Network


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

Hotspot


Dive into the research topics where Golareh Agha is active.

Publication


Featured researches published by Golareh Agha.


American Journal of Epidemiology | 2009

Life-Course Socioeconomic Position and Incidence of Coronary Heart Disease The Framingham Offspring Study

Eric B. Loucks; John Lynch; Louise Pilote; Rebecca Fuhrer; Nisha D. Almeida; Hugues Richard; Golareh Agha; Joanne M. Murabito; Emelia J. Benjamin

Cumulative exposure to socioeconomic disadvantage across the life course may be inversely associated with coronary heart disease (CHD); the mechanisms are not fully clear. An objective of this study was to determine whether cumulative life-course socioeconomic position (SEP) is associated with CHD incidence in a well-characterized US cohort that had directly assessed childhood and adulthood measures of SEP and prospectively measured CHD incidence. Furthermore, analyses aimed to evaluate whether adjustment for CHD risk factors reduces the association between cumulative life-course SEP and CHD. The authors examined 1,835 subjects who participated in the Framingham Heart Study Offspring Cohort from 1971 through 2003 (mean age, 35.0 years; 52.4% women). Childhood SEP was measured as fathers education; adulthood SEP was assessed as own education and occupation. CHD incidence included myocardial infarction, coronary insufficiency, and coronary death. Cox proportional hazards analyses indicated that cumulative SEP was associated with incident CHD after adjustment for age and sex (hazard ratio = 1.82, 95% confidence interval: 1.17, 2.85 for low vs. high cumulative SEP score). Adjustment for CHD risk factors reduced that magnitude of association (hazard ratio = 1.29, 95% confidence interval: 0.78, 2.13). These findings underscore the potential importance of CHD prevention and treatment efforts for those whose backgrounds include low SEP throughout life.


Genome Biology | 2016

DNA methylation signatures of chronic low-grade inflammation are associated with complex diseases

Symen Ligthart; Carola Marzi; Stella Aslibekyan; Michael M. Mendelson; Karen N. Conneely; Toshiko Tanaka; Elena Colicino; Lindsay L. Waite; Roby Joehanes; Weihua Guan; Jennifer A. Brody; Cathy E. Elks; Riccardo E. Marioni; Min A. Jhun; Golareh Agha; Jan Bressler; Cavin K. Ward-Caviness; Brian H. Chen; Tianxiao Huan; Kelly M. Bakulski; Elias Salfati; Giovanni Fiorito; Simone Wahl; Katharina Schramm; Jin Sha; Dena Hernandez; Allan C. Just; Jennifer A. Smith; Nona Sotoodehnia; Luke C. Pilling

BackgroundChronic low-grade inflammation reflects a subclinical immune response implicated in the pathogenesis of complex diseases. Identifying genetic loci where DNA methylation is associated with chronic low-grade inflammation may reveal novel pathways or therapeutic targets for inflammation.ResultsWe performed a meta-analysis of epigenome-wide association studies (EWAS) of serum C-reactive protein (CRP), which is a sensitive marker of low-grade inflammation, in a large European population (n = 8863) and trans-ethnic replication in African Americans (n = 4111). We found differential methylation at 218 CpG sites to be associated with CRP (P < 1.15 × 10–7) in the discovery panel of European ancestry and replicated (P < 2.29 × 10–4) 58 CpG sites (45 unique loci) among African Americans. To further characterize the molecular and clinical relevance of the findings, we examined the association with gene expression, genetic sequence variants, and clinical outcomes. DNA methylation at nine (16%) CpG sites was associated with whole blood gene expression in cis (P < 8.47 × 10–5), ten (17%) CpG sites were associated with a nearby genetic variant (P < 2.50 × 10–3), and 51 (88%) were also associated with at least one related cardiometabolic entity (P < 9.58 × 10–5). An additive weighted score of replicated CpG sites accounted for up to 6% inter-individual variation (R2) of age-adjusted and sex-adjusted CRP, independent of known CRP-related genetic variants.ConclusionWe have completed an EWAS of chronic low-grade inflammation and identified many novel genetic loci underlying inflammation that may serve as targets for the development of novel therapeutic interventions for inflammation.


