Network


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

Hotspot


Dive into the research topics where Thanneer M. Perumal is active.

Publication


Featured researches published by Thanneer M. Perumal.


Nature Neuroscience | 2016

Gene expression elucidates functional impact of polygenic risk for schizophrenia.

Menachem Fromer; Panos Roussos; Solveig K. Sieberts; Jessica S. Johnson; David H. Kavanagh; Thanneer M. Perumal; Douglas M. Ruderfer; Edwin C. Oh; Aaron Topol; Hardik Shah; Lambertus Klei; Robin Kramer; Dalila Pinto; Zeynep H. Gümüş; A. Ercument Cicek; Kristen Dang; Andrew Browne; Cong Lu; Lu Xie; Ben Readhead; Eli A. Stahl; Jianqiu Xiao; Mahsa Parvizi; Tymor Hamamsy; John F. Fullard; Ying-Chih Wang; Milind Mahajan; Jonathan Derry; Joel T. Dudley; Scott E. Hemby

Over 100 genetic loci harbor schizophrenia-associated variants, yet how these variants confer liability is uncertain. The CommonMind Consortium sequenced RNA from dorsolateral prefrontal cortex of people with schizophrenia (N = 258) and control subjects (N = 279), creating a resource of gene expression and its genetic regulation. Using this resource, ∼20% of schizophrenia loci have variants that could contribute to altered gene expression and liability. In five loci, only a single gene was involved: FURIN, TSNARE1, CNTN4, CLCN3 or SNAP91. Altering expression of FURIN, TSNARE1 or CNTN4 changed neurodevelopment in zebrafish; knockdown of FURIN in human neural progenitor cells yielded abnormal migration. Of 693 genes showing significant case-versus-control differential expression, their fold changes were ≤ 1.33, and an independent cohort yielded similar results. Gene co-expression implicates a network relevant for schizophrenia. Our findings show that schizophrenia is polygenic and highlight the utility of this resource for mechanistic interpretations of genetic liability for brain diseases.


American Journal of Human Genetics | 2017

Large-Scale Identification of Common Trait and Disease Variants Affecting Gene Expression

Mads E. Hauberg; Wen Zhang; Claudia Giambartolomei; Oscar Franzén; David L. Morris; Timothy J. Vyse; Arno Ruusalepp; Menachem Fromer; Solveig K. Sieberts; Jessica S. Johnson; Douglas M. Ruderfer; Hardik Shah; Lambertus Klei; Kristen Dang; Thanneer M. Perumal; Benjamin A. Logsdon; Milind Mahajan; Lara M. Mangravite; Laurent Essioux; Hiroyoshi Toyoshiba; Raquel E. Gur; Chang-Gyu Hahn; David A. Lewis; Vahram Haroutunian; Mette A. Peters; Barbara K. Lipska; Joseph D. Buxbaum; Keisuke Hirai; Enrico Domenici; Bernie Devlin

Genome-wide association studies (GWASs) have identified a multitude of genetic loci involved with traits and diseases. However, it is often unclear which genes are affected in such loci and whether the associated genetic variants lead to increased or decreased gene function. To mitigate this, we integrated associations of common genetic variants in 57 GWASs with 24 studies of expression quantitative trait loci (eQTLs) from a broad range of tissues by using a Mendelian randomization approach. We discovered a total of 3,484 instances of gene-trait-associated changes in expression at a false-discovery rate < 0.05. These genes were often not closest to the genetic variant and were primarily identified in eQTLs derived from pathophysiologically relevant tissues. For instance, genes with expression changes associated with lipid traits were mostly identified in the liver, and those associated with cardiovascular disease were identified in arterial tissue. The affected genes additionally point to biological processes implicated in the interrogated traits, such as the interleukin-27 pathway in rheumatoid arthritis. Further, comparing trait-associated gene expression changes across traits suggests that pleiotropy is a widespread phenomenon and points to specific instances of both agonistic and antagonistic pleiotropy. For instance, expression of SNX19 and ABCB9 is positively correlated with both the risk of schizophrenia and educational attainment. To facilitate interpretation, we provide this lexicon of how common trait-associated genetic variants alter gene expression in various tissues as the online database GWAS2Genes.


Scientific Data | 2017

Molecular, phenotypic, and sample-associated data to describe pluripotent stem cell lines and derivatives.

Kenneth Daily; Shannan J. Ho Sui; Lynn M. Schriml; Phillip Dexheimer; Nathan Salomonis; Robin Schroll; Stacy Bush; Mehdi Keddache; Christopher N. Mayhew; Samad Lotia; Thanneer M. Perumal; Kristen Dang; Lorena Pantano; Alexander R. Pico; Elke Grassman; Diana Nordling; Winston Hide; Antonis K. Hatzopoulos; Punam Malik; Jose A. Cancelas; Carolyn Lutzko; Bruce J. Aronow; Larsson Omberg

The use of induced pluripotent stem cells (iPSC) derived from independent patients and sources holds considerable promise to improve the understanding of development and disease. However, optimized use of iPSC depends on our ability to develop methods to efficiently qualify cell lines and protocols, monitor genetic stability, and evaluate self-renewal and differentiation potential. To accomplish these goals, 57 stem cell lines from 10 laboratories were differentiated to 7 different states, resulting in 248 analyzed samples. Cell lines were differentiated and characterized at a central laboratory using standardized cell culture methodologies, protocols, and metadata descriptors. Stem cell and derived differentiated lines were characterized using RNA-seq, miRNA-seq, copy number arrays, DNA methylation arrays, flow cytometry, and molecular histology. All materials, including raw data, metadata, analysis and processing code, and methodological and provenance documentation are publicly available for re-use and interactive exploration at https://www.synapse.org/pcbc. The goal is to provide data that can improve our ability to robustly and reproducibly use human pluripotent stem cells to understand development and disease.


Nature Communications | 2018

A community approach to mortality prediction in sepsis via gene expression analysis

Timothy E. Sweeney; Thanneer M. Perumal; Ricardo Henao; Marshall Nichols; Judith A. Howrylak; Augustine M. K. Choi; Jesus F. Bermejo-Martin; Raquel Almansa; Eduardo Tamayo; Emma E. Davenport; Katie L Burnham; Charles J. Hinds; Julian C. Knight; Christopher W. Woods; Stephen F. Kingsmore; Geoffrey S. Ginsburg; Hector R. Wong; Grant P. Parnell; Benjamin Tang; Lyle L. Moldawer; Frederick E. Moore; Larsson Omberg; Purvesh Khatri; Ephraim L. Tsalik; Lara M. Mangravite; Raymond J. Langley

Improved risk stratification and prognosis prediction in sepsis is a critical unmet need. Clinical severity scores and available assays such as blood lactate reflect global illness severity with suboptimal performance, and do not specifically reveal the underlying dysregulation of sepsis. Here, we present prognostic models for 30-day mortality generated independently by three scientific groups by using 12 discovery cohorts containing transcriptomic data collected from primarily community-onset sepsis patients. Predictive performance is validated in five cohorts of community-onset sepsis patients in which the models show summary AUROCs ranging from 0.765–0.89. Similar performance is observed in four cohorts of hospital-acquired sepsis. Combining the new gene-expression-based prognostic models with prior clinical severity scores leads to significant improvement in prediction of 30-day mortality as measured via AUROC and net reclassification improvement index These models provide an opportunity to develop molecular bedside tests that may improve risk stratification and mortality prediction in patients with sepsis.Sepsis is characterized by deregulated host response to infection. Efficient therapies are still needed but a limitation for sepsis treatment is the heterogeneity in patients. Here Sweeney et al. generate prognostic models based on gene expression to improve risk stratification classification and prediction for 30-day mortality of patients.


pacific symposium on biocomputing | 2016

PERSONALIZED HYPOTHESIS TESTS FOR DETECTING MEDICATION RESPONSE IN PARKINSON DISEASE PATIENTS USING iPHONE SENSOR DATA.

Elias Chaibub Neto; Brian M. Bot; Thanneer M. Perumal; Larsson Omberg; Justin Guinney; Mike Kellen; Arno Klein; Stephen H. Friend; Andrew D. Trister

We propose hypothesis tests for detecting dopaminergic medication response in Parkinson disease patients, using longitudinal sensor data collected by smartphones. The processed data is composed of multiple features extracted from active tapping tasks performed by the participant on a daily basis, before and after medication, over several months. Each extracted feature corresponds to a time series of measurements annotated according to whether the measurement was taken before or after the patient has taken his/her medication. Even though the data is longitudinal in nature, we show that simple hypothesis tests for detecting medication response, which ignore the serial correlation structure of the data, are still statistically valid, showing type I error rates at the nominal level. We propose two distinct personalized testing approaches. In the first, we combine multiple feature-specific tests into a single union-intersection test. In the second, we construct personalized classifiers of the before/after medication labels using all the extracted features of a given participant, and test the null hypothesis that the area under the receiver operating characteristic curve of the classifier is equal to 1/2. We compare the statistical power of the personalized classifier tests and personalized union-intersection tests in a simulation study, and illustrate the performance of the proposed tests using data from mPower Parkinsons disease study, recently launched as part of Apples ResearchKit mobile platform. Our results suggest that the personalized tests, which ignore the longitudinal aspect of the data, can perform well in real data analyses, suggesting they might be used as a sound baseline approach, to which more sophisticated methods can be compared to.


bioRxiv | 2017

Co-localization of Conditional eQTL and GWAS Signatures in Schizophrenia

Amanda Dobbyn; Laura M. Huckins; James Boocock; Laura G. Sloofman; Benjamin S. Glicksberg; Claudia Giambartolomei; Gabriel E. Hoffman; Thanneer M. Perumal; Kiran Girdhar; Yan Jiang; Douglas Ruderfer; Robin Kramer; Dalila Pinto; Schahram Akbarian; Panos Roussos; Enrico Domenici; Bernie Devlin; Pamela Sklar; Eli A. Stahl; Solveig K. Sieberts

Causal genes and variants within genome-wide association study (GWAS) loci can be identified by integrating GWAS statistics with expression quantitative trait loci (eQTL) and determining which SNPs underlie both GWAS and eQTL signals. Most analyses, however, consider only the marginal eQTL signal, rather than dissecting this signal into multiple independent eQTL for each gene. Here we show that analyzing conditional eQTL signatures, which could be important under specific cellular or temporal contexts, leads to improved fine mapping of GWAS associations. Using genotypes and gene expression levels from post-mortem human brain samples (N=467) reported by the CommonMind Consortium (CMC), we find that conditional eQTL are widespread; 63% of genes with primary eQTL also have conditional eQTL. In addition, genomic features associated with conditional eQTL are consistent with context specific (i.e. tissue, cell type, or developmental time point specific) regulation of gene expression. Integrating the Psychiatric Genomics Consortium schizophrenia (SCZ) GWAS and CMC conditional eQTL data reveals forty loci with strong evidence for co-localization (posterior probability >0.8), including six loci with co-localization of conditional eQTL. Our co-localization analyses support previously reported genes and identify novel genes for schizophrenia risk, and provide specific hypotheses for their functional follow-up.


bioRxiv | 2016

Mortality prediction in sepsis via gene expression analysis: a community approach

Timothy E. Sweeney; Thanneer M. Perumal; Ricardo Henao; Marshall Nichols; Judith A. Howrylak; Augustine M. K. Choi; Jesus F. Bermejo-Martin; Raquel Almansa; Eduardo Tamayo; Emma E Davenport; Katie L Burnham; Charles J. Hinds; Julian C. Knight; Stephen F. Kingsmore; Christopher W. Woods; Geoffrey S. Ginsburg; Hector R. Wong; Grant P Parnell; Benjamin Tang; Lyle L. Moldawer; Frederick E. Moore; Larsson Omberg; Purvesh Khatri; Ephraim L. Tsalik; Lara M. Mangravite; Raymond J. Langley

Improved risk stratification and prognosis in sepsis is a critical unmet need. Clinical severity scores and available assays such as blood lactate reflect global illness severity with suboptimal performance, and do not specifically reveal the underlying dysregulation of sepsis. Here three scientific groups were invited to independently generate prognostic models for 30-day mortality using 12 discovery cohorts (N=650) containing transcriptomic data collected from primarily community-onset sepsis patients. Predictive performance was validated in 5 cohorts of community-onset sepsis patients (N=189) in which the models showed summary AUROCs ranging from 0.765-0.89. Similar performance was observed in 4 cohorts of hospital-acquired sepsis (N=282). Combining the new gene-expression-based prognostic models with prior clinical severity scores led to significant improvement in prediction of 30-day mortality (p<0.01). These models provide an opportunity to develop molecular bedside tests that may improve risk stratification and mortality prediction in patients with sepsis, improving both resource allocation and prognostic enrichment in clinical trials.


American Journal of Human Genetics | 2018

Landscape of Conditional eQTL in Dorsolateral Prefrontal Cortex and Co-localization with Schizophrenia GWAS

Amanda Dobbyn; Laura M. Huckins; James Boocock; Laura G. Sloofman; Benjamin S. Glicksberg; Claudia Giambartolomei; Gabriel E. Hoffman; Thanneer M. Perumal; Kiran Girdhar; Yan Jiang; Towfique Raj; Douglas Ruderfer; Robin Kramer; Dalila Pinto; Pamela Sklar; Joseph D. Buxbaum; Bernie Devlin; David A. Lewis; Raquel E. Gur; Chang-Gyu Hahn; Keisuke Hirai; Hiroyoshi Toyoshiba; Enrico Domenici; Laurent Essioux; Lara M. Mangravite; Mette A. Peters; Thomas Lehner; Barbara K. Lipska; A. Ercument Cicek; Cong Lu

Causal genes and variants within genome-wide association study (GWAS) loci can be identified by integrating GWAS statistics with expression quantitative trait loci (eQTL) and determining which variants underlie both GWAS and eQTL signals. Most analyses, however, consider only the marginal eQTL signal, rather than dissect this signal into multiple conditionally independent signals for each gene. Here we show that analyzing conditional eQTL signatures, which could be important under specific cellular or temporal contexts, leads to improved fine mapping of GWAS associations. Using genotypes and gene expression levels from post-mortem human brain samples (n = 467) reported by the CommonMind Consortium (CMC), we find that conditional eQTL are widespread; 63% of genes with primary eQTL also have conditional eQTL. In addition, genomic features associated with conditional eQTL are consistent with context-specific (e.g., tissue-, cell type-, or developmental time point-specific) regulation of gene expression. Integrating the 2014 Psychiatric Genomics Consortium schizophrenia (SCZ) GWAS and CMC primary and conditional eQTL data reveals 40 loci with strong evidence for co-localization (posterior probability > 0.8), including six loci with co-localization of conditional eQTL. Our co-localization analyses support previously reported genes, identify novel genes associated with schizophrenia risk, and provide specific hypotheses for their functional follow-up.


Alzheimers & Dementia | 2018

SYSTEMS BIOLOGY RANKING OF CANDIDATE ALZHEIMER’S DRIVER GENES IDENTIFIES NEW GENETIC DRIVERS OF ALZHEIMER’S DISEASE ETIOLOGY

Benjamin A. Logsdon; Thanneer M. Perumal; Kenneth Daily; Solveig K. Sieberts; Larsson Omberg; Lara M. Mangravite; Christoph Preuss; Gregory W. Carter

Background: The standard approaches to understand gene expression and regulation in the brain include identification of differentially and co-expressed genes. To this end, the Accelerating Medicines Partnership Alzheimer’s Disease (AMP-AD) consortium has produced multiple, large RNA-seq datasets from several postmortem brain regions. Separately, the ENCODE project has produced DNAse Hypersensitivity (DHS) samples for various brain regions. We have integrated these large datasets into transcriptional regulatory networks (TRN), providing a directional and mechanistic list of putative transcription factors for nearly all expressed genes in the brain. Methods: We reprocessed all ENCODE brain DHS samples at scale, generating footprints—signatures of occupancy by DNA binding proteins—using the Wellington and HINT algorithms. We assembled motifs from JASPAR2016, HOCOMOCO, UniPROBE, and SwissRegulon, removing redundant motifs with Tomtom and intersecting our footprints with all possible overlapping motifs. This resulted in a total of 1,530motifs mapping to 1,515 different transcription factors.We developed and utilized Transcriptional Regulatory Network Analysis (TReNA), available as an R Bioconductor package. Gene regulatory regions considered in our model were obtained through Genehancer, thus enabling the inclusion of all known enhancer regions. TReNA utilizes an ensemble of machine learning techniques, including lassopv, square root lasso (flare) and randomForest to prioritize transcription factors based on the expression levels in RNA-seq for each target gene. The scores from the aforementioned techniques are scaled and normalized into a composite score, thereby ranking all associated transcription factors for each target gene. Results:We have identified transcriptional regulators for AD genes identified through GWAS. We have also identified putative targets for the AD-associated transcription factor MEF2C. We have identified multiple microglia-enriched transcription factors that regulate many differentially and co-expressed genes in AD, in particular, the AD-associated transcription factor SPI1. Conclusions:These resulting models can be applied to other datasets that generate lists of differentially or co-expressed genes as well as provide testable hypotheses for non-coding variants of interest. We are actively engaged in testing several hypotheses through experimental means and have made these TRNs publically available.


Alzheimers & Dementia | 2017

CROSS STUDY INTEGRATIVE NETWORK ANALYSIS IDENTIFICATION OF AD DISEASE ETIOLOGY

Benjamin A. Logsdon; Thanneer M. Perumal; Solveig K. Sieberts; Larsson Omberg; Lara M. Mangravite

immunohistochemistry analyses using two previously reported TSPO antibodies in human post mortem brain sections. As reported previously, these antibodies stain macrophages, astrocytes and microglia in AD brains with more limited staining in non-AD cases. We used astrocyte and microglia specific markers to determine which cells were differentially stained between TREM2+ and TREM2AD cases and determine differences in cells adjacent to amyloid or Tau which stain for TSPO. Conclusions:The biomarker TSPO has been used extensively to explore immune activation in vivo in the brain. Levels are increased in AD, particularly associated with the emergence of abnormal Tau and neuronal cell loss. Our results will facilitate interpretation of future PET imaging in this group of patients using the TSPO tracers [C]-PK11195 or [C]-PBR28.

Collaboration


Dive into the Thanneer M. Perumal'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge