Network


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

Hotspot


Dive into the research topics where Theru A. Sivakumaran is active.

Publication


Featured researches published by Theru A. Sivakumaran.


Blood | 2013

Abnormalities in the alternative pathway of complement in children with hematopoietic stem cell transplant-associated thrombotic microangiopathy

Sonata Jodele; Christoph Licht; Jens Goebel; Bradley P. Dixon; Kejian Zhang; Theru A. Sivakumaran; Stella M. Davies; Fred G. Pluthero; Lily Lu; Benjamin L. Laskin

Hematopoietic stem cell transplant (HSCT)-associated thrombotic microangiopathy (TMA) is a complication that occurs in 25% to 35% of HSCT recipients and shares histomorphologic similarities with hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). The hallmark of all thrombotic microangiopathies is vascular endothelial cell injury of various origins, resulting in microangiopathic hemolytic anemia, platelet consumption, fibrin deposition in the microcirculation, and tissue damage. Although significant advances have been made in understanding the pathogenesis of other thrombotic microangiopathies, post-HSCT TMA remains poorly understood. We report an analysis of the complement alternative pathway, which has recently been linked to the pathogenesis of both the Shiga toxin mediated and the atypical forms of HUS, with a focus on genetic variations in the complement Factor H (CFH) gene cluster and CFH autoantibodies in six children with post-HSCT TMA. We identified a high prevalence of deletions in CFH-related genes 3 and 1 (delCFHR3-CFHR1) and CFH autoantibodies in these patients with HSCT-TMA. Conversely, CFH autoantibodies were not detected in 18 children undergoing HSCT who did not develop TMA. Our observations suggest that complement alternative pathway dysregulation may be involved in the pathogenesis of post-HSCT TMA. These findings shed light on a novel mechanism of endothelial injury in transplant-TMA and may therefore guide the development of targeted treatment interventions.


PLOS Genetics | 2010

Four Novel Loci (19q13, 6q24, 12q24, and 5q14) Influence the Microcirculation In Vivo

M. Kamran Ikram; Sim Xueling; Richard Jensen; Mary Frances Cotch; Alex W. Hewitt; M. Arfan Ikram; Jie Jin Wang; Ronald Klein; Barbara E. K. Klein; Monique M.B. Breteler; Ning Cheung; Gerald Liew; Paul Mitchell; André G. Uitterlinden; Fernando Rivadeneira; Albert Hofman; Paulus T. V. M. de Jong; Cornelia M. van Duijn; Linda Kao; Ching-Yu Cheng; Albert V. Smith; Nicole L. Glazer; Thomas Lumley; Barbara McKnight; Bruce M. Psaty; Fridbert Jonasson; Gudny Eiriksdottir; Thor Aspelund; Tamara B. Harris; Lenore J. Launer

There is increasing evidence that the microcirculation plays an important role in the pathogenesis of cardiovascular diseases. Changes in retinal vascular caliber reflect early microvascular disease and predict incident cardiovascular events. We performed a genome-wide association study to identify genetic variants associated with retinal vascular caliber. We analyzed data from four population-based discovery cohorts with 15,358 unrelated Caucasian individuals, who are members of the Cohort for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium, and replicated findings in four independent Caucasian cohorts (n = 6,652). All participants had retinal photography and retinal arteriolar and venular caliber measured from computer software. In the discovery cohorts, 179 single nucleotide polymorphisms (SNP) spread across five loci were significantly associated (p<5.0×10−8) with retinal venular caliber, but none showed association with arteriolar caliber. Collectively, these five loci explain 1.0%–3.2% of the variation in retinal venular caliber. Four out of these five loci were confirmed in independent replication samples. In the combined analyses, the top SNPs at each locus were: rs2287921 (19q13; p = 1.61×10−25, within the RASIP1 locus), rs225717 (6q24; p = 1.25×10−16, adjacent to the VTA1 and NMBR loci), rs10774625 (12q24; p = 2.15×10−13, in the region of ATXN2,SH2B3 and PTPN11 loci), and rs17421627 (5q14; p = 7.32×10−16, adjacent to the MEF2C locus). In two independent samples, locus 12q24 was also associated with coronary heart disease and hypertension. Our population-based genome-wide association study demonstrates four novel loci associated with retinal venular caliber, an endophenotype of the microcirculation associated with clinical cardiovascular disease. These data provide further insights into the contribution and biological mechanisms of microcirculatory changes that underlie cardiovascular disease.