International Journal of Epidemiology | 2015

Adiposity is associated with DNA methylation profile in adipose tissue

Golareh Agha; E. Andres Houseman; Karl T. Kelsey; Charles B. Eaton; Stephen L. Buka; Eric B. Loucks

BACKGROUND Adiposity is a risk factor for type 2 diabetes and cardiovascular disease, suggesting an important role for adipose tissue in the development of these conditions. The epigenetic underpinnings of adiposity are not well understood, and studies of DNA methylation in relation to adiposity have rarely focused on target adipose tissue. Objectives were to evaluate whether genome-wide DNA methylation profiles in subcutaneous adipose tissue and peripheral blood leukocytes are associated with measures of adiposity, including central fat mass, body fat distribution and body mass index. METHODS Participants were 106 men and women (mean age 47 years) from the New England Family Study. DNA methylation was evaluated using the Infinium HumanMethylation450K BeadChip. Adiposity phenotypes included dual-energy X-ray absorptiometry-assessed android fat mass, android:gynoid fat ratio and trunk:limb fat ratio, as well as body mass index. RESULTS Adipose tissue genome-wide DNA methylation profiles were associated with all four adiposity phenotypes, after adjusting for race, sex and current smoking (omnibus p-values <0.001). After further adjustment for adipose cell-mixture effects, associations with android fat mass, android:gynoid fat ratio, and trunk:limb fat ratio remained. In gene-specific analyses, adiposity phenotypes were associated with adipose tissue DNA methylation in several genes that are biologically relevant to the development of adiposity, such as AOC3, LIPE, SOD3, AQP7 and CETP. Blood DNA methylation profiles were not associated with adiposity, before or after adjustment for blood leukocyte cell mixture effects. CONCLUSION Findings show that DNA methylation patterns in adipose tissue are associated with adiposity.


Circulation Research | 2016

The Role of DNA Methylation in Cardiovascular Risk and Disease Methodological Aspects, Study Design, and Data Analysis for Epidemiological Studies

Jia Zhong; Golareh Agha; Andrea Baccarelli

Epidemiological studies have demonstrated that genetic, environmental, behavioral, and clinical factors contribute to cardiovascular disease development. How these risk factors interact at the cellular level to cause cardiovascular disease is not well known. Epigenetic epidemiology enables researchers to explore critical links between genomic coding, modifiable exposures, and manifestation of disease phenotype. One epigenetic link, DNA methylation, is potentially an important mechanism underlying these associations. In the past decade, there has been a significant increase in the number of epidemiological studies investigating cardiovascular risk factors and outcomes in relation to DNA methylation, but many gaps remain in our understanding of the underlying cause and biological implications. In this review, we provide a brief overview of the biology and mechanisms of DNA methylation and its role in cardiovascular disease. In addition, we summarize the current evidence base in epigenetic epidemiology studies relevant to cardiovascular health and disease and discuss the limitations, challenges, and future directions of the field. Finally, we provide guidelines for well-designed epigenetic epidemiology studies, with particular focus on methodological aspects, study design, and analytical challenges.


Human Molecular Genetics | 2017

Maternal BMI at the start of pregnancy and offspring epigenome-wide DNA methylation: findings from the pregnancy and childhood epigenetics (PACE) consortium

Gemma C. Sharp; Lucas A. Salas; Claire Monnereau; Catherine Allard; Paul Yousefi; Todd M. Everson; Jon Bohlin; Zongli Xu; Rae-Chi Huang; Sarah E. Reese; Cheng-Jian Xu; Nour Baïz; Cathrine Hoyo; Golareh Agha; Ritu Roy; John W. Holloway; Akram Ghantous; Simon Kebede Merid; Kelly M. Bakulski; Leanne K. Küpers; Hongmei Zhang; Rebecca C. Richmond; Christian M. Page; Liesbeth Duijts; Rolv T. Lie; Phillip E. Melton; Judith M. Vonk; Ellen Aagaard Nohr; ClarLynda R. Williams-DeVane; Karen Huen

&NA; Pre‐pregnancy maternal obesity is associated with adverse offspring outcomes at birth and later in life. Individual studies have shown that epigenetic modifications such as DNA methylation could contribute. Within the Pregnancy and Childhood Epigenetics (PACE) Consortium, we meta‐analysed the association between pre‐pregnancy maternal BMI and methylation at over 450,000 sites in newborn blood DNA, across 19 cohorts (9,340 mother‐newborn pairs). We attempted to infer causality by comparing the effects of maternal versus paternal BMI and incorporating genetic variation. In four additional cohorts (1,817 mother‐child pairs), we meta‐analysed the association between maternal BMI at the start of pregnancy and blood methylation in adolescents. In newborns, maternal BMI was associated with small (<0.2% per BMI unit (1 kg/m2), P < 1.06 × 10‐7) methylation variation at 9,044 sites throughout the genome. Adjustment for estimated cell proportions greatly attenuated the number of significant CpGs to 104, including 86 sites common to the unadjusted model. At 72/86 sites, the direction of the association was the same in newborns and adolescents, suggesting persistence of signals. However, we found evidence for acausal intrauterine effect of maternal BMI on newborn methylation at just 8/86 sites. In conclusion, this well‐powered analysis identified robust associations between maternal adiposity and variations in newborn blood DNA methylation, but these small effects may be better explained by genetic or lifestyle factors than a causal intrauterine mechanism. This highlights the need for large‐scale collaborative approaches and the application of causal inference techniques in epigenetic epidemiology.


Scientific Reports | 2017

Persistent DNA methylation changes associated with prenatal mercury exposure and cognitive performance during childhood

Andres Cardenas; Sheryl L. Rifas-Shiman; Golareh Agha; Marie-France Hivert; Augusto A. Litonjua; Dawn L. DeMeo; Xihong Lin; Chitra Amarasiriwardena; Emily Oken; Matthew W. Gillman; Andrea Baccarelli

Prenatal exposure to mercury, a known neurotoxic metal, is associated with lower cognitive performance during childhood. Disruption of fetal epigenetic programming could explain mercury’s neurodevelopmental effects. We screened for epigenome-wide methylation differences associated with maternal prenatal blood mercury levels in 321 cord blood DNA samples and examined the persistence of these alterations during early (n = 75; 2.9–4.9 years) and mid-childhood (n = 291; 6.7–10.5 years). Among males, prenatal mercury levels were associated with lower regional cord blood DNA methylation at the Paraoxonase 1 gene (PON1) that persisted in early childhood and was attenuated in mid-childhood blood. Cord blood methylation at the PON1 locus predicted lower cognitive test scores measured during early childhood. Methylation at the PON1 locus was associated with PON1 expression in an independent set of cord blood samples. The observed persistent epigenetic disruption of the PON1 gene may modulate mercury toxicity in humans and might serve as a biomarker of exposure and disease susceptibility.


Environmental Health Perspectives | 2017

Exposure to Low Levels of Lead in Utero and Umbilical Cord Blood DNA Methylation in Project Viva: An Epigenome-Wide Association Study

Shaowei Wu; Marie-France Hivert; Andres Cardenas; Jia Zhong; Sheryl L. Rifas-Shiman; Golareh Agha; Elena Colicino; Allan C. Just; Chitra Amarasiriwardena; Xihong Lin; Augusto A. Litonjua; Dawn L. DeMeo; Matthew W. Gillman; Robert O. Wright; Emily Oken; Andrea Baccarelli

Background: Early-life exposure to lead is associated with deficits in neurodevelopment and with hematopoietic system toxicity. DNA methylation may be one of the underlying mechanisms for the adverse effects of prenatal lead on the offspring, but epigenome-wide methylation data for low levels of prenatal lead exposure are lacking. Objectives: We investigated the association between prenatal maternal lead exposure and epigenome-wide DNA methylation in umbilical cord blood nucleated cells in Project Viva, a prospective U.S.-based prebirth cohort with relatively low levels of lead exposure. Methods: Among 268 mother–infant pairs, we measured lead concentrations in red blood cells (RBC) from prenatal maternal blood samples, and using HumanMethylation450 Bead Chips, we measured genome-wide methylation levels at 482,397 CpG loci in umbilical cord blood and retained 394,460 loci after quality control. After adjustment for batch effects, cell types, and covariates, we used robust linear regression models to examine associations of prenatal lead exposure with DNA methylation in cord blood at epigenome-wide significance level [false discovery rate (FDR)<0.05]. Results: The mean [standard deviation (SD)] maternal RBC lead level was 1.22 (0.63) μg/dL. CpG cg10773601 showed an epigenome-wide significant negative association with prenatal lead exposure (−1.4% per doubling increase in lead exposure; p=2.3×10−7) and was annotated to C-Type Lectin Domain Family 11, Member A (CLEC11A), which functions as a growth factor for primitive hematopoietic progenitor cells. In sex-specific analyses, we identified more CpGs with FDR<0.05 among female infants (n=38) than among male infants (n=2). One CpG (cg24637308), which showed a strong negative association with prenatal lead exposure among female infants (−4.3% per doubling increase in lead exposure; p=1.1×10−06), was annotated to Dynein Heavy Chain Domain 1 gene (DNHD1) which is highly expressed in human brain. Interestingly, there were strong correlations between blood and brain methylation for CpG (cg24637308) based on another independent set of samples with a high proportion of female participants. Conclusion: Prenatal low-level lead exposure was associated with newborn DNA methylation, particularly in female infants. https://doi.org/10.1289/EHP1246


American Journal of Cardiology | 2011

Relation of Socioeconomic Position With Ankle–Brachial Index

Golareh Agha; Joanne M. Murabito; John Lynch; Michal Abrahamowicz; Sam Harper; Eric B. Loucks

Potential upstream determinants of coronary heart disease (CHD) include life-course socioeconomic position (e.g., childhood socioeconomic circumstances, own education and occupation); however, several plausible biological mechanisms by which socioeconomic position (SEP) may influence CHD are poorly understood. Several CHD risk factors appear to be more strongly associated with SEP in women than in men; little is known as to whether any CHD risk factors may be more strongly associated with SEP in men. Objectives were to evaluate whether cumulative life-course SEP is associated with a measurement of subclinical atherosclerosis, the ankle-brachial index (ABI), in men and women. This study was a prospective analysis of 1,454 participants from the Framingham Heart Study Offspring Cohort (mean age 57 years, 53.8% women). Cumulative SEP was calculated by summing tertile scores for fathers education, own education, and own occupation. ABI was dichotomized as low (≤1.1) and normal (>1.1 to 1.4). After adjustment for age and CHD risk factors cumulative life-course SEP was associated with low ABI in men (odds ratio [OR] 2.04, 95% confidence interval [CI] 1.22 to 3.42, for low vs high cumulative SEP score) but not in women (OR 0.86, 95% CI 0.56 to 1.33). Associations with low ABI in men were substantially driven by their own education (OR 4.13, 95% CI 1.86 to 9.16, for lower vs higher than high school education). In conclusion, cumulative life-course SEP was associated with low ABI in men but not in women.


JAMA Cardiology | 2018

Association of Methylation Signals With Incident Coronary Heart Disease in an Epigenome-Wide Assessment of Circulating Tumor Necrosis Factor α

Stella Aslibekyan; Golareh Agha; Elena Colicino; Anh N. Do; Jari Lahti; Symen Ligthart; Riccardo E. Marioni; Carola Marzi; Michael M. Mendelson; Toshiko Tanaka; Matthias Wielscher; Devin Absher; Luigi Ferrucci; Oscar H. Franco; Christian Gieger; Harald Grallert; Dena Hernandez; Tianxiao Huan; Stella Iurato; Roby Joehanes; Allan C. Just; Sonja Kunze; Honghuang Lin; Chunyu Liu; James B. Meigs; Joyce B. J. van Meurs; Ann Zenobia Moore; Annette Peters; Holger Prokisch; Katri Räikkönen

Importance Tumor necrosis factor &agr; (TNF-&agr;) is a proinflammatory cytokine with manifold consequences for mammalian pathophysiology, including cardiovascular disease. A deeper understanding of TNF-&agr; biology may enhance treatment precision. Objective To conduct an epigenome-wide analysis of blood-derived DNA methylation and TNF-&agr; levels and to assess the clinical relevance of findings. Design, Setting, and Participants This meta-analysis assessed epigenome-wide associations in circulating TNF-&agr; concentrations from 5 cohort studies and 1 interventional trial, with replication in 3 additional cohort studies. Follow-up analyses investigated associations of identified methylation loci with gene expression and incident coronary heart disease; this meta-analysis included 11 461 participants who experienced 1895 coronary events. Exposures Circulating TNF-&agr; concentration. Main Outcomes and Measures DNA methylation at approximately 450 000 loci, neighboring DNA sequence variation, gene expression, and incident coronary heart disease. Results The discovery cohort included 4794 participants, and the replication study included 816 participants (overall mean [SD] age, 60.7 [8.5] years). In the discovery stage, circulating TNF-&agr; levels were associated with methylation of 7 cytosine-phosphate-guanine (CpG) sites, 3 of which were located in or near DTX3L-PARP9 at cg00959259 (&bgr; [SE] = −0.01 [0.003]; P = 7.36 × 10−8), cg08122652 (&bgr; [SE] = −0.008 [0.002]; P = 2.24 × 10−7), and cg22930808(&bgr; [SE] = −0.01 [0.002]; P = 6.92 × 10−8); NLRC5 at cg16411857 (&bgr; [SE] = −0.01 [0.002]; P = 2.14 × 10−13) and cg07839457 (&bgr; [SE] = −0.02 [0.003]; P = 6.31 × 10−10); or ABO, at cg13683939 (&bgr; [SE] = 0.04 [0.008]; P = 1.42 × 10−7) and cg24267699 (&bgr; [SE] = −0.009 [0.002]; P = 1.67 × 10−7), after accounting for multiple testing. Of these, negative associations between TNF-&agr; concentration and methylation of 2 loci in NLRC5 and 1 in DTX3L-14 PARP9 were replicated. Replicated TNF-&agr;–linked CpG sites were associated with 9% to 19% decreased risk of incident coronary heart disease per 10% higher methylation per CpG site (cg16411857: hazard ratio [HR], 0.86; 95% CI, 0.78-1.95; P = .003; cg07839457: HR, 0.89; 95% CI, 0.80-0.94; P = 3.1 × 10−5; cg00959259: HR, 0.91; 95% CI, 0.84-0.97; P = .002; cg08122652: HR, 0.81; 95% CI, 0.74-0.89; P = 2.0 × 10−5). Conclusions and Relevance We identified and replicated novel epigenetic correlates of circulating TNF-&agr; concentration in blood samples and linked these loci to coronary heart disease risk, opening opportunities for validation and therapeutic applications.


International Journal of Epidemiology | 2018

Early life disadvantage and adult adiposity: tests of sensitive periods during childhood and behavioural mediation in adulthood

Stephen E. Gilman; Yen-Tsung Huang; Marcia P. Jimenez; Golareh Agha; Su H Chu; Charles B. Eaton; Risë B. Goldstein; Karl T. Kelsey; Stephen L. Buka; Eric B. Loucks

BACKGROUND Early exposure to socioeconomic disadvantage is associated with obesity. Here we investigated how early, and conducted mediation analyses to identify behavioural factors in adulthood that could explain why. METHODS Among 931 participants in the New England Family Study, we investigated the associations of family socioeconomic disadvantage measured before birth and at age 7 years with the following measures of adiposity in mid-adulthood (mean age = 44.4 years): body mass index (BMI), waist circumference and, among 400 participants, body composition from dual-energy X-ray absorption scans. RESULTS In linear regressions adjusting for age, sex, race and childhood BMI Z-score, participants in the highest tertile of socioeconomic disadvantage at birth had 2.6 additional BMI units in adulthood [95% confidence interval (CI) = 1.26, 3.96], 5.62 cm waist circumference (95% CI = 2.69, 8.55), 0.73 kg of android fat mass (95% CI = 0.25, 1.21), and 7.65 higher Fat Mass Index (95% CI = 2.22, 13.09). Conditional on disadvantage at birth, socioeconomic disadvantage at age 7 years was not associated with adult adiposity. In mediation analyses, 10-20% of these associations were explained by educational attainment and 5-10% were explained by depressive symptoms. CONCLUSIONS Infancy may be a sensitive period for exposure to socioeconomic disadvantage, as exposure in the earliest years of life confers a larger risk for overall and central adiposity in mid-adulthood than exposure during childhood. Intervention on these two adult risk factors for adiposity would, if all model assumptions were satisfied, only remediate up to one-fifth of the excess adult adiposity among individuals born into socioeconomically disadvantaged households.

Collaboration


Dive into the Golareh Agha's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa W. Martin

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Allan C. Just

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Augusto A. Litonjua

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dawn L. DeMeo

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge