Network


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

Hotspot


Dive into the research topics where Jihane Romanos is active.

Publication


Featured researches published by Jihane Romanos.


Nature Genetics | 2008

Newly identified genetic risk variants for celiac disease related to the immune response

Karen A. Hunt; Alexandra Zhernakova; Graham Turner; Graham A. Heap; Lude Franke; Marcel Bruinenberg; Jihane Romanos; Lotte C. Dinesen; Anthony W. Ryan; Davinder Panesar; Rhian Gwilliam; Fumihiko Takeuchi; William M. McLaren; Geoffrey Holmes; Peter D. Howdle; Julian R. Walters; David S. Sanders; Raymond J. Playford; Gosia Trynka; Chris Jj Mulder; M. Luisa Mearin; Wieke H. Verbeek; Valerie Trimble; Fiona M. Stevens; Colm O'Morain; N. P. Kennedy; Dermot Kelleher; Daniel J. Pennington; David P. Strachan; Wendy L. McArdle

Our genome-wide association study of celiac disease previously identified risk variants in the IL2–IL21 region. To identify additional risk variants, we genotyped 1,020 of the most strongly associated non-HLA markers in an additional 1,643 cases and 3,406 controls. Through joint analysis including the genome-wide association study data (767 cases, 1,422 controls), we identified seven previously unknown risk regions (P < 5 × 10−7). Six regions harbor genes controlling immune responses, including CCR3, IL12A, IL18RAP, RGS1, SH2B3 (nsSNP rs3184504) and TAGAP. Whole-blood IL18RAP mRNA expression correlated with IL18RAP genotype. Type 1 diabetes and celiac disease share HLA-DQ, IL2–IL21, CCR3 and SH2B3 risk regions. Thus, this extensive genome-wide association follow-up study has identified additional celiac disease risk variants in relevant biological pathways.


Nature Genetics | 2011

Dense genotyping identifies and localizes multiple common and rare variant association signals in celiac disease.

Gosia Trynka; Karen A. Hunt; Nicholas A. Bockett; Jihane Romanos; Vanisha Mistry; Agata Szperl; Sjoerd F. Bakker; Maria Teresa Bardella; Leena Bhaw-Rosun; Gemma Castillejo; Emilio G. de la Concha; Rodrigo Coutinho de Almeida; Kerith Rae M Dias; Cleo C. van Diemen; P Dubois; Richard H. Duerr; Sarah Edkins; Lude Franke; Karin Fransen; Javier Gutierrez; Graham A. Heap; Barbara Hrdlickova; Sarah Hunt; Leticia Plaza Izurieta; Valentina Izzo; Leo A. B. Joosten; Cordelia Langford; Maria Cristina Mazzilli; Charles A. Mein; Vandana Midah

Using variants from the 1000 Genomes Project pilot European CEU dataset and data from additional resequencing studies, we densely genotyped 183 non-HLA risk loci previously associated with immune-mediated diseases in 12,041 individuals with celiac disease (cases) and 12,228 controls. We identified 13 new celiac disease risk loci reaching genome-wide significance, bringing the number of known loci (including the HLA locus) to 40. We found multiple independent association signals at over one-third of these loci, a finding that is attributable to a combination of common, low-frequency and rare genetic variants. Compared to previously available data such as those from HapMap3, our dense genotyping in a large sample collection provided a higher resolution of the pattern of linkage disequilibrium and suggested localization of many signals to finer scale regions. In particular, 29 of the 54 fine-mapped signals seemed to be localized to single genes and, in some instances, to gene regulatory elements. Altogether, we define the complex genetic architecture of the risk regions of and refine the risk signals for celiac disease, providing the next step toward uncovering the causal mechanisms of the disease.


PLOS Genetics | 2011

Meta-Analysis of Genome-Wide Association Studies in Celiac Disease and Rheumatoid Arthritis Identifies Fourteen Non-HLA Shared Loci

Alexandra Zhernakova; Eli A. Stahl; Gosia Trynka; Soumya Raychaudhuri; Eleanora A. Festen; Lude Franke; Harm-Jan Westra; Rudolf S. N. Fehrmann; Fina Kurreeman; Brian Thomson; Namrata Gupta; Jihane Romanos; Ross McManus; Anthony W. Ryan; Graham Turner; E. Brouwer; Marcel D. Posthumus; Elaine F. Remmers; Francesca Tucci; René E. M. Toes; Elvira Grandone; Maria Cristina Mazzilli; Anna Rybak; Bozena Cukrowska; Marieke J. H. Coenen; Timothy R. D. J. Radstake; Piet L. C. M. van Riel; Yonghong Li; Paul I. W. de Bakker; Peter K. Gregersen

Epidemiology and candidate gene studies indicate a shared genetic basis for celiac disease (CD) and rheumatoid arthritis (RA), but the extent of this sharing has not been systematically explored. Previous studies demonstrate that 6 of the established non-HLA CD and RA risk loci (out of 26 loci for each disease) are shared between both diseases. We hypothesized that there are additional shared risk alleles and that combining genome-wide association study (GWAS) data from each disease would increase power to identify these shared risk alleles. We performed a meta-analysis of two published GWAS on CD (4,533 cases and 10,750 controls) and RA (5,539 cases and 17,231 controls). After genotyping the top associated SNPs in 2,169 CD cases and 2,255 controls, and 2,845 RA cases and 4,944 controls, 8 additional SNPs demonstrated P<5×10−8 in a combined analysis of all 50,266 samples, including four SNPs that have not been previously confirmed in either disease: rs10892279 near the DDX6 gene (Pcombined = 1.2×10−12), rs864537 near CD247 (Pcombined = 2.2×10−11), rs2298428 near UBE2L3 (Pcombined = 2.5×10−10), and rs11203203 near UBASH3A (Pcombined = 1.1×10−8). We also confirmed that 4 gene loci previously established in either CD or RA are associated with the other autoimmune disease at combined P<5×10−8 (SH2B3, 8q24, STAT4, and TRAF1-C5). From the 14 shared gene loci, 7 SNPs showed a genome-wide significant effect on expression of one or more transcripts in the linkage disequilibrium (LD) block around the SNP. These associations implicate antigen presentation and T-cell activation as a shared mechanism of disease pathogenesis and underscore the utility of cross-disease meta-analysis for identification of genetic risk factors with pleiotropic effects between two clinically distinct diseases.


The New England Journal of Medicine | 2014

Randomized Feeding Intervention in Infants at High Risk for Celiac Disease

Sabine L. Vriezinga; Renata Auricchio; E. Bravi; Gemma Castillejo; Anna Chmielewska; P. Crespo Escobar; Sanja Kolaček; S. Koletzko; Ilma Rita Korponay-Szabó; E. Mummert; Isabel Polanco; Hein Putter; Carmen Ribes-Koninckx; Raanan Shamir; H. Szajewska; Katharina J. Werkstetter; Luigi Greco; Judit Gyimesi; Corina Hartman; C. Hogen Esch; E.G.D. Hopman; Anneli Ivarsson; T. Koltai; Frits Koning; Eva Martínez-Ojinaga; C. te Marvelde; A. Mocic Pavic; Jihane Romanos; E. Stoopman; Vincenzo Villanacci

BACKGROUND A window of opportunity has been suggested for reducing the risk of celiac disease by introducing gluten to infants at 4 to 6 months of age. METHODS We performed a multicenter, randomized, double-blind, placebo-controlled dietary-intervention study involving 944 children who were positive for HLA-DQ2 or HLA-DQ8 and had at least one first-degree relative with celiac disease. From 16 to 24 weeks of age, 475 participants received 100 mg of immunologically active gluten daily, and 469 received placebo. Anti-transglutaminase type 2 and antigliadin antibodies were periodically measured. The primary outcome was the frequency of biopsy-confirmed celiac disease at 3 years of age. RESULTS Celiac disease was confirmed by means of biopsies in 77 children. To avoid underestimation of the frequency of celiac disease, 3 additional children who received a diagnosis of celiac disease according to the 2012 European Society for Pediatric Gastroenterology, Hepatology, and Nutrition diagnostic criteria (without having undergone biopsies) were included in the analyses (80 children; median age, 2.8 years; 59% were girls). The cumulative incidence of celiac disease among patients 3 years of age was 5.2% (95% confidence interval [CI], 3.6 to 6.8), with similar rates in the gluten group and the placebo group (5.9% [95% CI, 3.7 to 8.1] and 4.5% [95% CI, 2.5 to 6.5], respectively; hazard ratio in the gluten group, 1.23; 95% CI, 0.79 to 1.91). Rates of elevated levels of anti-transglutaminase type 2 and antigliadin antibodies were also similar in the two study groups (7.0% [95% CI, 4.7 to 9.4] in the gluten group and 5.7% [95% CI, 3.5 to 7.9] in the placebo group; hazard ratio, 1.14; 95% CI, 0.76 to 1.73). Breast-feeding, regardless of whether it was exclusive or whether it was ongoing during gluten introduction, did not significantly influence the development of celiac disease or the effect of the intervention. CONCLUSIONS As compared with placebo, the introduction of small quantities of gluten at 16 to 24 weeks of age did not reduce the risk of celiac disease by 3 years of age in this group of high-risk children. (Funded by the European Commission and others; PreventCD Current Controlled Trials number, ISRCTN74582487.).


Gut | 2009

Coeliac disease-associated risk variants in TNFAIP3 and REL implicate altered NF-κB signalling

Gosia Trynka; Alexandra Zhernakova; Jihane Romanos; Lude Franke; Karen A. Hunt; Graham Turner; Marcel Bruinenberg; Graham A. Heap; M Platteel; Anthony W. Ryan; C. de Kovel; Geoffrey Holmes; Peter D. Howdle; Julian R. Walters; David S. Sanders; Chris Jj Mulder; M L Mearin; Wieke H. Verbeek; Valerie Trimble; Fiona M. Stevens; Dermot Kelleher; Donatella Barisani; Maria Teresa Bardella; Ross McManus; D A van Heel; Cisca Wijmenga

Objective: Our previous coeliac disease genome-wide association study (GWAS) implicated risk variants in the human leucocyte antigen (HLA) region and eight novel risk regions. To identify more coeliac disease loci, we selected 458 single nucleotide polymorphisms (SNPs) that showed more modest association in the GWAS for genotyping and analysis in four independent cohorts. Design: 458 SNPs were assayed in 1682 cases and 3258 controls from three populations (UK, Irish and Dutch). We combined the results with the original GWAS cohort (767 UK cases and 1422 controls); six SNPs showed association with p<1×10−04 and were then genotyped in an independent Italian coeliac cohort (538 cases and 593 controls). Results: We identified two novel coeliac disease risk regions: 6q23.3 (OLIG3-TNFAIP3) and 2p16.1 (REL), both of which reached genome-wide significance in the combined analysis of all 2987 cases and 5273 controls (rs2327832 p = 1.3×10−08, and rs842647 p = 5.2×10−07). We investigated the expression of these genes in the RNA isolated from biopsies and from whole blood RNA. We did not observe any changes in gene expression, nor in the correlation of genotype with gene expression. Conclusions: Both TNFAIP3 (A20, at the protein level) and REL are key mediators in the nuclear factor kappa B (NF-κB) inflammatory signalling pathway. For the first time, a role for primary heritable variation in this important biological pathway predisposing to coeliac disease has been identified. Currently, the HLA risk factors and the 10 established non-HLA risk factors explain ∼40% of the heritability of coeliac disease.


American Journal of Human Genetics | 2010

Evolutionary and Functional Analysis of Celiac Risk Loci Reveals SH2B3 as a Protective Factor against Bacterial Infection

Alexandra Zhernakova; Clara C. Elbers; Bart Ferwerda; Jihane Romanos; Gosia Trynka; P Dubois; Carolien G.F. de Kovel; Lude Franke; Marije Oosting; Donatella Barisani; Maria Teresa Bardella; Leo A. B. Joosten; Päivi Saavalainen; David A. van Heel; Carlo Catassi; Mihai G. Netea; Cisca Wijmenga

Celiac disease (CD) is an intolerance to dietary proteins of wheat, barley, and rye. CD may have substantial morbidity, yet it is quite common with a prevalence of 1%-2% in Western populations. It is not clear why the CD phenotype is so prevalent despite its negative effects on human health, especially because appropriate treatment in the form of a gluten-free diet has only been available since the 1950s, when dietary gluten was discovered to be the triggering factor. The high prevalence of CD might suggest that genes underlying this disease may have been favored by the process of natural selection. We assessed signatures of selection for ten confirmed CD-associated loci in several genome-wide data sets, comprising 8154 controls from four European populations and 195 individuals from a North African population, by studying haplotype lengths via the integrated haplotype score (iHS) method. Consistent signs of positive selection for CD-associated derived alleles were observed in three loci: IL12A, IL18RAP, and SH2B3. For the SH2B3 risk allele, we also show a difference in allele frequency distribution (Fst) between HapMap phase II populations. Functional investigation of the effect of the SH2B3 genotype in response to lipopolysaccharide and muramyl dipeptide revealed that carriers of the SH2B3 rs3184504*A risk allele showed stronger activation of the NOD2 recognition pathway. This suggests that SH2B3 plays a role in protection against bacteria infection, and it provides a possible explanation for the selective sweep on SH2B3, which occurred sometime between 1200 and 1700 years ago.


PLOS ONE | 2008

Effective Detection of Human Leukocyte Antigen Risk Alleles in Celiac Disease Using Tag Single Nucleotide Polymorphisms

Alienke J. Monsuur; Paul I. W. de Bakker; Alexandra Zhernakova; Dalila Pinto; Willem Verduijn; Jihane Romanos; Renata Auricchio; Ana D. Lopez; David A. van Heel; J. Bart A. Crusius; Cisca Wijmenga

Background The HLA genes, located in the MHC region on chromosome 6p21.3, play an important role in many autoimmune disorders, such as celiac disease (CD), type 1 diabetes (T1D), rheumatoid arthritis, multiple sclerosis, psoriasis and others. Known HLA variants that confer risk to CD, for example, include DQA1*05/DQB1*02 (DQ2.5) and DQA1*03/DQB1*0302 (DQ8). To diagnose the majority of CD patients and to study disease susceptibility and progression, typing these strongly associated HLA risk factors is of utmost importance. However, current genotyping methods for HLA risk factors involve many reactions, and are complicated and expensive. We sought a simple experimental approach using tagging SNPs that predict the CD-associated HLA risk factors. Methodology Our tagging approach exploits linkage disequilibrium between single nucleotide polymorphism (SNPs) and the CD-associated HLA risk factors DQ2.5 and DQ8 that indicate direct risk, and DQA1*0201/DQB1*0202 (DQ2.2) and DQA1*0505/DQB1*0301 (DQ7) that attribute to the risk of DQ2.5 to CD. To evaluate the predictive power of this approach, we performed an empirical comparison of the predicted DQ types, based on these six tag SNPs, with those executed with current validated laboratory typing methods of the HLA-DQA1 and -DQB1 genes in three large cohorts. The results were validated in three European celiac populations. Conclusion Using this method, only six SNPs were needed to predict the risk types carried by >95% of CD patients. We determined that for this tagging approach the sensitivity was >0.991, specificity >0.996 and the predictive value >0.948. Our results show that this tag SNP method is very accurate and provides an excellent basis for population screening for CD. This method is broadly applicable in European populations.


Gastroenterology | 2009

Analysis of HLA and Non-HLA Alleles Can Identify Individuals at High Risk for Celiac Disease

Jihane Romanos; Cleo C. van Diemen; Ilja M. Nolte; Gosia Trynka; Alexandra Zhernakova; Jingyuan Fu; Maria Teresa Bardella; Donatella Barisani; Ross McManus; David A. van Heel; Cisca Wijmenga

BACKGROUND & AIMS Celiac disease (CD) is a common chronic disorder of the small intestine, resulting from aberrant cellular responses to gluten peptides, and often remains undiagnosed. It is a complex genetic disorder, although 95% of the patients carry the risk heterodimer human leukocyte antigen (HLA)-DQ2. Genome-wide association studies on CD have identified 9 non-HLA loci that also contribute to CD risk, most of which are shared with other immune-related diseases. Our aim is to predict the genetic risk for CD using HLA and non-HLA risk alleles. METHODS We selected 10 independent polymorphisms in 2,308 cases and 4,585 controls from Dutch, UK, and Irish populations and categorized the individuals into 3 risk groups, based on their HLA-DQ2 genotype. We used the summed number of non-HLA risk alleles per individual to analyze their cumulative effect on CD risk, adjusting for gender and population group in logistic regression analysis. We validated our findings in 436 Italian cases and 532 controls. RESULTS CD cases carried more non-HLA risk alleles than controls: individuals carrying > or = 13 risk alleles had a higher CD risk (odds ratio, 6.2; 95% confidence interval, 4.1-9.3) compared with those carrying 0-5 risk alleles. Combining HLA and non-HLA risk genotypes in one model increases sensitivity by 6.2% compared with using only HLA for identification of high-risk individuals with slight decrease in specificity. CONCLUSIONS We can use non-HLA risk factors for CD to improve identification of high-risk individuals. Our risk model is a first step toward better diagnosis and prognosis in high-risk families and population-based screening.


Gut | 2014

Improving coeliac disease risk prediction by testing non-HLA variants additional to HLA variants

Jihane Romanos; Anna Rosén; Vinod Kumar; Gosia Trynka; Lude Franke; Agata Szperl; Javier Gutierrez-Achury; Cleo C. van Diemen; Roan Kanninga; Soesma A. Jankipersadsing; Andrea K. Steck; Georges Eisenbarth; David A. van Heel; Bozena Cukrowska; Valentina Bruno; Maria Cristina Mazzilli; Concepción Núñez; Jose Ramon Bilbao; M. Luisa Mearin; Donatella Barisani; Marian Rewers; Jill M. Norris; Anneli Ivarsson; H. Marieke Boezen; Edwin Liu; Cisca Wijmenga

Background The majority of coeliac disease (CD) patients are not being properly diagnosed and therefore remain untreated, leading to a greater risk of developing CD-associated complications. The major genetic risk heterodimer, HLA-DQ2 and DQ8, is already used clinically to help exclude disease. However, approximately 40% of the population carry these alleles and the majority never develop CD. Objective We explored whether CD risk prediction can be improved by adding non-HLA-susceptible variants to common HLA testing. Design We developed an average weighted genetic risk score with 10, 26 and 57 single nucleotide polymorphisms (SNP) in 2675 cases and 2815 controls and assessed the improvement in risk prediction provided by the non-HLA SNP. Moreover, we assessed the transferability of the genetic risk model with 26 non-HLA variants to a nested case–control population (n=1709) and a prospective cohort (n=1245) and then tested how well this model predicted CD outcome for 985 independent individuals. Results Adding 57 non-HLA variants to HLA testing showed a statistically significant improvement compared to scores from models based on HLA only, HLA plus 10 SNP and HLA plus 26 SNP. With 57 non-HLA variants, the area under the receiver operator characteristic curve reached 0.854 compared to 0.823 for HLA only, and 11.1% of individuals were reclassified to a more accurate risk group. We show that the risk model with HLA plus 26 SNP is useful in independent populations. Conclusions Predicting risk with 57 additional non-HLA variants improved the identification of potential CD patients. This demonstrates a possible role for combined HLA and non-HLA genetic testing in diagnostic work for CD.


Journal of Medical Genetics | 2008

Six new coeliac disease loci replicated in an Italian population confirm association with coeliac disease

Jihane Romanos; Donatella Barisani; Gosia Trynka; Alessandra Zhernakova; Maria Teresa Bardella; Cisca Wijmenga

Background and aims: The first genome wide association study on coeliac disease (CD) and its follow-up have identified eight new loci that contribute significantly towards CD risk. Seven of these loci contain genes controlling adaptive immune responses, including IL2/IL21 (4q27), RGS1 (1q31), IL18RAP (2q11–2q12), CCR3 (3p21), IL12A (3q25–3q26), TAGAP (6q25) and SH2B3 (12q24). Methods: We selected the nine most associated single nucleotide polymorphisms to tag the eight new loci in an Italian cohort comprising 538 CD patients and 593 healthy controls. Results: Common variation in IL2/IL21, RGS1, IL12A/SCHIP and SH2B3 was associated with susceptibility to CD in our Italian cohort. The LPP and TAGAP regions also showed moderate association, whereas there was no association with CCR3 and IL18RAP. Conclusion: This is the first replication study of six of the eight new CD loci; it is also the first CD association study in a southern European cohort. Our results may imply there is a genuine population difference across Europe regarding the loci contributing to CD.

Collaboration


Dive into the Jihane Romanos's collaboration.

Top Co-Authors

Avatar

Cisca Wijmenga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Gosia Trynka

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Alexandra Zhernakova

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Lude Franke

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Maria Teresa Bardella

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Karen A. Hunt

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Graham A. Heap

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Donatella Barisani

University of Milano-Bicocca

View shared research outputs
Top Co-Authors

Avatar

David A. van Heel

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

P Dubois

University of Cambridge

View shared research outputs
Researchain Logo
Decentralizing Knowledge