Nature Genetics | 2012

Genome-wide association analyses identify three new susceptibility loci for primary angle closure glaucoma

Eranga N. Vithana; Chiea Chuen Khor; Chunyan Qiao; Monisha E. Nongpiur; Ronnie George; Li Jia Chen; Tan Do; Khaled K. Abu-Amero; Chor Kai Huang; Sancy Low; Liza-Sharmini Ahmad Tajudin; Shamira A. Perera; Ching-Yu Cheng; Liang Xu; Hongyan Jia; Ching-Lin Ho; Kar Seng Sim; Renyi Wu; Clement C.Y. Tham; Paul Chew; Daniel H. Su; Francis T.S. Oen; Sripriya Sarangapani; Nagaswamy Soumittra; Essam A. Osman; Hon-Tym Wong; Guangxian Tang; Sujie Fan; Hailin Meng; Dao T L Huong

Primary angle closure glaucoma (PACG) is a major cause of blindness worldwide. We conducted a genome-wide association study including 1,854 PACG cases and 9,608 controls across 5 sample collections in Asia. Replication experiments were conducted in 1,917 PACG cases and 8,943 controls collected from a further 6 sample collections. We report significant associations at three new loci: rs11024102 in PLEKHA7 (per-allele odds ratio (OR) = 1.22; P = 5.33 × 10−12), rs3753841 in COL11A1 (per-allele OR = 1.20; P = 9.22 × 10−10) and rs1015213 located between PCMTD1 and ST18 on chromosome 8q (per-allele OR = 1.50; P = 3.29 × 10−9). Our findings, accumulated across these independent worldwide collections, suggest possible mechanisms explaining the pathogenesis of PACG.


PLOS ONE | 2009

Mitochondrial DNA variants of respiratory complex I that uniquely characterize haplogroup T2 are associated with increased risk of age-related macular degeneration.

John Paul SanGiovanni; Dan E. Arking; Sudha K. Iyengar; Michael Elashoff; Traci E. Clemons; George F. Reed; Alice K. Henning; Theru A. Sivakumaran; Xuming Xu; Andrew T. DeWan; Elvira Agrón; Elena Rochtchina; Carolyn M. Sue; Jie Jin Wang; Paul Mitchell; Josephine Hoh; Peter J. Francis; Michael L. Klein; Emily Y. Chew; Aravinda Chakravarti

Background Age-related macular degeneration (AMD), a chronic neurodegenerative and neovascular retinal disease, is the leading cause of blindness in elderly people of western European origin. While structural and functional alterations in mitochondria (mt) and their metabolites have been implicated in the pathogenesis of chronic neurodegenerative and vascular diseases, the relationship of inherited variants in the mitochondrial genome and mt haplogroup subtypes with advanced AMD has not been reported in large prospective cohorts. Methodology/Prinicipal Findings We examined the relationship of inherited mtDNA variants with advanced AMD in 1168 people using a three-stage design on samples from 12-year and 10-year prospective studies on the natural history of age-related eye disease. In Stage I we resequenced the entire genome in 99 elderly AMD-free controls and 215 people with advanced AMD from the 12-year study. A consistent association with AMD in 14 of 17 SNPs characterizing the mtDNA T haplogroup emerged. Further analysis revealed these associations were driven entirely by the T2 haplogroup, and characterized by two variants in Complex I genes (A11812G of MT-ND4 and A14233G of MT-ND6). We genotyped T haplogroups in an independent sample of 490 cases and 61 controls from the same study (Stage II) and in 56 cases and 246 controls from the 10-year study (Stage III). People in the T2 haplogroup were approximately 2.5 times more likely to have advanced AMD than their peers (odds ratio [OR] = 2.54, 95%CI 1.36–4.80, P≤0.004) after considering the totality of evidence. Findings persisted after considering the impact of AMD-associated variants A69S and Y402H (OR = 5.19, 95%CI 1.19–22.69, P≤0.029). Conclusion Loci defining the mtDNA T2 haplogroup and Complex I are reasonable targets for novel functional analyses and therapeutic research in AMD.


JAMA Ophthalmology | 2014

Markers of Inflammation, Oxidative Stress, and Endothelial Dysfunction and the 20-Year Cumulative Incidence of Early Age-Related Macular Degeneration: The Beaver Dam Eye Study

Ronald Klein; Chelsea E. Myers; Karen J. Cruickshanks; Ronald E. Gangnon; Lorraine G. Danforth; Theru A. Sivakumaran; Sudha K. Iyengar; Michael Y. Tsai; Barbara E. K. Klein

IMPORTANCE Modifying levels of factors associated with age-related macular degeneration (AMD) may decrease the risk for visual impairment in older persons. OBJECTIVE To examine the relationships of markers of inflammation, oxidative stress, and endothelial dysfunction to the 20-year cumulative incidence of early AMD. DESIGN, SETTING, AND PARTICIPANTS This longitudinal population-based cohort study involved a random sample of 975 persons in the Beaver Dam Eye Study without signs of AMD who participated in the baseline examination in 1988-1990 and up to 4 follow-up examinations in 1993-1995, 1998-2000, 2003-2005, and 2008-2010. EXPOSURES Serum markers of inflammation (high-sensitivity C-reactive protein, tumor necrosis factor-α receptor 2, interleukin-6, and white blood cell count), oxidative stress (8-isoprostane and total carbonyl content), and endothelial dysfunction (soluble vascular cell adhesion molecule-1 and soluble intercellular adhesion molecule-1) were measured. Interactions with complement factor H (rs1061170), age-related maculopathy susceptibility 2 (rs10490924), complement component 3 (rs2230199), and complement component 2/complement factor B (rs4151667) were examined using multiplicative models. Age-related macular degeneration was assessed from fundus photographs. MAIN OUTCOMES AND MEASURES Early AMD defined by the presence of any size drusen and the presence of pigmentary abnormalities or by the presence of large-sized drusen (≥125-μm diameter) in the absence of late AMD. RESULTS The 20-year cumulative incidence of early AMD was 23.0%. Adjusting for age, sex, and other risk factors, high-sensitivity C-reactive protein (odds ratio comparing fourth with first quartile, 2.18; P = .005), tumor necrosis factor-α receptor 2 (odds ratio, 1.78; P = .04), and interleukin-6 (odds ratio, 1.78; P = .03) were associated with the incidence of early AMD. Increased incidence of early AMD was associated with soluble vascular cell adhesion molecule-1 (odds ratio per SD on the logarithmic scale, 1.21; P = .04). CONCLUSIONS AND RELEVANCE We found modest evidence of relationships of serum high-sensitivity C-reactive protein, tumor necrosis factor-α receptor 2, interleukin-6, and soluble vascular cell adhesion molecule-1 to the 20-year cumulative incidence of early AMD independent of age, smoking status, and other factors. It is not known whether these associations represent a cause and effect relationship or whether other unknown confounders accounted for the findings. Even if inflammatory processes are a cause of early AMD, it is not known whether interventions that reduce systemic inflammatory processes will reduce the incidence of early AMD.


Investigative Ophthalmology & Visual Science | 2008

Toll-like Receptor Polymorphisms and Age-Related Macular Degeneration

Albert O. Edwards; Dequan Chen; Brooke L. Fridley; Katherine M. James; Yanhong Wu; Gonçalo R. Abecasis; Anand Swaroop; Mohammad Othman; Kari Branham; Sudha K. Iyengar; Theru A. Sivakumaran; Ronald Klein; Barbara E. K. Klein; Nirubol Tosakulwong

PURPOSE Evidence from genetic-association studies in conjunction with the demonstration of complement deposition in the retina and choroid implicates noncellular pathways of innate immunity in the pathogenesis of age-related macular degeneration (AMD). The purpose of this study was to determine whether common variation in the 10 human toll-like receptors (TLRs) alters the risk of AMD. METHODS Sixty-eight SNPs were iteratively genotyped across the TLR genes in a cohort of 577 subjects, with and without AMD. Two additional cohorts were used for replication studies. Standard genetic-association methods were used to analyze the results for association with disease and interaction with other loci. RESULTS Coding SNPs in TLR3 (rs3775291) and TLR7 (rs179008) showed association with AMD in one group (P = 0.01 and P = 0.02, respectively) before correction for multiple testing. For both SNPs, the association with AMD arose due to an excess of heterozygotes compared with homozygotes for the major allele. The two coding SNPs were not associated with AMD in another case-control cohort or an extended-family cohort. Although an intronic SNP in TLR4 was associated marginally with AMD (P = 0.03), it was not possible to replicate a previous association with the rare coding SNP D299G in this gene (P = 0.6). CONCLUSIONS Although borderline support for association between polymorphisms in TLR genes and AMD was reported for some cohorts, these initial observations of coding SNPs in TLR3, TLR4, and TLR7 were not replicated. TLR variants are unlikely to have a major impact on overall AMD risk, and the common variants studied were not associated with AMD.


American Journal of Epidemiology | 2008

Combined Effects of Complement Factor H Genotypes, Fish Consumption, and Inflammatory Markers on Long-Term Risk for Age-related Macular Degeneration in a Cohort

Jie Jin Wang; Elena Rochtchina; Wayne Smith; Ronald Klein; Barbara E. K. Klein; Tripti Joshi; Theru A. Sivakumaran; Sudha K. Iyengar; Paul Mitchell

At baseline in 1992-1994, the authors assessed the combined effects of complement factor H (CFH) genotypes with smoking, fish consumption, and inflammatory markers on the risk of age-related macular degeneration (AMD) in 3,654 persons aged > or =49 years. They reexamined 75% of the survivors after 5 and 10 years, confirming incident AMD by side-by-side photographic grading. Of the 2,452 persons followed in the Blue Mountains Eye Study, 1,881 were genotyped (rs1061170), with CC, CT, and TT identified in 13.6%, 46.7%, and 39.7%, respectively. AMD risk increased with each additional C allele (early AMD: age- and sex-adjusted relative risk (RR) = 1.6, 95% confidence interval (CI): 1.2, 1.9; late AMD: RR = 2.3, 95% CI: 1.5, 3.6). Late AMD risk among current smokers with the CC/CT genotypes (RR = 10.7, 95% CI: 3.4, 33.9) was 5-fold that for genotypically similar nonsmokers (RR = 2.2, 95% CI: 0.9, 5.5) versus current nonsmokers with TT genotypes. Weekly compared with less than weekly consumption of fish was associated with reduced late AMD risk in participants with the CC genotype (RR = 0.15, 95% CI: 0.03, 0.8) but not the CT (RR = 0.7, 95% CI: 0.3, 2.0) or TT (RR = 1.3, 95% CI: 0.2, 7.2) genotypes. This study documents joint contributions from genetic and systemic factors in determining the progression of AMD.


Genes and Immunity | 2010

Genome-wide association identifies SKIV2L and MYRIP as protective factors for age-related macular degeneration

Laura J. Kopplin; Robert P. Igo; Yang Wang; Theru A. Sivakumaran; Stephanie A. Hagstrom; Neal S. Peachey; Peter J. Francis; Michael L. Klein; John Paul SanGiovanni; Emily Y. Chew; Gayle J. T. Pauer; Gwen M. Sturgill; Tripti Joshi; Liping Tian; Quansheng Xi; Alice K. Henning; Kristine E. Lee; Ronald Klein; Barbara Ek Klein; Sudha K. Iyengar

Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly in the developed world. We conducted a genome-wide association study in a series of families enriched for AMD and completed a meta-analysis of this new data with results from reanalysis of an existing study of a late-stage case–control cohort. We tested the top findings for replication in 1896 cases and 1866 controls and identified two novel genetic protective factors for AMD. In addition to the complement factor H (CFH) (P=2.3 × 10−64) and age-related maculopathy susceptibility 2 (ARMS2) (P=1.2 × 10−60) loci, we observed a protective effect at rs429608, an intronic SNP in SKIV2L (P=5.3 × 10−15), a gene near the complement component 2 (C2)/complement factor B (BF) locus, that indicates the protective effect may be mediated by variants other than the C2/BF variants previously studied. Haplotype analysis at this locus identified three protective haplotypes defined by the rs429608 protective allele. We also identified a new potentially protective effect at rs2679798 in MYRIP (P=2.9 × 10−4), a gene involved in retinal pigment epithelium melanosome trafficking. Interestingly, MYRIP was initially identified in the family-based scan and was confirmed in the case–control set. From these efforts, we report the identification of two novel protective factors for AMD and confirm the previously known associations at CFH, ARMS2 and C3.


American Journal of Ophthalmology | 2014

Lipids, Lipid Genes, and Incident Age-Related Macular Degeneration: The Three Continent Age-Related Macular Degeneration Consortium

Ronald Klein; Chelsea E. Myers; Gabriëlle H.S. Buitendijk; Elena Rochtchina; Xiaoyi Gao; Paulus T. V. M. de Jong; Theru A. Sivakumaran; George Burlutsky; Roberta McKean-Cowdin; Albert Hofman; Sudha K. Iyengar; Kristine E. Lee; Bruno H. Stricker; Johannes R. Vingerling; Paul Mitchell; Barbara E. K. Klein; Caroline C. W. Klaver; Jie Jin Wang

PURPOSE To describe associations of serum lipid levels and lipid pathway genes to the incidence of age-related macular degeneration (AMD). DESIGN Meta-analysis. METHODS setting: Three population-based cohorts. population: A total of 6950 participants from the Beaver Dam Eye Study (BDES), Blue Mountains Eye Study (BMES), and Rotterdam Study (RS). observation procedures: Participants were followed over 20 years and examined at 5-year intervals. Hazard ratios associated with lipid levels per standard deviation above the mean or associated with each additional risk allele for each lipid pathway gene were calculated using random-effects inverse-weighted meta-analysis models, adjusting for known AMD risk factors. main outcome measures: Incidence of AMD. RESULTS The average 5-year incidences of early AMD were 8.1%, 15.1%, and 13.0% in the BDES, BMES, and RS, respectively. Substantial heterogeneity in the effect of cholesterol and lipid pathway genes on the incidence and progression of AMD was evident when the data from the 3 studies were combined in meta-analysis. After correction for multiple comparisons, we did not find a statistically significant association between any of the cholesterol measures, statin use, or serum lipid genes and any of the AMD outcomes in the meta-analysis. CONCLUSION In a meta-analysis, there were no associations of cholesterol measures, history of statin use, or lipid pathway genes to the incidence and progression of AMD. These findings add to inconsistencies in earlier reports from our studies and others showing weak associations, no associations, or inverse associations of high-density lipoprotein cholesterol and total cholesterol with AMD.


PLOS ONE | 2011

A 32 kb critical region excluding Y402H in CFH mediates risk for age-related macular degeneration

Theru A. Sivakumaran; Robert P. Igo; Jeffrey M. Kidd; Andy Itsara; Laura J. Kopplin; Wei Chen; Stephanie A. Hagstrom; Neal S. Peachey; Peter J. Francis; Michael L. Klein; Emily Y. Chew; Vedam Lakshmi Ramprasad; Wan Ting Tay; Paul Mitchell; Mark Seielstad; Dwight Stambolian; Albert O. Edwards; Kristine E. Lee; Dmitry V. Leontiev; Gyungah Jun; Yang Wang; Liping Tian; Feiyou Qiu; Alice K. Henning; Thomas LaFramboise; Parveen Sen; Manoharan Aarthi; Ronnie George; Rajiv Raman; Manmath K Das

Complement factor H shows very strong association with Age-related Macular Degeneration (AMD), and recent data suggest that multiple causal variants are associated with disease. To refine the location of the disease associated variants, we characterized in detail the structural variation at CFH and its paralogs, including two copy number polymorphisms (CNP), CNP147 and CNP148, and several rare deletions and duplications. Examination of 34 AMD-enriched extended families (N = 293) and AMD cases (White N = 4210 Indian = 134; Malay = 140) and controls (White N = 3229; Indian = 117; Malay = 2390) demonstrated that deletion CNP148 was protective against AMD, independent of SNPs at CFH. Regression analysis of seven common haplotypes showed three haplotypes, H1, H6 and H7, as conferring risk for AMD development. Being the most common haplotype H1 confers the greatest risk by increasing the odds of AMD by 2.75-fold (95% CI = [2.51, 3.01]; p = 8.31×10−109); Caucasian (H6) and Indian-specific (H7) recombinant haplotypes increase the odds of AMD by 1.85-fold (p = 3.52×10−9) and by 15.57-fold (P = 0.007), respectively. We identified a 32-kb region downstream of Y402H (rs1061170), shared by all three risk haplotypes, suggesting that this region may be critical for AMD development. Further analysis showed that two SNPs within the 32 kb block, rs1329428 and rs203687, optimally explain disease association. rs1329428 resides in 20 kb unique sequence block, but rs203687 resides in a 12 kb block that is 89% similar to a noncoding region contained in ΔCNP148. We conclude that causal variation in this region potentially encompasses both regulatory effects at single markers and copy number.

Collaboration


Dive into the Theru A. Sivakumaran's collaboration.

Top Co-Authors

Avatar

Ronald Klein

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Sudha K. Iyengar

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Barbara E. K. Klein

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ronald E. Gangnon

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kristine E. Lee

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Chelsea E. Myers

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jie Jin Wang

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Karen J. Cruickshanks

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge