scispace - formally typeset
Search or ask a question
Author

Karna Robinson

Bio: Karna Robinson is an academic researcher from Leidos. The author has contributed to research in topics: Regulation of gene expression & Genome-wide association study. The author has an hindex of 13, co-authored 14 publications receiving 10924 citations. Previous affiliations of Karna Robinson include Science Applications International Corporation.

Papers
More filters
Journal ArticleDOI
John T. Lonsdale, Jeffrey Thomas, Mike Salvatore, Rebecca Phillips, Edmund Lo, Saboor Shad, Richard Hasz, Gary Walters, Fernando U. Garcia1, Nancy Young2, Barbara A. Foster3, Mike Moser3, Ellen Karasik3, Bryan Gillard3, Kimberley Ramsey3, Susan L. Sullivan, Jason Bridge, Harold Magazine, John Syron, Johnelle Fleming, Laura A. Siminoff4, Heather M. Traino4, Maghboeba Mosavel4, Laura Barker4, Scott D. Jewell5, Daniel C. Rohrer5, Dan Maxim5, Dana Filkins5, Philip Harbach5, Eddie Cortadillo5, Bree Berghuis5, Lisa Turner5, Eric Hudson5, Kristin Feenstra5, Leslie H. Sobin6, James A. Robb6, Phillip Branton, Greg E. Korzeniewski6, Charles Shive6, David Tabor6, Liqun Qi6, Kevin Groch6, Sreenath Nampally6, Steve Buia6, Angela Zimmerman6, Anna M. Smith6, Robin Burges6, Karna Robinson6, Kim Valentino6, Deborah Bradbury6, Mark Cosentino6, Norma Diaz-Mayoral6, Mary Kennedy6, Theresa Engel6, Penelope Williams6, Kenyon Erickson, Kristin G. Ardlie7, Wendy Winckler7, Gad Getz8, Gad Getz7, David S. DeLuca7, MacArthur Daniel MacArthur7, MacArthur Daniel MacArthur8, Manolis Kellis7, Alexander Thomson7, Taylor Young7, Ellen Gelfand7, Molly Donovan7, Yan Meng7, George B. Grant7, Deborah C. Mash9, Yvonne Marcus9, Margaret J. Basile9, Jun Liu8, Jun Zhu10, Zhidong Tu10, Nancy J. Cox11, Dan L. Nicolae11, Eric R. Gamazon11, Hae Kyung Im11, Anuar Konkashbaev11, Jonathan K. Pritchard11, Jonathan K. Pritchard12, Matthew Stevens11, Timothée Flutre11, Xiaoquan Wen11, Emmanouil T. Dermitzakis13, Tuuli Lappalainen13, Roderic Guigó, Jean Monlong, Michael Sammeth, Daphne Koller14, Alexis Battle14, Sara Mostafavi14, Mark I. McCarthy15, Manual Rivas15, Julian Maller15, Ivan Rusyn16, Andrew B. Nobel16, Fred A. Wright16, Andrey A. Shabalin16, Mike Feolo17, Nataliya Sharopova17, Anne Sturcke17, Justin Paschal17, James M. Anderson17, Elizabeth L. Wilder17, Leslie Derr17, Eric D. Green17, Jeffery P. Struewing17, Gary F. Temple17, Simona Volpi17, Joy T. Boyer17, Elizabeth J. Thomson17, Mark S. Guyer17, Cathy Ng17, Assya Abdallah17, Deborah Colantuoni17, Thomas R. Insel17, Susan E. Koester17, Roger Little17, Patrick Bender17, Thomas Lehner17, Yin Yao17, Carolyn C. Compton17, Jimmie B. Vaught17, Sherilyn Sawyer17, Nicole C. Lockhart17, Joanne P. Demchok17, Helen F. Moore17 
TL;DR: The Genotype-Tissue Expression (GTEx) project is described, which will establish a resource database and associated tissue bank for the scientific community to study the relationship between genetic variation and gene expression in human tissues.
Abstract: Genome-wide association studies have identified thousands of loci for common diseases, but, for the majority of these, the mechanisms underlying disease susceptibility remain unknown. Most associated variants are not correlated with protein-coding changes, suggesting that polymorphisms in regulatory regions probably contribute to many disease phenotypes. Here we describe the Genotype-Tissue Expression (GTEx) project, which will establish a resource database and associated tissue bank for the scientific community to study the relationship between genetic variation and gene expression in human tissues.

6,545 citations

Journal ArticleDOI
Kristin G. Ardlie, David S. DeLuca, Ayellet V. Segrè, Timothy J. Sullivan, Taylor Young, Ellen Gelfand, Casandra A. Trowbridge, Julian Maller, Taru Tukiainen, Monkol Lek, Lucas D. Ward, Pouya Kheradpour, Benjamin Iriarte, Yan Meng, Cameron D. Palmer, Tõnu Esko, Wendy Winckler, Joel N. Hirschhorn, Manolis Kellis, Daniel G. MacArthur, Gad Getz, Andrey A. Shabalin, Gen Li, Yi-Hui Zhou, Andrew B. Nobel, Ivan Rusyn, Fred A. Wright, Tuuli Lappalainen, Pedro G. Ferreira, Halit Ongen, Manuel A. Rivas, Alexis Battle, Sara Mostafavi, Jean Monlong, Michael Sammeth, Marta Melé, Ferran Reverter, Jakob M. Goldmann, Daphne Koller, Roderic Guigó, Mark I. McCarthy, Emmanouil T. Dermitzakis, Eric R. Gamazon, Hae Kyung Im, Anuar Konkashbaev, Dan L. Nicolae, Nancy J. Cox, Timothée Flutre, Xiaoquan Wen, Matthew Stephens, Jonathan K. Pritchard, Zhidong Tu, Bin Zhang, Tao Huang, Quan Long, Luan Lin, Jialiang Yang, Jun Zhu, Jun Liu, Amanda Brown, Bernadette Mestichelli, Denee Tidwell, Edmund Lo, Mike Salvatore, Saboor Shad, Jeffrey A. Thomas, John T. Lonsdale, Michael T. Moser, Bryan Gillard, Ellen Karasik, Kimberly Ramsey, Christopher Choi, Barbara A. Foster, John Syron, Johnell Fleming, Harold Magazine, Rick Hasz, Gary Walters, Jason Bridge, Mark Miklos, Susan L. Sullivan, Laura Barker, Heather M. Traino, Maghboeba Mosavel, Laura A. Siminoff, Dana R. Valley, Daniel C. Rohrer, Scott D. Jewell, Philip A. Branton, Leslie H. Sobin, Mary Barcus, Liqun Qi, Jeffrey McLean, Pushpa Hariharan, Ki Sung Um, Shenpei Wu, David Tabor, Charles Shive, Anna M. Smith, Stephen A. Buia, Anita H. Undale, Karna Robinson, Nancy Roche, Kimberly M. Valentino, Angela Britton, Robin Burges, Debra Bradbury, Kenneth W. Hambright, John Seleski, Greg E. Korzeniewski, Kenyon Erickson, Yvonne Marcus, Jorge Tejada, Mehran Taherian, Chunrong Lu, Margaret J. Basile, Deborah C. Mash, Simona Volpi, Jeffery P. Struewing, Gary F. Temple, Joy T. Boyer, Deborah Colantuoni, Roger Little, Susan E. Koester, Latarsha J. Carithers, Helen M. Moore, Ping Guan, Carolyn C. Compton, Sherilyn Sawyer, Joanne P. Demchok, Jimmie B. Vaught, Chana A. Rabiner, Nicole C. Lockhart 
08 May 2015-Science
TL;DR: The landscape of gene expression across tissues is described, thousands of tissue-specific and shared regulatory expression quantitative trait loci (eQTL) variants are cataloged, complex network relationships are described, and signals from genome-wide association studies explained by eQTLs are identified.
Abstract: Understanding the functional consequences of genetic variation, and how it affects complex human disease and quantitative traits, remains a critical challenge for biomedicine. We present an analysi...

4,418 citations

Journal ArticleDOI
TL;DR: How a successful infrastructure for biospecimen procurement was developed and implemented by multiple research partners to support the prospective collection, annotation, and distribution of blood, tissues, and cell lines for the GTEx project is described.
Abstract: The Genotype-Tissue Expression (GTEx) project, sponsored by the NIH Common Fund, was established to study the correlation between human genetic variation and tissue-specific gene expression in non-diseased individuals. A significant challenge was the collection of high-quality biospecimens for extensive genomic analyses. Here we describe how a successful infrastructure for biospecimen procurement was developed and implemented by multiple research partners to support the prospective collection, annotation, and distribution of blood, tissues, and cell lines for the GTEx project. Other research projects can follow this model and form beneficial partnerships with rapid autopsy and organ procurement organizations to collect high quality biospecimens and associated clinical data for genomic studies. Biospecimens, clinical and genomic data, and Standard Operating Procedures guiding biospecimen collection for the GTEx project are available to the research community.

669 citations

Journal ArticleDOI
TL;DR: A mathematical expression is derived to compute PrediXcan results using summary data, and the effects of gene expression variation on human phenotypes in 44 GTEx tissues and >100 phenotypes are investigated.
Abstract: Scalable, integrative methods to understand mechanisms that link genetic variants with phenotypes are needed. Here we derive a mathematical expression to compute PrediXcan (a gene mapping approach) results using summary data (S-PrediXcan) and show its accuracy and general robustness to misspecified reference sets. We apply this framework to 44 GTEx tissues and 100+ phenotypes from GWAS and meta-analysis studies, creating a growing public catalog of associations that seeks to capture the effects of gene expression variation on human phenotypes. Replication in an independent cohort is shown. Most of the associations are tissue specific, suggesting context specificity of the trait etiology. Colocalized significant associations in unexpected tissues underscore the need for an agnostic scanning of multiple contexts to improve our ability to detect causal regulatory mechanisms. Monogenic disease genes are enriched among significant associations for related traits, suggesting that smaller alterations of these genes may cause a spectrum of milder phenotypes.

657 citations


Cited by
More filters
28 Jul 2005
TL;DR: PfPMP1)与感染红细胞、树突状组胞以及胎盘的单个或多个受体作用,在黏附及免疫逃避中起关键的作�ly.
Abstract: 抗原变异可使得多种致病微生物易于逃避宿主免疫应答。表达在感染红细胞表面的恶性疟原虫红细胞表面蛋白1(PfPMP1)与感染红细胞、内皮细胞、树突状细胞以及胎盘的单个或多个受体作用,在黏附及免疫逃避中起关键的作用。每个单倍体基因组var基因家族编码约60种成员,通过启动转录不同的var基因变异体为抗原变异提供了分子基础。

18,940 citations

Journal ArticleDOI
Monkol Lek, Konrad J. Karczewski1, Konrad J. Karczewski2, Eric Vallabh Minikel2, Eric Vallabh Minikel1, Kaitlin E. Samocha, Eric Banks2, Timothy Fennell2, Anne H. O’Donnell-Luria2, Anne H. O’Donnell-Luria1, Anne H. O’Donnell-Luria3, James S. Ware, Andrew J. Hill4, Andrew J. Hill2, Andrew J. Hill1, Beryl B. Cummings1, Beryl B. Cummings2, Taru Tukiainen1, Taru Tukiainen2, Daniel P. Birnbaum2, Jack A. Kosmicki, Laramie E. Duncan1, Laramie E. Duncan2, Karol Estrada1, Karol Estrada2, Fengmei Zhao2, Fengmei Zhao1, James Zou2, Emma Pierce-Hoffman2, Emma Pierce-Hoffman1, Joanne Berghout5, David Neil Cooper6, Nicole A. Deflaux7, Mark A. DePristo2, Ron Do, Jason Flannick2, Jason Flannick1, Menachem Fromer, Laura D. Gauthier2, Jackie Goldstein2, Jackie Goldstein1, Namrata Gupta2, Daniel P. Howrigan1, Daniel P. Howrigan2, Adam Kiezun2, Mitja I. Kurki2, Mitja I. Kurki1, Ami Levy Moonshine2, Pradeep Natarajan, Lorena Orozco, Gina M. Peloso1, Gina M. Peloso2, Ryan Poplin2, Manuel A. Rivas2, Valentin Ruano-Rubio2, Samuel A. Rose2, Douglas M. Ruderfer8, Khalid Shakir2, Peter D. Stenson6, Christine Stevens2, Brett Thomas1, Brett Thomas2, Grace Tiao2, María Teresa Tusié-Luna, Ben Weisburd2, Hong-Hee Won9, Dongmei Yu, David Altshuler2, David Altshuler10, Diego Ardissino, Michael Boehnke11, John Danesh12, Stacey Donnelly2, Roberto Elosua, Jose C. Florez2, Jose C. Florez1, Stacey Gabriel2, Gad Getz1, Gad Getz2, Stephen J. Glatt13, Christina M. Hultman14, Sekar Kathiresan, Markku Laakso15, Steven A. McCarroll1, Steven A. McCarroll2, Mark I. McCarthy16, Mark I. McCarthy17, Dermot P.B. McGovern18, Ruth McPherson19, Benjamin M. Neale1, Benjamin M. Neale2, Aarno Palotie, Shaun Purcell8, Danish Saleheen20, Jeremiah M. Scharf, Pamela Sklar, Patrick F. Sullivan14, Patrick F. Sullivan21, Jaakko Tuomilehto22, Ming T. Tsuang23, Hugh Watkins17, Hugh Watkins16, James G. Wilson24, Mark J. Daly2, Mark J. Daly1, Daniel G. MacArthur2, Daniel G. MacArthur1 
18 Aug 2016-Nature
TL;DR: The aggregation and analysis of high-quality exome (protein-coding region) DNA sequence data for 60,706 individuals of diverse ancestries generated as part of the Exome Aggregation Consortium (ExAC) provides direct evidence for the presence of widespread mutational recurrence.
Abstract: Large-scale reference data sets of human genetic variation are critical for the medical and functional interpretation of DNA sequence changes. Here we describe the aggregation and analysis of high-quality exome (protein-coding region) DNA sequence data for 60,706 individuals of diverse ancestries generated as part of the Exome Aggregation Consortium (ExAC). This catalogue of human genetic diversity contains an average of one variant every eight bases of the exome, and provides direct evidence for the presence of widespread mutational recurrence. We have used this catalogue to calculate objective metrics of pathogenicity for sequence variants, and to identify genes subject to strong selection against various classes of mutation; identifying 3,230 genes with near-complete depletion of predicted protein-truncating variants, with 72% of these genes having no currently established human disease phenotype. Finally, we demonstrate that these data can be used for the efficient filtering of candidate disease-causing variants, and for the discovery of human 'knockout' variants in protein-coding genes.

8,758 citations

Journal ArticleDOI
TL;DR: A significant update to one of the tools in this domain called Enrichr, a comprehensive resource for curated gene sets and a search engine that accumulates biological knowledge for further biological discoveries is presented.
Abstract: Enrichment analysis is a popular method for analyzing gene sets generated by genome-wide experiments. Here we present a significant update to one of the tools in this domain called Enrichr. Enrichr currently contains a large collection of diverse gene set libraries available for analysis and download. In total, Enrichr currently contains 180 184 annotated gene sets from 102 gene set libraries. New features have been added to Enrichr including the ability to submit fuzzy sets, upload BED files, improved application programming interface and visualization of the results as clustergrams. Overall, Enrichr is a comprehensive resource for curated gene sets and a search engine that accumulates biological knowledge for further biological discoveries. Enrichr is freely available at: http://amp.pharm.mssm.edu/Enrichr.

6,201 citations

Journal ArticleDOI
Zefang Tang1, Chenwei Li1, Boxi Kang1, Ge Gao1, Cheng Li1, Zemin Zhang 
TL;DR: GEPIA (Gene Expression Profiling Interactive Analysis) fills in the gap between cancer genomics big data and the delivery of integrated information to end users, thus helping unleash the value of the current data resources.
Abstract: Tremendous amount of RNA sequencing data have been produced by large consortium projects such as TCGA and GTEx, creating new opportunities for data mining and deeper understanding of gene functions. While certain existing web servers are valuable and widely used, many expression analysis functions needed by experimental biologists are still not adequately addressed by these tools. We introduce GEPIA (Gene Expression Profiling Interactive Analysis), a web-based tool to deliver fast and customizable functionalities based on TCGA and GTEx data. GEPIA provides key interactive and customizable functions including differential expression analysis, profiling plotting, correlation analysis, patient survival analysis, similar gene detection and dimensionality reduction analysis. The comprehensive expression analyses with simple clicking through GEPIA greatly facilitate data mining in wide research areas, scientific discussion and the therapeutic discovery process. GEPIA fills in the gap between cancer genomics big data and the delivery of integrated information to end users, thus helping unleash the value of the current data resources. GEPIA is available at http://gepia.cancer-pku.cn/.

5,980 citations

Journal ArticleDOI
TL;DR: This work presents a method named HISAT2 (hierarchical indexing for spliced alignment of transcripts 2) that can align both DNA and RNA sequences using a graph Ferragina Manzini index, and uses it to represent and search an expanded model of the human reference genome.
Abstract: The human reference genome represents only a small number of individuals, which limits its usefulness for genotyping. We present a method named HISAT2 (hierarchical indexing for spliced alignment of transcripts 2) that can align both DNA and RNA sequences using a graph Ferragina Manzini index. We use HISAT2 to represent and search an expanded model of the human reference genome in which over 14.5 million genomic variants in combination with haplotypes are incorporated into the data structure used for searching and alignment. We benchmark HISAT2 using simulated and real datasets to demonstrate that our strategy of representing a population of genomes, together with a fast, memory-efficient search algorithm, provides more detailed and accurate variant analyses than other methods. We apply HISAT2 for HLA typing and DNA fingerprinting; both applications form part of the HISAT-genotype software that enables analysis of haplotype-resolved genes or genomic regions. HISAT-genotype outperforms other computational methods and matches or exceeds the performance of laboratory-based assays. A graph-based genome indexing scheme enables variant-aware alignment of sequences with very low memory requirements.

4,855 citations