Network


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

Hotspot


Dive into the research topics where María Soler Artigas is active.

Publication


Featured researches published by María Soler Artigas.


PLOS ONE | 2011

A Comprehensive Evaluation of Potential Lung Function Associated Genes in the SpiroMeta General Population Sample

Ma’en Obeidat; Louise V. Wain; Nick Shrine; Noor Kalsheker; María Soler Artigas; Emmanouela Repapi; Paul R. Burton; Toby Johnson; Adaikalavan Ramasamy; Jing Hua Zhao; Guangju Zhai; Jennifer E. Huffman; Veronique Vitart; Eva Albrecht; Wilmar Igl; Anna-Liisa Hartikainen; Anneli Pouta; Gemma Cadby; Jennie Hui; Lyle J. Palmer; David Hadley; Wendy L. McArdle; Alicja R. Rudnicka; Inês Barroso; Ruth J. F. Loos; Nicholas J. Wareham; Massimo Mangino; Nicole Soranzo; Tim D. Spector; Sven Gläser

Rationale Lung function measures are heritable traits that predict population morbidity and mortality and are essential for the diagnosis of chronic obstructive pulmonary disease (COPD). Variations in many genes have been reported to affect these traits, but attempts at replication have provided conflicting results. Recently, we undertook a meta-analysis of Genome Wide Association Study (GWAS) results for lung function measures in 20,288 individuals from the general population (the SpiroMeta consortium). Objectives To comprehensively analyse previously reported genetic associations with lung function measures, and to investigate whether single nucleotide polymorphisms (SNPs) in these genomic regions are associated with lung function in a large population sample. Methods We analysed association for SNPs tagging 130 genes and 48 intergenic regions (+/−10 kb), after conducting a systematic review of the literature in the PubMed database for genetic association studies reporting lung function associations. Results The analysis included 16,936 genotyped and imputed SNPs. No loci showed overall significant association for FEV1 or FEV1/FVC traits using a carefully defined significance threshold of 1.3×10−5. The most significant loci associated with FEV1 include SNPs tagging MACROD2 (P = 6.81×10−5), CNTN5 (P = 4.37×10−4), and TRPV4 (P = 1.58×10−3). Among ever-smokers, SERPINA1 showed the most significant association with FEV1 (P = 8.41×10−5), followed by PDE4D (P = 1.22×10−4). The strongest association with FEV1/FVC ratio was observed with ABCC1 (P = 4.38×10−4), and ESR1 (P = 5.42×10−4) among ever-smokers. Conclusions Polymorphisms spanning previously associated lung function genes did not show strong evidence for association with lung function measures in the SpiroMeta consortium population. Common SERPINA1 polymorphisms may affect FEV1 among smokers in the general population.


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 | 2012

Genome-Wide Association Studies Identify CHRNA5/3 and HTR4 in the Development of Airflow Obstruction

Jemma B. Wilk; Nick Shrine; Laura R. Loehr; Jing Hua Zhao; Ani Manichaikul; Lorna M. Lopez; Albert V. Smith; Susan R. Heckbert; Joanna Smolonska; Wenbo Tang; Daan W. Loth; Ivan Curjuric; Jennie Hui; Michael H. Cho; Jeanne C. Latourelle; Amanda P. Henry; Melinda C. Aldrich; Per Bakke; Terri H. Beaty; Amy R. Bentley; Ingrid B. Borecki; Guy Brusselle; Kristin M. Burkart; Ting Hsu Chen; David Couper; James D. Crapo; Gail Davies; Josée Dupuis; Nora Franceschini; Amund Gulsvik

RATIONALE Genome-wide association studies (GWAS) have identified loci influencing lung function, but fewer genes influencing chronic obstructive pulmonary disease (COPD) are known. OBJECTIVES Perform meta-analyses of GWAS for airflow obstruction, a key pathophysiologic characteristic of COPD assessed by spirometry, in population-based cohorts examining all participants, ever smokers, never smokers, asthma-free participants, and more severe cases. METHODS Fifteen cohorts were studied for discovery (3,368 affected; 29,507 unaffected), and a population-based family study and a meta-analysis of case-control studies were used for replication and regional follow-up (3,837 cases; 4,479 control subjects). Airflow obstruction was defined as FEV(1) and its ratio to FVC (FEV(1)/FVC) both less than their respective lower limits of normal as determined by published reference equations. MEASUREMENTS AND MAIN RESULTS The discovery meta-analyses identified one region on chromosome 15q25.1 meeting genome-wide significance in ever smokers that includes AGPHD1, IREB2, and CHRNA5/CHRNA3 genes. The region was also modestly associated among never smokers. Gene expression studies confirmed the presence of CHRNA5/3 in lung, airway smooth muscle, and bronchial epithelial cells. A single-nucleotide polymorphism in HTR4, a gene previously related to FEV(1)/FVC, achieved genome-wide statistical significance in combined meta-analysis. Top single-nucleotide polymorphisms in ADAM19, RARB, PPAP2B, and ADAMTS19 were nominally replicated in the COPD meta-analysis. CONCLUSIONS These results suggest an important role for the CHRNA5/3 region as a genetic risk factor for airflow obstruction that may be independent of smoking and implicate the HTR4 gene in the etiology of airflow obstruction.


Nature Genetics | 2017

Genetic loci associated with chronic obstructive pulmonary disease overlap with loci for lung function and pulmonary fibrosis

Brian D. Hobbs; Kim de Jong; Maxime Lamontagne; Yohan Bossé; Nick Shrine; María Soler Artigas; Louise V. Wain; Ian P. Hall; Victoria E. Jackson; Annah B. Wyss; Stephanie J. London; Kari E. North; Nora Franceschini; David P. Strachan; Terri H. Beaty; John E. Hokanson; James D. Crapo; Peter J. Castaldi; Robert Chase; Traci M. Bartz; Susan R. Heckbert; Bruce M. Psaty; Sina A. Gharib; Pieter Zanen; Jan Willem J. Lammers; Matthijs Oudkerk; Harry J.M. Groen; Nicholas Locantore; Ruth Tal-Singer; Stephen I. Rennard

Chronic obstructive pulmonary disease (COPD) is a leading cause of mortality worldwide. We performed a genetic association study in 15,256 cases and 47,936 controls, with replication of select top results (P < 5 × 10−6) in 9,498 cases and 9,748 controls. In the combined meta-analysis, we identified 22 loci associated at genome-wide significance, including 13 new associations with COPD. Nine of these 13 loci have been associated with lung function in general population samples, while 4 (EEFSEC, DSP, MTCL1, and SFTPD) are new. We noted two loci shared with pulmonary fibrosis (FAM13A and DSP) but that had opposite risk alleles for COPD. None of our loci overlapped with genome-wide associations for asthma, although one locus has been implicated in joint susceptibility to asthma and obesity. We also identified genetic correlation between COPD and asthma. Our findings highlight new loci associated with COPD, demonstrate the importance of specific loci associated with lung function to COPD, and identify potential regions of genetic overlap between COPD and other respiratory diseases.


Nature Communications | 2015

Sixteen new lung function signals identified through 1000 Genomes Project reference panel imputation.

María Soler Artigas; Louise V. Wain; Suzanne Miller; Abdul Kader Kheirallah; Jennifer E. Huffman; Ioanna Ntalla; Nick Shrine; Ma’en Obeidat; Holly Trochet; Wendy L. McArdle; Alexessander Couto Alves; Jennie Hui; Jing Hua Zhao; Peter K. Joshi; Alexander Teumer; Eva Albrecht; Medea Imboden; Rajesh Rawal; Lorna M. Lopez; Jonathan Marten; Stefan Enroth; Ida Surakka; Ozren Polasek; Leo-Pekka Lyytikäinen; Raquel Granell; Pirro G. Hysi; Claudia Flexeder; Anubha Mahajan; John Beilby; Yohan Bossé

Lung function measures are used in the diagnosis of chronic obstructive pulmonary disease. In 38,199 European ancestry individuals, we studied genome-wide association of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC with 1000 Genomes Project (phase 1)-imputed genotypes and followed up top associations in 54,550 Europeans. We identify 14 novel loci (P<5 × 10−8) in or near ENSA, RNU5F-1, KCNS3, AK097794, ASTN2, LHX3, CCDC91, TBX3, TRIP11, RIN3, TEKT5, LTBP4, MN1 and AP1S2, and two novel signals at known loci NPNT and GPR126, providing a basis for new understanding of the genetic determinants of these traits and pulmonary diseases in which they are altered.


PLOS ONE | 2014

Large-Scale Genome-Wide Association Studies and Meta-Analyses of Longitudinal Change in Adult Lung Function

Wenbo Tang; Matthew Kowgier; Daan W. Loth; María Soler Artigas; Bonnie R. Joubert; Emily Hodge; Sina A. Gharib; Albert V. Smith; Ingo Ruczinski; Vilmundur Gudnason; Rasika A. Mathias; Tamara B. Harris; Nadia N. Hansel; Lenore J. Launer; Kathleen C. Barnes; J Hansen; Eva Albrecht; Melinda C. Aldrich; Michael Allerhand; R. Graham Barr; Guy Brusselle; David J. Couper; Ivan Curjuric; Gail Davies; Ian J. Deary; Josée Dupuis; Tove Fall; Millennia Foy; Nora Franceschini; Wei Gao

Background Genome-wide association studies (GWAS) have identified numerous loci influencing cross-sectional lung function, but less is known about genes influencing longitudinal change in lung function. Methods We performed GWAS of the rate of change in forced expiratory volume in the first second (FEV1) in 14 longitudinal, population-based cohort studies comprising 27,249 adults of European ancestry using linear mixed effects model and combined cohort-specific results using fixed effect meta-analysis to identify novel genetic loci associated with longitudinal change in lung function. Gene expression analyses were subsequently performed for identified genetic loci. As a secondary aim, we estimated the mean rate of decline in FEV1 by smoking pattern, irrespective of genotypes, across these 14 studies using meta-analysis. Results The overall meta-analysis produced suggestive evidence for association at the novel IL16/STARD5/TMC3 locus on chromosome 15 (P  =  5.71 × 10-7). In addition, meta-analysis using the five cohorts with ≥3 FEV1 measurements per participant identified the novel ME3 locus on chromosome 11 (P  =  2.18 × 10-8) at genome-wide significance. Neither locus was associated with FEV1 decline in two additional cohort studies. We confirmed gene expression of IL16, STARD5, and ME3 in multiple lung tissues. Publicly available microarray data confirmed differential expression of all three genes in lung samples from COPD patients compared with controls. Irrespective of genotypes, the combined estimate for FEV1 decline was 26.9, 29.2 and 35.7 mL/year in never, former, and persistent smokers, respectively. Conclusions In this large-scale GWAS, we identified two novel genetic loci in association with the rate of change in FEV1 that harbor candidate genes with biologically plausible functional links to lung function.


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.


PLOS ONE | 2013

GSTCD and INTS12 regulation and expression in the human lung.

Ma’en Obeidat; Suzanne Miller; Kelly Probert; Charlotte K. Billington; Amanda P. Henry; Emily Hodge; Carl P. Nelson; Ceri E. Stewart; Caroline Swan; Louise V. Wain; María Soler Artigas; Erik Melén; Kevin Ushey; Ke Hao; Maxime Lamontagne; Yohan Bossé; Dirkje S. Postma; Martin D. Tobin; Ian Sayers; Ian P. Hall

Genome-Wide Association Study (GWAS) meta-analyses have identified a strong association signal for lung function, which maps to a region on 4q24 containing two oppositely transcribed genes: glutathione S-transferase, C-terminal domain containing (GSTCD) and integrator complex subunit 12 (INTS12). Both genes were found to be expressed in a range of human airway cell types. The promoter regions and transcription start sites were determined in mRNA from human lung and a novel splice variant was identified for each gene. We obtained the following evidence for GSTCD and INTS12 co-regulation and expression: (i) correlated mRNA expression was observed both via Q-PCR and in a lung expression quantitative trait loci (eQTL) study, (ii) induction of both GSTCD and INTS12 mRNA expression in human airway smooth muscle cells was seen in response to TGFβ1, (iii) a lung eQTL study revealed that both GSTCD and INTS12 mRNA levels positively correlate with percent predicted FEV1, and (iv) FEV1 GWAS associated SNPs in 4q24 were found to act as an eQTL for INTS12 in a number of tissues. In fixed sections of human lung tissue, GSTCD protein expression was ubiquitous, whereas INTS12 expression was predominantly in epithelial cells and pneumocytes. During human fetal lung development, GSTCD protein expression was observed to be highest at the earlier pseudoglandular stage (10-12 weeks) compared with the later canalicular stage (17-19 weeks), whereas INTS12 expression levels did not alter throughout these stages. Knowledge of the transcriptional and translational regulation and expression of GSTCD and INTS12 provides important insights into the potential role of these genes in determining lung function. Future work is warranted to fully define the functions of INTS12 and GSTCD.


Human Molecular Genetics | 2012

A major recombination hotspot in the XqYq pseudoautosomal region gives new insight into processing of human gene conversion events

Shriparna Sarbajna; Matthew Denniff; Alec J. Jeffreys; Rita Neumann; María Soler Artigas; Amelia Veselis; Celia A. May

Recombination plays a fundamental role in meiosis. Non-exchange gene conversion (non-crossover, NCO) may facilitate homologue pairing, while reciprocal crossover (CO) physically connects homologues so they orientate appropriately on the meiotic spindle. In males, X-Y homologous pairing and exchange occurs within the two pseudoautosomal regions (PARs) together comprising <5% of the human sex chromosomes. Successful meiosis depends on an obligatory CO within PAR1, while the nature and role of exchange within PAR2 is unclear. Here, we describe the identification and characterization of a typical ~1 kb wide recombination hotspot within PAR2. We find that both COs and NCOs are strongly modulated in trans by the presumed chromatin remodelling protein PRDM9, and in cis by a single nucleotide polymorphism (SNP) located at the hotspot centre that appears to influence recombination initiation and which causes biased gene conversion in SNP heterozygotes. This, the largest survey to date of human NCOs reveals for the first time substantial inter-individual variation in the NCO:CO ratio. Although the extent of biased transmission at the central marker in COs is similar across men, it is highly variable among NCO recombinants. This suggests that cis-effects are mediated not only through recombination initiation frequencies varying between haplotypes but also through subsequent processing, with the potential to significantly intensify meiotic drive of hotspot-suppressing alleles. The NCO:CO ratio and extent of transmission distortion among NCOs appear to be inter-related, suggesting the existence of two NCO pathways in humans.


European Respiratory Journal | 2014

APOM and high-density lipoprotein cholesterol are associated with lung function and per cent emphysema

Kristin M. Burkart; Ani Manichaikul; Jemma B. Wilk; Firas S. Ahmed; Gregory L. Burke; Paul L. Enright; Nadia N. Hansel; Demondes Haynes; Susan R. Heckbert; Eric A. Hoffman; Joel D. Kaufman; Jun Kurai; Laura R. Loehr; Stephanie J. London; Yang Meng; George T. O'Connor; Elizabeth C. Oelsner; Marcy F. Petrini; Tess D. Pottinger; Charles A. Powell; Susan Redline; Jerome I. Rotter; Lewis J. Smith; María Soler Artigas; Martin D. Tobin; Michael Y. Tsai; Karol E. Watson; Wendy White; Taylor R. Young; Stephen S. Rich

Chronic obstructive pulmonary disease (COPD) is linked to cardiovascular disease; however, there are few studies on the associations of cardiovascular genes with COPD. We assessed the association of lung function with 2100 genes selected for cardiovascular diseases among 20 077 European-Americans and 6900 African-Americans. We performed replication of significant loci in the other racial group and an independent consortium of Europeans, tested the associations of significant loci with per cent emphysema and examined gene expression in an independent sample. We then tested the association of a related lipid biomarker with forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) ratio and per cent emphysema. We identified one new polymorphism for FEV1/FVC (rs805301) in European-Americans (p=1.3×10−6) and a second (rs707974) in the combined European-American and African-American analysis (p=1.38×10−7). Both single-nucleotide polymorphisms (SNPs) flank the gene for apolipoprotein M (APOM), a component of high-density lipoprotein (HDL) cholesterol. Both were replicated in an independent cohort. SNPs in a second gene related to apolipoprotein M and HDL, PCSK9, were associated with FEV1/FVC ratio among African-Americans. rs707974 was associated with per cent emphysema among European-Americans and African-Americans and APOM expression was related to FEV1/FVC ratio and per cent emphysema. Higher HDL levels were associated with lower FEV1/FVC ratio and greater per cent emphysema. These findings suggest a novel role for the apolipoprotein M/HDL pathway in the pathogenesis of COPD and emphysema. Findings from this study suggest a novel effect of the APOM/HDL pathway in the pathogenesis of COPD and emphysema http://ow.ly/t8Ao9

Collaboration


Dive into the María Soler Artigas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian P. Hall

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Nick Shrine

University of Leicester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennie Hui

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Daan W. Loth

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Ian Sayers

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge