Network


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

Hotspot


Dive into the research topics where Sonia Shah is active.

Publication


Featured researches published by Sonia Shah.


Genome Biology | 2015

DNA methylation age of blood predicts all-cause mortality in later life

Riccardo E. Marioni; Sonia Shah; Allan F. McRae; Brian H. Chen; Elena Colicino; Sarah E. Harris; Jude Gibson; Anjali K. Henders; Paul Redmond; Simon R. Cox; Alison Pattie; Janie Corley; Lee Murphy; Nicholas G. Martin; Grant W. Montgomery; Andrew P. Feinberg; M. Daniele Fallin; Michael L Multhaup; Andrew E. Jaffe; Roby Joehanes; Joel Schwartz; Allan C. Just; Kathryn L. Lunetta; Joanne M. Murabito; Steve Horvath; Andrea Baccarelli; Daniel Levy; Peter M. Visscher; Naomi R. Wray; Ian J. Deary

BackgroundDNA methylation levels change with age. Recent studies have identified biomarkers of chronological age based on DNA methylation levels. It is not yet known whether DNA methylation age captures aspects of biological age.ResultsHere we test whether differences between people’s chronological ages and estimated ages, DNA methylation age, predict all-cause mortality in later life. The difference between DNA methylation age and chronological age (Δage) was calculated in four longitudinal cohorts of older people. Meta-analysis of proportional hazards models from the four cohorts was used to determine the association between Δage and mortality. A 5-year higher Δage is associated with a 21% higher mortality risk, adjusting for age and sex. After further adjustments for childhood IQ, education, social class, hypertension, diabetes, cardiovascular disease, and APOE e4 status, there is a 16% increased mortality risk for those with a 5-year higher Δage. A pedigree-based heritability analysis of Δage was conducted in a separate cohort. The heritability of Δage was 0.43.ConclusionsDNA methylation-derived measures of accelerated aging are heritable traits that predict mortality independently of health status, lifestyle factors, and known genetic factors.


The Lancet | 2013

Use of low-density lipoprotein cholesterol gene score to distinguish patients with polygenic and monogenic familial hypercholesterolaemia: a case-control study

Philippa J. Talmud; Sonia Shah; Ros Whittall; Marta Futema; Philip Howard; Jackie A. Cooper; Seamus C. Harrison; KaWah Li; Fotios Drenos; Frederik Karpe; H. Andrew W. Neil; Olivier S. Descamps; Claudia Langenberg; Nicholas Lench; Mika Kivimäki; John C. Whittaker; Aroon D. Hingorani; Meena Kumari; Steve E. Humphries

BACKGROUND Familial hypercholesterolaemia is a common autosomal-dominant disorder caused by mutations in three known genes. DNA-based cascade testing is recommended by UK guidelines to identify affected relatives; however, about 60% of patients are mutation-negative. We assessed the hypothesis that familial hypercholesterolaemia can also be caused by an accumulation of common small-effect LDL-C-raising alleles. METHODS In November, 2011, we assembled a sample of patients with familial hypercholesterolaemia from three UK-based sources and compared them with a healthy control sample from the UK Whitehall II (WHII) study. We also studied patients from a Belgian lipid clinic (Hôpital de Jolimont, Haine St-Paul, Belgium) for validation analyses. We genotyped participants for 12 common LDL-C-raising alleles identified by the Global Lipid Genetics Consortium and constructed a weighted LDL-C-raising gene score. We compared the gene score distribution among patients with familial hypercholesterolaemia with no confirmed mutation, those with an identified mutation, and controls from WHII. FINDINGS We recruited 321 mutation-negative UK patients (451 Belgian), 319 mutation-positive UK patients (273 Belgian), and 3020 controls from WHII. The mean weighted LDL-C gene score of the WHII participants (0.90 [SD 0.23]) was strongly associated with LDL-C concentration (p=1.4 x 10(-77); R(2)=0.11). Mutation-negative UK patients had a significantly higher mean weighted LDL-C score (1.0 [SD 0.21]) than did WHII controls (p=4.5 x 10(-16)), as did the mutation-negative Belgian patients (0.99 [0.19]; p=5.2 x 10(-20)). The score was also higher in UK (0.95 [0.20]; p=1.6 x 10(-5)) and Belgian (0.92 [0.20]; p=0.04) mutation-positive patients than in WHII controls. 167 (52%) of 321 mutation-negative UK patients had a score within the top three deciles of the WHII weighted LDL-C gene score distribution, and only 35 (11%) fell within the lowest three deciles. INTERPRETATION In a substantial proportion of patients with familial hypercholesterolaemia without a known mutation, their raised LDL-C concentrations might have a polygenic cause, which could compromise the efficiency of cascade testing. In patients with a detected mutation, a substantial polygenic contribution might add to the variable penetrance of the disease. FUNDING British Heart Foundation, Pfizer, AstraZeneca, Schering-Plough, National Institute for Health Research, Medical Research Council, Health and Safety Executive, Department of Health, National Heart Lung and Blood Institute, National Institute on Aging, Agency for Health Care Policy Research, John D and Catherine T MacArthur Foundation Research Networks on Successful Midlife Development and Socio-economic Status and Health, Unilever, and Departments of Health and Trade and Industry.


European Heart Journal | 2015

Mendelian randomization of blood lipids for coronary heart disease.

Michael V. Holmes; Folkert W. Asselbergs; Tom Palmer; Fotios Drenos; Matthew B. Lanktree; Christopher P. Nelson; Caroline Dale; Sandosh Padmanabhan; Chris Finan; Daniel I. Swerdlow; Vinicius Tragante; Erik P A Van Iperen; Suthesh Sivapalaratnam; Sonia Shah; Clara C. Elbers; Tina Shah; Jorgen Engmann; Claudia Giambartolomei; Jon White; Delilah Zabaneh; Reecha Sofat; Stela McLachlan; Pieter A. Doevendans; Anthony J. Balmforth; Alistair S. Hall; Kari E. North; Berta Almoguera; Ron C. Hoogeveen; Mary Cushman; Myriam Fornage

Aims To investigate the causal role of high-density lipoprotein cholesterol (HDL-C) and triglycerides in coronary heart disease (CHD) using multiple instrumental variables for Mendelian randomization. Methods and results We developed weighted allele scores based on single nucleotide polymorphisms (SNPs) with established associations with HDL-C, triglycerides, and low-density lipoprotein cholesterol (LDL-C). For each trait, we constructed two scores. The first was unrestricted, including all independent SNPs associated with the lipid trait identified from a prior meta-analysis (threshold P < 2 × 10−6); and the second a restricted score, filtered to remove any SNPs also associated with either of the other two lipid traits at P ≤ 0.01. Mendelian randomization meta-analyses were conducted in 17 studies including 62,199 participants and 12,099 CHD events. Both the unrestricted and restricted allele scores for LDL-C (42 and 19 SNPs, respectively) associated with CHD. For HDL-C, the unrestricted allele score (48 SNPs) was associated with CHD (OR: 0.53; 95% CI: 0.40, 0.70), per 1 mmol/L higher HDL-C, but neither the restricted allele score (19 SNPs; OR: 0.91; 95% CI: 0.42, 1.98) nor the unrestricted HDL-C allele score adjusted for triglycerides, LDL-C, or statin use (OR: 0.81; 95% CI: 0.44, 1.46) showed a robust association. For triglycerides, the unrestricted allele score (67 SNPs) and the restricted allele score (27 SNPs) were both associated with CHD (OR: 1.62; 95% CI: 1.24, 2.11 and 1.61; 95% CI: 1.00, 2.59, respectively) per 1-log unit increment. However, the unrestricted triglyceride score adjusted for HDL-C, LDL-C, and statin use gave an OR for CHD of 1.01 (95% CI: 0.59, 1.75). Conclusion The genetic findings support a causal effect of triglycerides on CHD risk, but a causal role for HDL-C, though possible, remains less certain.


Blood | 2011

Transcriptomic analyses of murine resolution-phase macrophages

Melanie Stables; Sonia Shah; Evelyn Camon; Ruth C. Lovering; Justine Newson; Jonas Bystrom; Stuart N. Farrow; Derek W. Gilroy

Macrophages are either classically (M1) or alternatively-activated (M2). Whereas this nomenclature was generated from monocyte-derived macrophages treated in vitro with defined cytokine stimuli, the phenotype of in vivo-derived macrophages is less understood. We completed Affymetrix-based transcriptomic analysis of macrophages from the resolution phase of a zymosan-induced peritonitis. Compared with macrophages from hyperinflamed mice possessing a pro-inflammatory nature as well as naive macrophages from the uninflamed peritoneum, resolution-phase macrophages (rM) are similar to monocyte-derived dendritic cells (DCs), being CD209a positive but lacking CD11c. They are enriched for antigen processing/presentation (MHC class II [H2-Eb1, H2-Ab1, H2-Ob, H2-Aa], CD74, CD86), secrete T- and B-lymphocyte chemokines (Xcl1, Ccl5, Cxcl13) as well as factors that enhance macrophage/DC development, and promote DC/T cell synapse formation (Clec2i, Tnfsf4, Clcf1). rM are also enriched for cell cycle/proliferation genes as well as Alox15, Timd4, and Tgfb2, key systems in the termination of leukocyte trafficking and clearance of inflammatory cells. Finally, comparison with in vitro-derived M1/M2 shows that rM are neither classically nor alternatively activated but possess aspects of both definitions consistent with an immune regulatory phenotype. We propose that macrophages in situ cannot be rigidly categorized as they can express many shades of the inflammatory spectrum determined by tissue, stimulus, and phase of inflammation.


American Journal of Human Genetics | 2009

Gene-centric Association Signals for Lipids and Apolipoproteins Identified via the HumanCVD BeadChip

Philippa J. Talmud; Fotios Drenos; Sonia Shah; Tina Shah; Jutta Palmen; Claudio Verzilli; Tom R. Gaunt; Jacky Pallas; Ruth C. Lovering; KaWah Li; Juan P. Casas; Reecha Sofat; Meena Kumari; Santiago Rodriguez; Toby Johnson; Stephen Newhouse; Anna F. Dominiczak; Nilesh J. Samani; Mark J. Caulfield; Peter Sever; Alice Stanton; Denis C. Shields; Sandosh Padmanabhan; Olle Melander; Claire E. Hastie; Christian Delles; Shah Ebrahim; Michael Marmot; George Davey Smith; Debbie A. Lawlor

Blood lipids are important cardiovascular disease (CVD) risk factors with both genetic and environmental determinants. The Whitehall II study (n=5592) was genotyped with the gene-centric HumanCVD BeadChip (Illumina). We identified 195 SNPs in 16 genes/regions associated with 3 major lipid fractions and 2 apolipoprotein components at p<10(-5), with the associations being broadly concordant with prior genome-wide analysis. SNPs associated with LDL cholesterol and apolipoprotein B were located in LDLR, PCSK9, APOB, CELSR2, HMGCR, CETP, the TOMM40-APOE-C1-C2-C4 cluster, and the APOA5-A4-C3-A1 cluster; SNPs associated with HDL cholesterol and apolipoprotein AI were in CETP, LPL, LIPC, APOA5-A4-C3-A1, and ABCA1; and SNPs associated with triglycerides in GCKR, BAZ1B, MLXIPL, LPL, and APOA5-A4-C3-A1. For 48 SNPs in previously unreported loci that were significant at p<10(-4) in Whitehall II, in silico analysis including the British Womens Heart and Health Study, BRIGHT, ASCOT, and NORDIL studies (total n>12,500) revealed previously unreported associations of SH2B3 (p<2.2x10(-6)), BMPR2 (p<2.3x10(-7)), BCL3/PVRL2 (flanking APOE; p<4.4x10(-8)), and SMARCA4 (flanking LDLR; p<2.5x10(-7)) with LDL cholesterol. Common alleles in these genes explained 6.1%-14.7% of the variance in the five lipid-related traits, and individuals at opposite tails of the additive allele score exhibited substantial differences in trait levels (e.g., >1 mmol/L in LDL cholesterol [approximately 1 SD of the trait distribution]). These data suggest that multiple common alleles of small effect can make important contributions to individual differences in blood lipids potentially relevant to the assessment of CVD risk. These genes provide further insights into lipid metabolism and the likely effects of modifying the encoded targets therapeutically.


