Try a new search

Format these results:

Searched for:

person:chakra01

in-biosketch:yes

Total Results:

518


Genome-wide pleiotropy analysis identifies novel blood pressure variants and improves its polygenic risk scores

Zhu, Xiaofeng; Zhu, Luke; Wang, Heming; Cooper, Richard S; Chakravarti, Aravinda
Systolic and diastolic blood pressure (S/DBP) are highly correlated modifiable risk factors for cardiovascular disease (CVD). We report here a bidirectional Mendelian Randomization (MR) and horizontal pleiotropy analysis of S/DBP summary statistics from the UK Biobank (UKB)-International Consortium for Blood Pressure (ICBP) (UKB-ICBP) BP genome-wide association study and construct a composite genetic risk score (GRS) by including pleiotropic variants. The composite GRS captures greater (1.11-3.26 fold) heritability for BP traits and increases (1.09- and 2.01-fold) Nagelkerke's R2 for hypertension and CVD. We replicated 118 novel BP horizontal pleiotropic variants including 18 novel BP loci using summary statistics from the Million Veteran Program (MVP) study. An additional 219 novel BP signals and 40 novel loci were identified after a meta-analysis of the UKB-ICBP and MVP summary statistics but without further independent replication. Our study provides further insight into BP regulation and provides a novel way to construct a GRS by including pleiotropic variants for other complex diseases.
PMID: 34989438
ISSN: 1098-2272
CID: 5107252

Rare coding variants in RCN3 are associated with blood pressure

He, Karen Y; Kelly, Tanika N; Wang, Heming; Liang, Jingjing; Zhu, Luke; Cade, Brian E; Assimes, Themistocles L; Becker, Lewis C; Beitelshees, Amber L; Bielak, Lawrence F; Bress, Adam P; Brody, Jennifer A; Chang, Yen-Pei Christy; Chang, Yi-Cheng; de Vries, Paul S; Duggirala, Ravindranath; Fox, Ervin R; Franceschini, Nora; Furniss, Anna L; Gao, Yan; Guo, Xiuqing; Haessler, Jeffrey; Hung, Yi-Jen; Hwang, Shih-Jen; Irvin, Marguerite Ryan; Kalyani, Rita R; Liu, Ching-Ti; Liu, Chunyu; Martin, Lisa Warsinger; Montasser, May E; Muntner, Paul M; Mwasongwe, Stanford; Naseri, Take; Palmas, Walter; Reupena, Muagututi'a Sefuiva; Rice, Kenneth M; Sheu, Wayne H-H; Shimbo, Daichi; Smith, Jennifer A; Snively, Beverly M; Yanek, Lisa R; Zhao, Wei; Blangero, John; Boerwinkle, Eric; Chen, Yii-Der Ida; Correa, Adolfo; Cupples, L Adrienne; Curran, Joanne E; Fornage, Myriam; He, Jiang; Hou, Lifang; Kaplan, Robert C; Kardia, Sharon L R; Kenny, Eimear E; Kooperberg, Charles; Lloyd-Jones, Donald; Loos, Ruth J F; Mathias, Rasika A; McGarvey, Stephen T; Mitchell, Braxton D; North, Kari E; Peyser, Patricia A; Psaty, Bruce M; Raffield, Laura M; Rao, D C; Redline, Susan; Reiner, Alex P; Rich, Stephen S; Rotter, Jerome I; Taylor, Kent D; Tracy, Russell; Vasan, Ramachandran S; Morrison, Alanna C; Levy, Daniel; Chakravarti, Aravinda; Arnett, Donna K; Zhu, Xiaofeng
BACKGROUND:While large genome-wide association studies have identified nearly one thousand loci associated with variation in blood pressure, rare variant identification is still a challenge. In family-based cohorts, genome-wide linkage scans have been successful in identifying rare genetic variants for blood pressure. This study aims to identify low frequency and rare genetic variants within previously reported linkage regions on chromosomes 1 and 19 in African American families from the Trans-Omics for Precision Medicine (TOPMed) program. Genetic association analyses weighted by linkage evidence were completed with whole genome sequencing data within and across TOPMed ancestral groups consisting of 60,388 individuals of European, African, East Asian, Hispanic, and Samoan ancestries. RESULTS:). CONCLUSIONS:Low frequency and rare variants in RCN3 contributes blood pressure variation. This study demonstrates that focusing association analyses in linkage regions greatly reduces multiple-testing burden and improves power to identify novel rare variants associated with blood pressure traits.
PMCID:8858539
PMID: 35183128
ISSN: 1471-2164
CID: 5174962

Multiple, independent, common variants at RET, SEMA3 and NRG1 gut enhancers specify Hirschsprung disease risk in European ancestry subjects

Kapoor, Ashish; Nandakumar, Priyanka; Auer, Dallas R; Sosa, Maria X; Ross, Holly; Bollinger, Juli; Yan, Jia; Berrios, Courtney; Chakravarti, Aravinda
PURPOSE/OBJECTIVE:Hirschsprung disease (HSCR) is a developmental disorder of the enteric nervous system (ENS) characterized by congenital aganglionosis arising from coding variants in ENS genes causing partial or total loss-of-function. Low-penetrance, common, noncoding variants at RET, SEMA3 and NRG1 loci are also associated with HSCR, with small-to-moderate loss of gene expression mediated through sequence variants in cis-regulatory elements (CRE) as another causal mechanism. Since these latter variants are common, many individuals carry multiple risk variants. However, the extent and combinatorial effects of all putative CRE variants within and across these loci on HSCR is unknown. METHODS:Using 583 HSCR subjects, one of the largest samples of European ancestry studied, and genotyping 56 tag variants, we evaluated association of all common variants overlapping putative gut CREs and fine-mapped causal variants at RET, SEMA3 and NRG1. RESULTS:We demonstrate that 28 and 8 tag variants, several of which are genetically independent, overlap putative-enhancers at the RET and SEMA3 loci, respectively, as well as two fine-mapped tag variants at the NRG1 locus, are significantly associated with HSCR. Importantly, disease risk increases with increasing numbers of risk alleles from multiple variants within and across these loci, varying >25-fold across individuals. CONCLUSION/CONCLUSIONS:This increasing allele number-dependent risk, we hypothesize, arises from HSCR-relevant ENS cells sensing the reduced gene expression at multiple ENS genes since their developmental effects are integrated through gene regulatory networks.
PMID: 34006365
ISSN: 1531-5037
CID: 4889482

A multi-enhancer RET regulatory code is disrupted in Hirschsprung disease

Chatterjee, Sumantra; Karasaki, Kameko M; Fries, Lauren E; Kapoor, Ashish; Chakravarti, Aravinda
The major genetic risk factors for Hirschsprung disease (HSCR) are three common polymorphisms within cis-regulatory elements (CREs) of the receptor tyrosine kinase gene RET, which reduce its expression during enteric nervous system (ENS) development. These risk variants attenuate binding of the transcription factors RARB, GATA2, and SOX10 to their cognate CREs, reduce RET gene expression, and dysregulate other ENS and HSCR genes in the RET-EDNRB gene regulatory network (GRN). Here, we use siRNA, ChIP, and CRISPR-Cas9 deletion analyses in the SK-N-SH cell line to ask how many additional HSCR-associated risk variants reside in RET CREs that affect its gene expression. We identify 22 HSCR-associated variants in candidate RET CREs, of which seven have differential allele-specific in vitro enhancer activity, and four of these seven affect RET gene expression; of these, two enhancers are bound by the transcription factor PAX3. We also show that deleting multiple variant-containing enhancers leads to synergistic effects on RET gene expression. These, coupled with our prior results, show that common sequence variants in at least 10 RET enhancers affect HSCR risk, seven with experimental evidence of affecting RET gene expression, extending the known RET-EDNRB GRN to reveal an extensive regulatory code modulating disease risk at a single gene.
PMID: 34782358
ISSN: 1549-5469
CID: 5049002

Interferon pathway lupus risk alleles modulate risk of death from acute COVID-19

Nln, Ilona; Fernandez-Ruiz, Ruth; Wampler Muskardin, Theresa L; Paredes, Jacqueline L; Blazer, Ashira D; Tuminello, Stephanie; Attur, Mukundan; Iturrate, Eduardo; Petrilli, Christopher M; Abramson, Steven B; Chakravarti, Aravinda; Niewold, Timothy B
Type I interferon (IFN) is critical in our defense against viral infections. Increased type I IFN pathway activation is a genetic risk factor for systemic lupus erythematosus (SLE), and a number of common risk alleles contribute to the high IFN trait. We hypothesized that these common gain-of-function IFN pathway alleles may be associated with protection from mortality in acute COVID-19. We studied patients admitted with acute COVID-19 (756 European-American and 398 African-American ancestry). Ancestral backgrounds were analyzed separately, and mortality after acute COVID-19 was the primary outcome. In European-American ancestry, we found that a haplotype of interferon regulatory factor 5 (IRF5) and alleles of protein kinase cGMP-dependent 1 (PRKG1) were associated with mortality from COVID-19. Interestingly, these were much stronger risk factors in younger patients (OR=29.2 for PRKG1 in ages 45-54). Variants in the IRF7 and IRF8 genes were associated with mortality from COVID-19 in African-American subjects, and these genetic effects were more pronounced in older subjects. Combining genetic information with blood biomarker data such as C-reactive protein, troponin, and D-dimer resulted in significantly improved predictive capacity, and in both ancestral backgrounds the risk genotypes were most relevant in those with positive biomarkers (OR for death between 14 and 111 in high risk genetic/biomarker groups). This study confirms the critical role of the IFN pathway in defense against COVID-19 and viral infections, and supports the idea that some common SLE risk alleles exert protective effects in anti-viral immunity.
PMID: 34751274
ISSN: n/a
CID: 5362212

Methylome Analysis of Rare Sinonasal Malignancies: Novel Venues for Clinical Applications

Mladkova, N; Dogan, S; Yaney, A; Snuderl, M; Palmer, J D; Gamez, M E; Bonomi, M; Prevedello, D; Raval, R; Chakravarti, A; Carrau, R; Blakaj, D M
PURPOSE/OBJECTIVE(S): Olfactory neuroblastoma (ONB) and sinonasal undifferentiated carcinoma (SNUC) are rare sinonasal cancers with distinct clinical courses. Methylomic differences between these entities have not been previously explored. The aims of this study were to compare genome-wide methylomes of ONB and SNUC, and to explore candidate tissue of origin of SNUC using a comparative methylomics approach. MATERIALS/METHODS: We included a total of 87 sinonasal malignancy samples. Genome-wide methylome data were obtained with an SNP-based human microarray for 71 ONB-like samples (44 "definitive" ONB samples, 27 ONB-resembling samples) and 10 IDH2-mutatnt SNUC samples. RNA sequencing data for a further 3 ONB and 3 SNUC samples were included. Methylomes were compared between ONB and SNUC samples and to publicly available methylation data for 195 tissue samples representing an array of tumors and normal tissues using R. Strict quality control was performed.
RESULT(S): A total of 41 ONB and 9 SNUC samples passed QC and were included in the final analysis. A total of 89,016 differentially methylated probes had adjusted P-value < 0.01, the majority (90.4%) of which were hypermethylated in SNUC and located predominantly in regulatory regions, likely reflecting IDH2 mutation. The remaining 9.6% were hypomethylated in SNUC samples and were commonly located in intergenic regions. Top differentially methylated regions were GATA3, IGF2, LSP1, NKX6-2 and UNCX. NKX6-2 and UNCX were not expressed in SNUC RNAseq data. Gene set enrichment analysis (GSEA) of differentially hypermethylated CpG-island associated probes with at least 2.5-fold-decrease in SNUC RNA-seq data showed enrichment of EZH2 targets, H3K27 bound genes consistent with previous GSEA in IDH2-mutat sarcomas, in addition to WNT pathway, olfactory transduction, lipid & carbohydrate metabolism and genes pertaining to the nervous system (P < 0.0001). On Pearson's correlation-based analysis, ONB clustered with neuronal samples while SNUC clustered with medulloblastoma (MB) and malignant peripheral nerve sheath tumor (MPNST). On separate correlation, SNUC clustered with T-cell lymphoma, MPNST and MB. On examination of the top 300 differentially methylated probes, SNUC again clustered with MPNST and MB and with a neural crest cluster. On deconvolution analysis, both ONB and SNUC samples showed a predominant T-cell presence, which was significantly higher in SNUC (P < 0.001).
CONCLUSION(S): ONB and SNUC possess distinct methylomes. PRC2 complex dysregulation may represent a key driver of a genome-wide repressive phenotype in IDH2-mutant SNUC samples leading to a dedifferentiated phenotype and may be a potential avenue for targeted therapies in the future. The presence of T-cell infiltration in both tumors represents a potential avenue for exploration of therapeutic checkpoint inhibition. Genome-wide methylomics suggests SNUC may be derived from neuronal tissue and we are currently involved in further transomic approaches to independently validate this hypothesis.
Copyright
EMBASE:636626413
ISSN: 1879-355x
CID: 5077782

Magnitude of Mendelian versus complex inheritance of rare disorders

Chakravarti, Aravinda
In medical genetics, the vast majority of patients with a currently known genetic basis harbor a rare deleterious allele explaining its Mendelian inheritance. Increasingly, for these phenotypes, we recognize exceptions to Mendelian expectations from non-penetrance of clinical disease to significant inter-individual variation in clinical manifestations, likely reflecting the actions of additional modifier genes. Despite recent progress, we still remain ignorant about the molecular basis for many rare disorders presumed to be Mendelian. The molecular evidence increasingly suggests a role for multiple genes in some of these cases, but for how many? In this article, I discuss why equating a phenotype as Mendelian or complex may be short-sighted or even erroneous. As we learn more about the functions of the human genome with its genes in networks, we should view the phenotype of an individual patient as arising from his or her total genomic deleterious burden in a set of functionally inter-related genes affecting that phenotype. This can sometimes arise from deleterious allele(s) at a single gene (Mendelian inheritance) creating a specific biochemical deficiency (or excess) but could just as frequently arise from the cumulative effects of multiple disease alleles (complex inheritance) leading to the same biochemical deficiency (or excess).
PMID: 34418293
ISSN: 1552-4833
CID: 5006452

Genome wide pleiotropic analysis to identify novel variants and improve genetic risk score construction [Meeting Abstract]

Zhu, X; Zhu, L; Wang, H; Cooper, R S; Chakravarti, A
Systolic and diastolic blood pressure (S/
EMBASE:636152913
ISSN: 1098-2272
CID: 5024952

Sequence-based correction of barcode bias in massively parallel reporter assays

Lee, Dongwon; Kapoor, Ashish; Lee, Changhee; Mudgett, Michael; Beer, Michael A; Chakravarti, Aravinda
Massively parallel reporter assays (MPRAs) are a high-throughput method for evaluating in vitro activities of thousands of candidate cis-regulatory elements (CREs). In these assays, candidate sequences are cloned upstream or downstream from a reporter gene tagged by unique DNA sequences. However, tag sequences may themselves affect reporter gene expression and lead to major potential biases in the measured cis-regulatory activity. Here, we present a sequence-based method for correcting tag-sequence-specific effects and show that our method can significantly reduce this source of variation and improve the identification of functional regulatory variants by MPRAs. We also show that our model captures sequence features associated with post-transcriptional regulation of mRNA. Thus, this new method helps not only to improve detection of regulatory signals in MPRA experiments but also to design better MPRA protocols.
PMID: 34285053
ISSN: 1549-5469
CID: 4979642

Interferon pathway lupus risk alleles modulate risk of death from acute covid-19 [Meeting Abstract]

Nln, I; Ruiz, R F; Muskardin, T W; Tuminello, S; Attur, M; Itturate, E; Petrilli, C; Abramson, S B; Chakravarti, A; Niewold, T
Background/Purpose: Type I interferon (IFN) is critical in our defense against viral infections. Increased type I IFN pathway activation is a genetic risk factor for systemic lupus erythematosus (SLE), and a number of common alleles contribute to the genetic high IFN trait. In this study, we examine whether these common gain-of-function alleles in the type I IFN pathway are associated with protection from mortality in acute COVID-19.
Method(s): We studied IFN pathway SLE risk genes in patients with acute COVID-19 admitted to NYU Langone hospitals (751 European-American and 398 African-American ancestry). The samples were genotyped using low depth sequencing and imputation, and we analyzed data from the following SNPs: IRF5 (rs2004640, rs3807306, rs10488631, rs2280714), IRF7/PHRF (rs1131665, rs4963128), IRF8 (rs17445836, rs12444486), and PRKG1 (rs7897633). Ancestral backgrounds were analyzed separately, and mortality after acute COVID-19 was the primary outcome.
Result(s): We observed specific IRF5 haplotypes that are protective against SLE risk were associated with increased risk of mortality in acute COVID-19 patients in European-American ancestry (OR=3.74, p=0.015). Alleles of PRKG1 were also associated with mortality from COVID-19 in the European-American ancestry cohort (OR=1.80, p=0.0057), and this risk factor was particularly strong in younger patients (OR=29.2, p=0.01 in ages 45-54). IRF8 genotype at rs1244486 was associated with protection from mortality in COVID-19 in African-American subjects aged 65 and older (OR=0.34, p=0.04).
Conclusion(s): We find that a number of type I IFN pathway genes associated with risk of SLE also modulate risk of death during acute COVID-19. Similar to their associations with SLE, these alleles are variably associated with COVID-19 mortality across ancestral backgrounds, suggesting ancestral differences in the genetic regulation of the IFN pathway. These data confirm the critical role of the IFN pathway in our defense against viral infections, and support the idea that some common SLE risk alleles exert protective effects in anti-viral immunity
PMCID:
EMBASE:637275920
ISSN: 2326-5205
CID: 5164662