsanna Hollander; Raymond T. Ng; Bruce McManus; Stephen I. Rennard; Avrum Spira; Tillie-Louise Hackett; Wan L. Lam; Stephen Lam; Rosa Faner; Alvar Agusti; James C. Hogg; Don D. Sin; Peter D. Paré

Abstract Chronic obstructive pulmonary disease is the third leading cause of death worldwide. Gene expression profiling across multiple regions of the same lung identified genes significantly related to emphysema. We sought to determine whether the lung and epithelial expression of 127 emphysema‐related genes was also related to lung function in independent cohorts, and whether any of these genes could be used as biomarkers in the peripheral blood of patients with chronic obstructive pulmonary disease. To that end, we examined whether the expression levels of these genes were under genetic control in lung tissue (n = 1,111). We then determined whether the mRNA levels of these genes in lung tissue (n = 727), small airway epithelial cells (n = 238), and peripheral blood (n = 620) were significantly related to lung function measurements. The expression of 63 of the 127 genes (50%) was under genetic control in lung tissue. The lung and epithelial mRNA expression of a subset of the emphysema‐associated genes, including ASRGL1, LPHN2, and EDNRB, was strongly associated with lung function. In peripheral blood, the expression of 40 genes was significantly associated with lung function. Twenty‐nine of these genes (73%) were also associated with lung function in lung tissue, but with the opposite direction of effect for 24 of the 29 genes, including those involved in hypoxia and B cell‐related responses. The integrative genomics approach uncovered a significant overlap of emphysema genes associations with lung function between lung and blood with opposite directions between the two. These results support the use of peripheral blood to detect disease biomarkers.

Collaboration


Dive into the Ma'en Obeidat's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian Sayers

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Ke Hao

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian P. Hall

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennie Hui

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nick Shrine

University of Leicester

View shared research outputs
Researchain Logo
Decentralizing Knowledge