International Journal of Epidemiology | 2015

The epigenetic clock is correlated with physical and cognitive fitness in the Lothian Birth Cohort 1936

Riccardo E. Marioni; Sonia Shah; Allan F. McRae; Stuart J Ritchie; Graciela Muniz-Terrera; Sarah E. Harris; Jude Gibson; Paul Redmond; Simon R. Cox; Alison Pattie; J. Corley; Adele Taylor; Lee Murphy; John M. Starr; Steve Horvath; Peter M. Visscher; Naomi R. Wray; Ian J. Deary

Background: The DNA methylation-based ‘epigenetic clock’ correlates strongly with chronological age, but it is currently unclear what drives individual differences. We examine cross-sectional and longitudinal associations between the epigenetic clock and four mortality-linked markers of physical and mental fitness: lung function, walking speed, grip strength and cognitive ability. Methods: DNA methylation-based age acceleration (residuals of the epigenetic clock estimate regressed on chronological age) were estimated in the Lothian Birth Cohort 1936 at ages 70 (n = 920), 73 (n = 299) and 76 (n = 273) years. General cognitive ability, walking speed, lung function and grip strength were measured concurrently. Cross-sectional correlations between age acceleration and the fitness variables were calculated. Longitudinal change in the epigenetic clock estimates and the fitness variables were assessed via linear mixed models and latent growth curves. Epigenetic age acceleration at age 70 was used as a predictor of longitudinal change in fitness. Epigenome-wide association studies (EWASs) were conducted on the four fitness measures. Results: Cross-sectional correlations were significant between greater age acceleration and poorer performance on the lung function, cognition and grip strength measures (r range: −0.07 to −0.05, P range: 9.7 x 10−3 to 0.024). All of the fitness variables declined over time but age acceleration did not correlate with subsequent change over 6 years. There were no EWAS hits for the fitness traits. Conclusions: Markers of physical and mental fitness are associated with the epigenetic clock (lower abilities associated with age acceleration). However, age acceleration does not associate with decline in these measures, at least over a relatively short follow-up.


Aging (Albany NY) , 8 (9) pp. 1844-1865. (2016) | 2016

DNA methylation-based measures of biological age: meta-analysis predicting time to death.

Brian H. Chen; Riccardo E. Marioni; Elena Colicino; Marjolein J. Peters; Cavin K. Ward-Caviness; Pei-Chien Tsai; Nicholas S. Roetker; Allan C. Just; Ellen W. Demerath; Weihua Guan; Jan Bressler; Myriam Fornage; Stephanie A. Studenski; Amy Vandiver; Ann Zenobia Moore; Toshiko Tanaka; Douglas P. Kiel; Liming Liang; Pantel S. Vokonas; Joel Schwartz; Kathryn L. Lunetta; Joanne M. Murabito; Stefania Bandinelli; Dena Hernandez; David Melzer; Michael A. Nalls; Luke C. Pilling; Timothy R. Price; Andrew Singleton; Christian Gieger

Estimates of biological age based on DNA methylation patterns, often referred to as “epigenetic age”, “DNAm age”, have been shown to be robust biomarkers of age in humans. We previously demonstrated that independent of chronological age, epigenetic age assessed in blood predicted all-cause mortality in four human cohorts. Here, we expanded our original observation to 13 different cohorts for a total sample size of 13,089 individuals, including three racial/ethnic groups. In addition, we examined whether incorporating information on blood cell composition into the epigenetic age metrics improves their predictive power for mortality. All considered measures of epigenetic age acceleration were predictive of mortality (p≤8.2×10−9), independent of chronological age, even after adjusting for additional risk factors (p<5.4×10−4), and within the racial/ethnic groups that we examined (non-Hispanic whites, Hispanics, African Americans). Epigenetic age estimates that incorporated information on blood cell composition led to the smallest p-values for time to death (p=7.5×10−43). Overall, this study a) strengthens the evidence that epigenetic age predicts all-cause mortality above and beyond chronological age and traditional risk factors, and b) demonstrates that epigenetic age estimates that incorporate information on blood cell counts lead to highly significant associations with all-cause mortality.


Circulation-cardiovascular Genetics | 2016

Epigenetic Signatures of Cigarette Smoking

Roby Joehanes; Allan C. Just; Riccardo E. Marioni; Luke C. Pilling; Lindsay M. Reynolds; Pooja R. Mandaviya; Weihua Guan; Tao Xu; Cathy E. Elks; Stella Aslibekyan; Hortensia Moreno-Macías; Jennifer A. Smith; Jennifer A. Brody; Radhika Dhingra; Paul Yousefi; James S. Pankow; Sonja Kunze; Sonia Shah; Allan F. McRae; Kurt Lohman; Jin Sha; Devin M. Absher; Luigi Ferrucci; Wei Zhao; Ellen W. Demerath; Jan Bressler; Megan L. Grove; Tianxiao Huan; Chunyu Liu; Michael M. Mendelson

Background—DNA methylation leaves a long-term signature of smoking exposure and is one potential mechanism by which tobacco exposure predisposes to adverse health outcomes, such as cancers, osteoporosis, lung, and cardiovascular disorders. Methods and Results—To comprehensively determine the association between cigarette smoking and DNA methylation, we conducted a meta-analysis of genome-wide DNA methylation assessed using the Illumina BeadChip 450K array on 15 907 blood-derived DNA samples from participants in 16 cohorts (including 2433 current, 6518 former, and 6956 never smokers). Comparing current versus never smokers, 2623 cytosine–phosphate–guanine sites (CpGs), annotated to 1405 genes, were statistically significantly differentially methylated at Bonferroni threshold of P<1×10−7 (18 760 CpGs at false discovery rate <0.05). Genes annotated to these CpGs were enriched for associations with several smoking-related traits in genome-wide studies including pulmonary function, cancers, inflammatory diseases, and heart disease. Comparing former versus never smokers, 185 of the CpGs that differed between current and never smokers were significant P<1×10−7 (2623 CpGs at false discovery rate <0.05), indicating a pattern of persistent altered methylation, with attenuation, after smoking cessation. Transcriptomic integration identified effects on gene expression at many differentially methylated CpGs. Conclusions—Cigarette smoking has a broad impact on genome-wide methylation that, at many loci, persists many years after smoking cessation. Many of the differentially methylated genes were novel genes with respect to biological effects of smoking and might represent therapeutic targets for prevention or treatment of tobacco-related diseases. Methylation at these sites could also serve as sensitive and stable biomarkers of lifetime exposure to tobacco smoke.


Genome Biology | 2014

Contribution of genetic variation to transgenerational inheritance of DNA methylation

Allan F. McRae; Joseph E. Powell; Anjali K. Henders; Lisa Bowdler; Gibran Hemani; Sonia Shah; Jodie N. Painter; Nicholas G. Martin; Peter M. Visscher; Grant W. Montgomery

BackgroundDespite the important role DNA methylation plays in transcriptional regulation, the transgenerational inheritance of DNA methylation is not well understood. The genetic heritability of DNA methylation has been estimated using twin pairs, although concern has been expressed whether the underlying assumption of equal common environmental effects are applicable due to intrauterine differences between monozygotic and dizygotic twins. We estimate the heritability of DNA methylation on peripheral blood leukocytes using Illumina HumanMethylation450 array using a family based sample of 614 people from 117 families, allowing comparison both within and across generations.ResultsThe correlations from the various available relative pairs indicate that on average the similarity in DNA methylation between relatives is predominantly due to genetic effects with any common environmental or zygotic effects being limited. The average heritability of DNA methylation measured at probes with no known SNPs is estimated as 0.187. The ten most heritable methylation probes were investigated with a genome-wide association study, all showing highly statistically significant cis mQTLs. Further investigation of one of these cis mQTL, found in the MHC region of chromosome 6, showed the most significantly associated SNP was also associated with over 200 other DNA methylation probes in this region and the gene expression level of 9 genes.ConclusionsThe majority of transgenerational similarity in DNA methylation is attributable to genetic effects, and approximately 20% of individual differences in DNA methylation in the population are caused by DNA sequence variation that is not located within CpG sites.


Clinical Chemistry | 2015

Refinement of Variant Selection for the LDL Cholesterol Genetic Risk Score in the Diagnosis of the Polygenic Form of Clinical Familial Hypercholesterolemia and Replication in Samples from 6 Countries

Marta Futema; Sonia Shah; Jackie A. Cooper; KaWah Li; Ros Whittall; Mahtab Sharifi; Olivia Goldberg; Euridiki Drogari; Vasiliki Mollaki; Albert Wiegman; Joep C. Defesche; Maria Nicoletta D'Agostino; Antonietta D'Angelo; Paolo Rubba; Giuliana Fortunato; Małgorzata Waluś-Miarka; Robert A. Hegele; Mary Aderayo Bamimore; Ronen Durst; Eran Leitersdorf; Monique Mulder; Jeanine E. Roeters van Lennep; Eric J.G. Sijbrands; John C. Whittaker; Philippa J. Talmud; Steve E. Humphries

BACKGROUND Familial hypercholesterolemia (FH) is an autosomal-dominant disorder caused by mutations in 1 of 3 genes. In the 60% of patients who are mutation negative, we have recently shown that the clinical phenotype can be associated with an accumulation of common small-effect LDL cholesterol (LDL-C)-raising alleles by use of a 12-single nucleotide polymorphism (12-SNP) score. The aims of the study were to improve the selection of SNPs and replicate the results in additional samples. METHODS We used ROC curves to determine the optimum number of LDL-C SNPs. For replication analysis, we genotyped patients with a clinical diagnosis of FH from 6 countries for 6 LDL-C-associated alleles. We compared the weighted SNP score among patients with no confirmed mutation (FH/M-), those with a mutation (FH/M+), and controls from a UK population sample (WHII). RESULTS Increasing the number of SNPs to 33 did not improve the ability of the score to discriminate between FH/M- and controls, whereas sequential removal of SNPs with smaller effects/lower frequency showed that a weighted score of 6 SNPs performed as well as the 12-SNP score. Metaanalysis of the weighted 6-SNP score, on the basis of polymorphisms in CELSR2 (cadherin, EGF LAG 7-pass G-type receptor 2), APOB (apolipoprotein B), ABCG5/8 [ATP-binding cassette, sub-family G (WHITE), member 5/8], LDLR (low density lipoprotein receptor), and APOE (apolipoprotein E) loci, in the independent FH/M- cohorts showed a consistently higher score in comparison to the WHII population (P < 2.2 × 10(-16)). Modeling in individuals with a 6-SNP score in the top three-fourths of the score distribution indicated a >95% likelihood of a polygenic explanation of their increased LDL-C. CONCLUSIONS A 6-SNP LDL-C score consistently distinguishes FH/M- patients from healthy individuals. The hypercholesterolemia in 88% of mutation-negative patients is likely to have a polygenic basis.

Collaboration


Dive into the Sonia Shah's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mika Kivimäki

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tina Shah

University College London

View shared research outputs
Top Co-Authors

Avatar

Allan F. McRae

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge