35 results on '"Pal, Lipika R."'
Search Results
2. CAGI, the Critical Assessment of Genome Interpretation, establishes progress and prospects for computational genetic variant interpretation methods
- Author
-
Jain, Shantanu, Bakolitsa, Constantina, Brenner, Steven E, Radivojac, Predrag, Moult, John, Repo, Susanna, Hoskins, Roger A, Andreoletti, Gaia, Barsky, Daniel, Chellapan, Ajithavalli, Chu, Hoyin, Dabbiru, Navya, Kollipara, Naveen K, Ly, Melissa, Neumann, Andrew J, Pal, Lipika R, Odell, Eric, Pandey, Gaurav, Peters-Petrulewicz, Robin C, Srinivasan, Rajgopal, Yee, Stephen F, Yeleswarapu, Sri Jyothsna, Zuhl, Maya, Adebali, Ogun, Patra, Ayoti, Beer, Michael A, Hosur, Raghavendra, Peng, Jian, Bernard, Brady M, Berry, Michael, Dong, Shengcheng, Boyle, Alan P, Adhikari, Aashish, Chen, Jingqi, Hu, Zhiqiang, Wang, Robert, Wang, Yaqiong, Miller, Maximilian, Wang, Yanran, Bromberg, Yana, Turina, Paola, Capriotti, Emidio, Han, James J, Ozturk, Kivilcim, Carter, Hannah, Babbi, Giulia, Bovo, Samuele, Di Lena, Pietro, Martelli, Pier Luigi, Savojardo, Castrense, Casadio, Rita, Cline, Melissa S, De Baets, Greet, Bonache, Sandra, Díez, Orland, Gutiérrez-Enríquez, Sara, Fernández, Alejandro, Montalban, Gemma, Ootes, Lars, Özkan, Selen, Padilla, Natàlia, Riera, Casandra, De la Cruz, Xavier, Diekhans, Mark, Huwe, Peter J, Wei, Qiong, Xu, Qifang, Dunbrack, Roland L, Gotea, Valer, Elnitski, Laura, Margolin, Gennady, Fariselli, Piero, Kulakovskiy, Ivan V, Makeev, Vsevolod J, Penzar, Dmitry D, Vorontsov, Ilya E, Favorov, Alexander V, Forman, Julia R, Hasenahuer, Marcia, Fornasari, Maria S, Parisi, Gustavo, Avsec, Ziga, Çelik, Muhammed H, Nguyen, Thi Yen Duong, Gagneur, Julien, Shi, Fang-Yuan, Edwards, Matthew D, Guo, Yuchun, Tian, Kevin, Zeng, Haoyang, Gifford, David K, Göke, Jonathan, Zaucha, Jan, Gough, Julian, Ritchie, Graham RS, Frankish, Adam, Mudge, Jonathan M, Harrow, Jennifer, Young, Erin L, and Yu, Yao
- Subjects
Biological Sciences ,Genetics ,Human Genome ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Humans ,Computational Biology ,Mutation ,Missense ,Phenotype ,Critical Assessment of Genome Interpretation Consortium ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
BackgroundThe Critical Assessment of Genome Interpretation (CAGI) aims to advance the state-of-the-art for computational prediction of genetic variant impact, particularly where relevant to disease. The five complete editions of the CAGI community experiment comprised 50 challenges, in which participants made blind predictions of phenotypes from genetic data, and these were evaluated by independent assessors.ResultsPerformance was particularly strong for clinical pathogenic variants, including some difficult-to-diagnose cases, and extends to interpretation of cancer-related variants. Missense variant interpretation methods were able to estimate biochemical effects with increasing accuracy. Assessment of methods for regulatory variants and complex trait disease risk was less definitive and indicates performance potentially suitable for auxiliary use in the clinic.ConclusionsResults show that while current methods are imperfect, they have major utility for research and clinical applications. Emerging methods and increasingly large, robust datasets for training and assessment promise further progress ahead.
- Published
- 2024
3. Metabolic dependency mapping identifies Peroxiredoxin 1 as a driver of resistance to ATM inhibition
- Author
-
Li, Haojian, Furusawa, Takashi, Cavero, Renzo, Xiao, Yunjie, Chari, Raj, Wu, Xiaolin, Sun, David, Hartmann, Oliver, Dhall, Anjali, Holewinski, Ronald, Andresson, Thorkell, Karim, Baktiar, Villamor-Payà, Marina, Gallardo, Devorah, Day, Chi-Ping, Pal, Lipika R., Nair, Nishanth Ulhas, Ruppin, Eytan, Aladjem, Mirit I., Pommier, Yves, Diefenbacher, Markus E., Lim, Jung Mi, Levine, Rodney L., Stracker, Travis H., and Weyemi, Urbain
- Published
- 2025
- Full Text
- View/download PDF
4. Assessing computational predictions of the phenotypic effect of cystathionine‐beta‐synthase variants
- Author
-
Kasak, Laura, Bakolitsa, Constantina, Hu, Zhiqiang, Yu, Changhua, Rine, Jasper, Dimster‐Denk, Dago F, Pandey, Gaurav, Baets, Greet, Bromberg, Yana, Cao, Chen, Capriotti, Emidio, Casadio, Rita, Durme, Joost, Giollo, Manuel, Karchin, Rachel, Katsonis, Panagiotis, Leonardi, Emanuela, Lichtarge, Olivier, Martelli, Pier Luigi, Masica, David, Mooney, Sean D, Olatubosun, Ayodeji, Radivojac, Predrag, Rousseau, Frederic, Pal, Lipika R, Savojardo, Castrense, Schymkowitz, Joost, Thusberg, Janita, Tosatto, Silvio CE, Vihinen, Mauno, Väliaho, Jouni, Repo, Susanna, Moult, John, Brenner, Steven E, and Friedberg, Iddo
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Networking and Information Technology R&D (NITRD) ,Aetiology ,2.1 Biological and endogenous factors ,Generic health relevance ,Good Health and Well Being ,Amino Acid Substitution ,Computational Biology ,Cystathionine ,Cystathionine beta-Synthase ,Homocysteine ,Humans ,Phenotype ,Precision Medicine ,CAGI challenge ,critical assessment ,cystathionine-beta-synthase ,machine learning ,phenotype prediction ,single amino acid substitution ,Clinical Sciences ,Genetics & Heredity ,Clinical sciences - Abstract
Accurate prediction of the impact of genomic variation on phenotype is a major goal of computational biology and an important contributor to personalized medicine. Computational predictions can lead to a better understanding of the mechanisms underlying genetic diseases, including cancer, but their adoption requires thorough and unbiased assessment. Cystathionine-beta-synthase (CBS) is an enzyme that catalyzes the first step of the transsulfuration pathway, from homocysteine to cystathionine, and in which variations are associated with human hyperhomocysteinemia and homocystinuria. We have created a computational challenge under the CAGI framework to evaluate how well different methods can predict the phenotypic effect(s) of CBS single amino acid substitutions using a blinded experimental data set. CAGI participants were asked to predict yeast growth based on the identity of the mutations. The performance of the methods was evaluated using several metrics. The CBS challenge highlighted the difficulty of predicting the phenotype of an ex vivo system in a model organism when classification models were trained on human disease data. We also discuss the variations in difficulty of prediction for known benign and deleterious variants, as well as identify methodological and experimental constraints with lessons to be learned for future challenges.
- Published
- 2019
5. Antibody interfaces revealed through structural mining
- Author
-
Yin, Yizhou, Romei, Matthew G., Sankar, Kannan, Pal, Lipika R., Hoi, Kam Hon, Yang, Yanli, Leonard, Brandon, De Leon Boenig, Gladys, Kumar, Nikit, Matsumoto, Marissa, Payandeh, Jian, Harris, Seth F., Moult, John, and Lazar, Greg A.
- Published
- 2022
- Full Text
- View/download PDF
6. Matching phenotypes to whole genomes: Lessons learned from four iterations of the personal genome project community challenges
- Author
-
Cai, Binghuang, Li, Biao, Kiga, Nikki, Thusberg, Janita, Bergquist, Timothy, Chen, Yun‐Ching, Niknafs, Noushin, Carter, Hannah, Tokheim, Collin, Beleva‐Guthrie, Violeta, Douville, Christopher, Bhattacharya, Rohit, Yeo, Hui Ting Grace, Fan, Jean, Sengupta, Sohini, Kim, Dewey, Cline, Melissa, Turner, Tychele, Diekhans, Mark, Zaucha, Jan, Pal, Lipika R, Cao, Chen, Yu, Chen‐Hsin, Yin, Yizhou, Carraro, Marco, Giollo, Manuel, Ferrari, Carlo, Leonardi, Emanuela, Tosatto, Silvio CE, Bobe, Jason, Ball, Madeleine, Hoskins, Roger A, Repo, Susanna, Church, George, Brenner, Steven E, Moult, John, Gough, Julian, Stanke, Mario, Karchin, Rachel, and Mooney, Sean D
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Human Genome ,Biotechnology ,Good Health and Well Being ,Area Under Curve ,Genetic Predisposition to Disease ,High-Throughput Nucleotide Sequencing ,Human Genome Project ,Humans ,Phenotype ,Quantitative Trait Loci ,Whole Genome Sequencing ,biomedical informatics ,community challenge ,critical assessment ,genome ,genome interpretation ,open consent ,personal genome project ,phenotype ,Clinical Sciences ,Genetics & Heredity ,Clinical sciences - Abstract
The advent of next-generation sequencing has dramatically decreased the cost for whole-genome sequencing and increased the viability for its application in research and clinical care. The Personal Genome Project (PGP) provides unrestricted access to genomes of individuals and their associated phenotypes. This resource enabled the Critical Assessment of Genome Interpretation (CAGI) to create a community challenge to assess the bioinformatics community's ability to predict traits from whole genomes. In the CAGI PGP challenge, researchers were asked to predict whether an individual had a particular trait or profile based on their whole genome. Several approaches were used to assess submissions, including ROC AUC (area under receiver operating characteristic curve), probability rankings, the number of correct predictions, and statistical significance simulations. Overall, we found that prediction of individual traits is difficult, relying on a strong knowledge of trait frequency within the general population, whereas matching genomes to trait profiles relies heavily upon a small number of common traits including ancestry, blood type, and eye color. When a rare genetic disorder is present, profiles can be matched when one or more pathogenic variants are identified. Prediction accuracy has improved substantially over the last 6 years due to improved methodology and a better understanding of features.
- Published
- 2017
7. Lessons from the CAGI‐4 Hopkins clinical panel challenge
- Author
-
Chandonia, John‐Marc, Adhikari, Aashish, Carraro, Marco, Chhibber, Aparna, Cutting, Garry R, Fu, Yao, Gasparini, Alessandra, Jones, David T, Kramer, Andreas, Kundu, Kunal, Lam, Hugo YK, Leonardi, Emanuela, Moult, John, Pal, Lipika R, Searls, David B, Shah, Sohela, Sunyaev, Shamil, Tosatto, Silvio CE, Yin, Yizhou, and Buckley, Bethany A
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Genetics ,Genetic Testing ,4.2 Evaluation of markers and technologies ,Computational Biology ,Databases ,Genetic ,Genetic Predisposition to Disease ,Humans ,Phenotype ,Sequence Analysis ,DNA ,CAGI ,genetic testing ,phenotype prediction ,variant interpretation ,Genetics & Heredity ,Clinical sciences - Abstract
The CAGI-4 Hopkins clinical panel challenge was an attempt to assess state-of-the-art methods for clinical phenotype prediction from DNA sequence. Participants were provided with exonic sequences of 83 genes for 106 patients from the Johns Hopkins DNA Diagnostic Laboratory. Five groups participated in the challenge, predicting both the probability that each patient had each of the 14 possible classes of disease, as well as one or more causal variants. In cases where the Hopkins laboratory reported a variant, at least one predictor correctly identified the disease class in 36 of the 43 patients (84%). Even in cases where the Hopkins laboratory did not find a variant, at least one predictor correctly identified the class in 39 of the 63 patients (62%). Each prediction group correctly diagnosed at least one patient that was not successfully diagnosed by any other group. We discuss the causal variant predictions by different groups and their implications for further development of methods to assess variants of unknown significance. Our results suggest that clinically relevant variants may be missed when physicians order small panels targeted on a specific phenotype. We also quantify the false-positive rate of DNA-guided analysis in the absence of prior phenotypic indication.
- Published
- 2017
8. Genome‐scale metabolic modeling reveals SARS‐CoV‐2‐induced metabolic changes and antiviral targets
- Author
-
Cheng, Kuoyuan, Martin‐Sancho, Laura, Pal, Lipika R, Pu, Yuan, Riva, Laura, Yin, Xin, Sinha, Sanju, Nair, Nishanth Ulhas, Chanda, Sumit K, and Ruppin, Eytan
- Published
- 2021
- Full Text
- View/download PDF
9. REPLY TO HU ET AL. : On the interpretation of gasdermin-B expression quantitative trait loci data
- Author
-
Pal, Lipika R., Chao, Kinlin L., Moult, John, and Herzberg, Osnat
- Published
- 2017
10. CAGI, the Critical Assessment of Genome Interpretation, establishes progress and prospects for computational genetic variant interpretation methods.
- Author
-
The Critical Assessment of Genome Interpretation Consortium, Jain, Shantanu, Bakolitsa, Constantina, Brenner, Steven E., Radivojac, Predrag, Moult, John, Repo, Susanna, Hoskins, Roger A., Andreoletti, Gaia, Barsky, Daniel, Chellapan, Ajithavalli, Chu, Hoyin, Dabbiru, Navya, Kollipara, Naveen K., Ly, Melissa, Neumann, Andrew J., Pal, Lipika R., Odell, Eric, Pandey, Gaurav, and Peters-Petrulewicz, Robin C.
- Published
- 2024
- Full Text
- View/download PDF
11. Abstract 3118: Predicting response to PARP inhibitors in pediatric cancer via synthetic lethal networks
- Author
-
Nagy, Matthew, primary, Schischlik, Fiorella, additional, Wang, Kun, additional, Nair, Nishanth Ulhas, additional, Gertz, E. Michael, additional, Pal, Lipika R., additional, Jaeger, Natalie, additional, Previti, Christopher, additional, ElHarouni, Dina, additional, Pfister, Stefan M., additional, and Ruppin, Eytan, additional
- Published
- 2023
- Full Text
- View/download PDF
12. Cross-species identification of cancer resistance–associated genes that may mediate human cancer risk
- Author
-
Nair, Nishanth Ulhas, primary, Cheng, Kuoyuan, additional, Naddaf, Lamis, additional, Sharon, Elad, additional, Pal, Lipika R., additional, Rajagopal, Padma S., additional, Unterman, Irene, additional, Aldape, Kenneth, additional, Hannenhalli, Sridhar, additional, Day, Chi-Ping, additional, Tabach, Yuval, additional, and Ruppin, Eytan, additional
- Published
- 2022
- Full Text
- View/download PDF
13. Abstract 3583: Identifying and testing cancer-derived synthetic-lethal anti-SARS-CoV-2 targets
- Author
-
Pal, Lipika R., primary, Cheng, Kuoyuan, additional, Nair, Nishanth Ulhas, additional, Martin-Sancho, Laura, additional, Sinha, Sanju, additional, Pu, Yuan, additional, Riva, Laura, additional, Yin, Xin, additional, Schischlik, Fiorella, additional, Lee, Joo Sang, additional, Chanda, Sumit, additional, and Ruppin, Eytan, additional
- Published
- 2022
- Full Text
- View/download PDF
14. Synthetic lethality-based prediction of anti-SARS-CoV-2 targets
- Author
-
Pal, Lipika R., primary, Cheng, Kuoyuan, additional, Nair, Nishanth Ulhas, additional, Martin-Sancho, Laura, additional, Sinha, Sanju, additional, Pu, Yuan, additional, Riva, Laura, additional, Yin, Xin, additional, Schischlik, Fiorella, additional, Lee, Joo Sang, additional, Chanda, Sumit K., additional, and Ruppin, Eytan, additional
- Published
- 2022
- Full Text
- View/download PDF
15. Working toward precision medicine: Predicting phenotypes from exomes in the Critical Assessment of Genome Interpretation (CAGI) challenges
- Author
-
Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Daneshjou, Roxana, Wang, Yanran, Bromberg, Yana, Bovo, Samuele, Martelli, Pier L, Babbi, Giulia, Di Lena, Pietro, Casadio, Rita, Edwards, Matthew D, Gifford, David K, Jones, David T, Sundaram, Laksshman, Bhat, Rajendra, Li, Xiaolin, Pal, Lipika R., Kundu, Kunal, Yin, Yizhou, Moult, John, Jiang, Yuxiang, Pejaver, Vikas, Pagel, Kymberleigh A., Li, Biao, Mooney, Sean D., Radivojac, Predrag, Shah, Sohela, Carraro, Marco, Gasparini, Alessandra, Leonardi, Emanuela, Giollo, Manuel, Ferrari, Carlo, Tosatto, Silvio C E, Bachar, Eran, Azaria, Johnathan R., Ofran, Yanay, Unger, Ron, Niroula, Abhishek, Vihinen, Mauno, Chang, Billy, Wang, Maggie H, Franke, Andre, Petersen, Britt-Sabina, Pirooznia, Mehdi, Zandi, Peter, McCombie, Richard, Potash, James B, Altman, Russ, Klein, Teri E., Hoskins, Roger, Repo, Susanna, Brenner, Steve E, Morgan, Alexander A, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Daneshjou, Roxana, Wang, Yanran, Bromberg, Yana, Bovo, Samuele, Martelli, Pier L, Babbi, Giulia, Di Lena, Pietro, Casadio, Rita, Edwards, Matthew D, Gifford, David K, Jones, David T, Sundaram, Laksshman, Bhat, Rajendra, Li, Xiaolin, Pal, Lipika R., Kundu, Kunal, Yin, Yizhou, Moult, John, Jiang, Yuxiang, Pejaver, Vikas, Pagel, Kymberleigh A., Li, Biao, Mooney, Sean D., Radivojac, Predrag, Shah, Sohela, Carraro, Marco, Gasparini, Alessandra, Leonardi, Emanuela, Giollo, Manuel, Ferrari, Carlo, Tosatto, Silvio C E, Bachar, Eran, Azaria, Johnathan R., Ofran, Yanay, Unger, Ron, Niroula, Abhishek, Vihinen, Mauno, Chang, Billy, Wang, Maggie H, Franke, Andre, Petersen, Britt-Sabina, Pirooznia, Mehdi, Zandi, Peter, McCombie, Richard, Potash, James B, Altman, Russ, Klein, Teri E., Hoskins, Roger, Repo, Susanna, Brenner, Steve E, and Morgan, Alexander A
- Abstract
Precision medicine aims to predict a patient's disease risk and best therapeutic options by using that individual's genetic sequencing data. The Critical Assessment of Genome Interpretation (CAGI) is a community experiment consisting of genotype–phenotype prediction challenges; participants build models, undergo assessment, and share key findings. For CAGI 4, three challenges involved using exome-sequencing data: Crohn's disease, bipolar disorder, and warfarin dosing. Previous CAGI challenges included prior versions of the Crohn's disease challenge. Here, we discuss the range of techniques used for phenotype prediction as well as the methods used for assessing predictive models. Additionally, we outline some of the difficulties associated with making predictions and evaluating them. The lessons learned from the exome challenges can be applied to both research and clinical efforts to improve phenotype prediction from genotype. In addition, these challenges serve as a vehicle for sharing clinical and research exome data in a secure manner with scientists who have a broad range of expertise, contributing to a collaborative effort to advance our understanding of genotype–phenotype relationships.
- Published
- 2021
16. DMAPS: a database of multiple alignments for protein structures
- Author
-
Guda, Chittibabu, Pal, Lipika R., and Shindyalov, Ilya N.
- Published
- 2006
17. Genetic basis of common human disease: insight into the role of nonsynonymous SNPs from genome-wide association studies
- Author
-
Pal, Lipika R and Moult, John
- Published
- 2011
- Full Text
- View/download PDF
18. Harnessing formal concepts of biological mechanism to analyze human disease
- Author
-
Darden, Lindley, Kundu, Kunal, Pal, Lipika R., and Moult, John
- Subjects
0301 basic medicine ,Biomedical Research ,Heredity ,Databases, Factual ,Crohn's Disease ,Ignorance ,Notation ,Infographics ,Human disease ,0302 clinical medicine ,Medicine and Health Sciences ,Disease ,Biology (General) ,Physiological Phenomena ,Biological Phenomena ,media_common ,0303 health sciences ,Ecology ,Representation (systemics) ,Genomics ,Ambiguity ,Classification ,Phenotypes ,Computational Theory and Mathematics ,Mechanism (philosophy) ,Modeling and Simulation ,030220 oncology & carcinogenesis ,Perspective ,Identification (biology) ,Graphs ,Computer and Information Sciences ,QH301-705.5 ,media_common.quotation_subject ,Systems biology ,Immunology ,Gastroenterology and Hepatology ,Autoimmune Diseases ,Molecular Genetics ,Cellular and Molecular Neuroscience ,03 medical and health sciences ,Genome-Wide Association Studies ,Genetics ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Philosophy of science ,Complex Traits ,Data Visualization ,Inflammatory Bowel Disease ,Computational Biology ,Biology and Life Sciences ,Human Genetics ,Genome Analysis ,Data science ,030104 developmental biology ,Genetic Loci ,Genetics of Disease ,Clinical Immunology ,Clinical Medicine - Abstract
Mechanism is a widely used concept in biology: in 2017 more than 10% of PubMed abstracts used the term. Thus, searching for and reasoning about mechanisms is fundamental to much of biomedical research, but until now there has been almost no computational infrastructure for this purpose. Recent work in the philosophy of science has explored the central role that the search for mechanistic accounts of biological phenomena plays in biomedical research, providing a conceptual basis for representing and analyzing biological mechanism. The foundational categories for components of mechanisms - entities and activities - guide the development of general, abstract types of biological mechanism parts. Building on that analysis, we have developed a formal framework for describing and representing biological mechanism, MecCog, and applied it to describing mechanisms underlying human genetic disease. Mechanisms are depicted using a graphical notation. Key features are assignment of mechanism components to stages of biological organization and classes; visual representation of uncertainty, ignorance, and ambiguity; and tight integration with literature sources. The MecCog framework facilitates analysis of many aspects of disease mechanism, including the prioritization of future experiments, probing of gene-drug and gene-environment interactions, identification of possible new drug targets, personalized drug choice, analysis of non-linear interactions between relevant genetic loci, and classification of disease based on mechanism.
- Published
- 2018
- Full Text
- View/download PDF
19. Tracing the origin of functional and conserved domains in the human proteome: implications for protein evolution at the modular level
- Author
-
Guda Chittibabu and Pal Lipika R
- Subjects
Evolution ,QH359-425 - Abstract
Abstract Background The functional repertoire of the human proteome is an incremental collection of functions accomplished by protein domains evolved along the Homo sapiens lineage. Therefore, knowledge on the origin of these functionalities provides a better understanding of the domain and protein evolution in human. The lack of proper comprehension about such origin has impelled us to study the evolutionary origin of human proteome in a unique way as detailed in this study. Results This study reports a unique approach for understanding the evolution of human proteome by tracing the origin of its constituting domains hierarchically, along the Homo sapiens lineage. The uniqueness of this method lies in subtractive searching of functional and conserved domains in the human proteome resulting in higher efficiency of detecting their origins. From these analyses the nature of protein evolution and trends in domain evolution can be observed in the context of the entire human proteome data. The method adopted here also helps delineate the degree of divergence of functional families occurred during the course of evolution. Conclusion This approach to trace the evolutionary origin of functional domains in the human proteome facilitates better understanding of their functional versatility as well as provides insights into the functionality of hypothetical proteins present in the human proteome. This work elucidates the origin of functional and conserved domains in human proteins, their distribution along the Homo sapiens lineage, occurrence frequency of different domain combinations and proteome-wide patterns of their distribution, providing insights into the evolutionary solution to the increased complexity of the human proteome.
- Published
- 2006
- Full Text
- View/download PDF
20. Harnessing formal concepts of biological mechanism to analyze human disease
- Author
-
Darden, Lindley, primary, Kundu, Kunal, additional, Pal, Lipika R., additional, and Moult, John, additional
- Published
- 2018
- Full Text
- View/download PDF
21. Insights from GWAS: emerging landscape of mechanisms underlying complex trait disease
- Author
-
Pal, Lipika R, Pal, Lipika R, Yu, Chen-Hsin, Mount, Stephen M, Moult, John, Pal, Lipika R, Pal, Lipika R, Yu, Chen-Hsin, Mount, Stephen M, and Moult, John
- Abstract
There are now over 2000 loci in the human genome where genome wide association studies (GWAS) have found one or more SNPs to be associated with altered risk of a complex trait disease. At each of these loci, there must be some molecular level mechanism relevant to the disease. What are these mechanisms and how do they contribute to disease? Here we consider the roles of three primary mechanism classes: changes that directly alter protein function (missense SNPs), changes that alter transcript abundance as a consequence of variants close-by in sequence, and changes that affect splicing. Missense SNPs are divided into those predicted to have a high impact on in vivo protein function, and those with a low impact. Splicing is divided into SNPs with a direct impact on splice sites, and those with a predicted effect on auxiliary splicing signals. The analysis was based on associations found for seven complex trait diseases in the classic Wellcome Trust Case Control Consortium (WTCCC1) GWA study and subsequent studies and meta-analyses, collected from the GWAS catalog. Linkage disequilibrium information was used to identify possible candidate SNPs for involvement in disease mechanism in each of the 356 loci associated with these seven diseases. With the parameters used, we find that 76% of loci have at least of these mechanisms. Overall, except for the low incidence of direct impact on splice sites, the mechanisms are found at similar frequencies, with changes in transcript abundance the most common. But the distribution of mechanisms over diseases varies markedly, as does the fraction of loci with assigned mechanisms. Many of the implicated proteins have previously been suggested as relevant, but the specific mechanism assignments are new. In addition, a number of new disease relevant proteins are proposed. The high fraction of GWAS loci with proposed mechanisms suggests that these classes of mechanism play a major role. Other mechanism types, such as variants affecting e
- Published
- 2015
22. Lessons from the CAGI-4 Hopkins clinical panel challenge.
- Author
-
Chandonia, John-Marc, Chandonia, John-Marc, Adhikari, Aashish, Carraro, Marco, Chhibber, Aparna, Cutting, Garry R, Fu, Yao, Gasparini, Alessandra, Jones, David T, Kramer, Andreas, Kundu, Kunal, Lam, Hugo YK, Leonardi, Emanuela, Moult, John, Pal, Lipika R, Searls, David B, Shah, Sohela, Sunyaev, Shamil, Tosatto, Silvio CE, Yin, Yizhou, Buckley, Bethany A, Chandonia, John-Marc, Chandonia, John-Marc, Adhikari, Aashish, Carraro, Marco, Chhibber, Aparna, Cutting, Garry R, Fu, Yao, Gasparini, Alessandra, Jones, David T, Kramer, Andreas, Kundu, Kunal, Lam, Hugo YK, Leonardi, Emanuela, Moult, John, Pal, Lipika R, Searls, David B, Shah, Sohela, Sunyaev, Shamil, Tosatto, Silvio CE, Yin, Yizhou, and Buckley, Bethany A
- Abstract
The CAGI-4 Hopkins clinical panel challenge was an attempt to assess state-of-the-art methods for clinical phenotype prediction from DNA sequence. Participants were provided with exonic sequences of 83 genes for 106 patients from the Johns Hopkins DNA Diagnostic Laboratory. Five groups participated in the challenge, predicting both the probability that each patient had each of the 14 possible classes of disease, as well as one or more causal variants. In cases where the Hopkins laboratory reported a variant, at least one predictor correctly identified the disease class in 36 of the 43 patients (84%). Even in cases where the Hopkins laboratory did not find a variant, at least one predictor correctly identified the class in 39 of the 63 patients (62%). Each prediction group correctly diagnosed at least one patient that was not successfully diagnosed by any other group. We discuss the causal variant predictions by different groups and their implications for further development of methods to assess variants of unknown significance. Our results suggest that clinically relevant variants may be missed when physicians order small panels targeted on a specific phenotype. We also quantify the false-positive rate of DNA-guided analysis in the absence of prior phenotypic indication.
- Published
- 2017
23. The Product Guides the Process: Discovering Disease Mechanisms
- Author
-
Darden, Lindley, Pal, Lipika R., Kundu, Kunal, Moult, John, Darden, Lindley, Pal, Lipika R., Kundu, Kunal, and Moult, John
- Abstract
The nature of the product to be discovered guides the reasoning to discover it. Biologists and medical researchers often search for mechanisms. The "new mechanistic philosophy of science" provides resources about the nature of biological mechanisms that aid the discovery of mechanisms. Here, we apply these resources to the discovery of mechanisms in medicine. A new diagrammatic representation of a disease mechanism chain indicates both what is known and, most significantly, what is not known at a given time, thereby guiding the researcher and collaborators in discovery. Mechanisms of genetic diseases provide the examples.
- Published
- 2017
24. Performance of in silico tools for the evaluation of p16INK4a (CDKN2A) variants in CAGI
- Author
-
Carraro, Marco, Minervini, Giovanni, Giollo, Manuel, Bromberg, Yana, Capriotti, Emidio, Casadio, Rita, Dunbrack, Roland, Elefanti, Lisa, Fariselli, Pietro, Ferrari, Carlo, Gough, Julian, Katsonis, Panagiotis, Leonardi, Emanuela, Lichtarge, Olivier, Menin, Chiara, Martelli, Pier Luigi, Niroula, Abhishek, Pal, Lipika R., Repo, Susanna, Scaini, Maria Chiara, Vihinen, Mauno, Wei, Qiong, Xu, Qifang, Yang, Yuedong, Yin, Yizhou, Zaucha, Jan, Zhao, Huiying, Zhou, Yaoqi, Brenner, Steven E, Moult, John, Tosatto, Silvio C.E., Carraro, Marco, Minervini, Giovanni, Giollo, Manuel, Bromberg, Yana, Capriotti, Emidio, Casadio, Rita, Dunbrack, Roland, Elefanti, Lisa, Fariselli, Pietro, Ferrari, Carlo, Gough, Julian, Katsonis, Panagiotis, Leonardi, Emanuela, Lichtarge, Olivier, Menin, Chiara, Martelli, Pier Luigi, Niroula, Abhishek, Pal, Lipika R., Repo, Susanna, Scaini, Maria Chiara, Vihinen, Mauno, Wei, Qiong, Xu, Qifang, Yang, Yuedong, Yin, Yizhou, Zaucha, Jan, Zhao, Huiying, Zhou, Yaoqi, Brenner, Steven E, Moult, John, and Tosatto, Silvio C.E.
- Abstract
Correct phenotypic interpretation of variants of unknown significance for cancer-associated genes is a diagnostic challenge as genetic screenings gain in popularity in the next-generation sequencing era. The Critical Assessment of Genome Interpretation (CAGI) experiment aims to test and define the state of the art of genotype-phenotype interpretation. Here, we present the assessment of the CAGI p16INK4a challenge. Participants were asked to predict the effect on cellular proliferation of 10 variants for the p16INK4a tumor suppressor, a cyclin-dependent kinase inhibitor encoded by the CDKN2A gene. Twenty-two pathogenicity predictors were assessed with a variety of accuracy measures for reliability in a medical context. Different assessment measures were combined in an overall ranking to provide more robust results. The R scripts used for assessment are publicly available from a GitHub repository for future use in similar assessment exercises. Despite a limited test-set size, our findings show a variety of results, with some methods performing significantly better. Methods combining different strategies frequently outperform simpler approaches. The best predictor, Yang&Zhou lab, uses a machine learning method combining an empirical energy function measuring protein stability with an evolutionary conservation term. The p16INK4a challenge highlights how subtle structural effects can neutralize otherwise deleterious variants.
- Published
- 2017
25. Consensus Genome-Wide Expression Quantitative Trait Loci and Their Relationship with Human Complex Trait Disease
- Author
-
Yu, Chen-Hsin, primary, Pal, Lipika R., additional, and Moult, John, additional
- Published
- 2016
- Full Text
- View/download PDF
26. Insights from GWAS: emerging landscape of mechanisms underlying complex trait disease
- Author
-
Pal, Lipika R, primary, Yu, Chen-Hsin, additional, Mount, Stephen M, additional, and Moult, John, additional
- Published
- 2015
- Full Text
- View/download PDF
27. Tracing the origin of functional and conserved domains in the human proteome: implications for protein evolution at the modular level
- Author
-
Pal, Lipika R and Guda, Chittibabu
- Subjects
Evolution, Molecular ,Proteomics ,Epidermal Growth Factor ,Evolution ,QH359-425 ,Animals ,Computational Biology ,Humans ,Databases, Protein ,Biological Evolution ,Research Article ,Protein Structure, Tertiary - Abstract
Background The functional repertoire of the human proteome is an incremental collection of functions accomplished by protein domains evolved along the Homo sapiens lineage. Therefore, knowledge on the origin of these functionalities provides a better understanding of the domain and protein evolution in human. The lack of proper comprehension about such origin has impelled us to study the evolutionary origin of human proteome in a unique way as detailed in this study. Results This study reports a unique approach for understanding the evolution of human proteome by tracing the origin of its constituting domains hierarchically, along the Homo sapiens lineage. The uniqueness of this method lies in subtractive searching of functional and conserved domains in the human proteome resulting in higher efficiency of detecting their origins. From these analyses the nature of protein evolution and trends in domain evolution can be observed in the context of the entire human proteome data. The method adopted here also helps delineate the degree of divergence of functional families occurred during the course of evolution. Conclusion This approach to trace the evolutionary origin of functional domains in the human proteome facilitates better understanding of their functional versatility as well as provides insights into the functionality of hypothetical proteins present in the human proteome. This work elucidates the origin of functional and conserved domains in human proteins, their distribution along the Homo sapiens lineage, occurrence frequency of different domain combinations and proteome-wide patterns of their distribution, providing insights into the evolutionary solution to the increased complexity of the human proteome.
- Published
- 2006
28. Protein Characterization of a Candidate Mechanism SNP for Crohn's Disease: The Macrophage Stimulating Protein R689C Substitution
- Author
-
Gorlatova, Natalia, primary, Chao, Kinlin, additional, Pal, Lipika R., additional, Araj, Rawan Hanna, additional, Galkin, Andrey, additional, Turko, Illarion, additional, Moult, John, additional, and Herzberg, Osnat, additional
- Published
- 2011
- Full Text
- View/download PDF
29. A Top-Down Approach to Infer and Compare Domain-Domain Interactions across Eight Model Organisms
- Author
-
Guda, Chittibabu, primary, King, Brian R., additional, Pal, Lipika R., additional, and Guda, Purnima, additional
- Published
- 2009
- Full Text
- View/download PDF
30. Predicting venous thromboembolism risk from exomes in the Critical Assessment of Genome Interpretation (CAGI) challenges
- Author
-
Moses Stamboulian, Rita Casadio, Rajgopal Srinivasan, Emidio Capriotti, Predrag Radivojac, Yana Bromberg, Sadhna Rana, Sean D. Mooney, Castrense Savojardo, Russ B. Altman, Yanran Wang, Panagiostis Katsonis, Steven E. Brenner, Yuxiang Jiang, Roxana Daneshjou, Kymberleigh A. Pagel, Samuele Bovo, John Moult, Gregory McInnes, Lipika R. Pal, Olivier Lichtarge, Pier Luigi Martelli, McInnes, Gregory, Daneshjou, Roxana, Katsonis, Panagiosti, Lichtarge, Olivier, Srinivasan, Raj G, Rana, Sadhna, Radivojac, Predrag, Mooney, Sean D, Pagel, Kymberleigh A, Stamboulian, Mose, Jiang, Yuxiang, Capriotti, Emidio, Wang, Yanran, Bromberg, Yana, Bovo, Samuele, Savojardo, Castrense, Martelli, Pier Luigi, Casadio, Rita, Pal, Lipika R, Moult, John, Brenner, Steven, and Altman, Russ
- Subjects
Male ,medicine.medical_specialty ,venous thromboembolism ,Disease ,Biology ,Genome ,Article ,03 medical and health sciences ,Exome Sequencing ,Genetics ,medicine ,Cluster Analysis ,Humans ,Genetic Predisposition to Disease ,cardiovascular diseases ,Exome ,Allele frequency ,Genetics (clinical) ,Exome sequencing ,030304 developmental biology ,0303 health sciences ,030305 genetics & heredity ,Confounding ,Warfarin ,Computational Biology ,prediction challenge ,Congresses as Topic ,equipment and supplies ,machine learning ,ROC Curve ,phenotype prediction ,Family medicine ,Female ,Venous thromboembolism ,exome ,Unsupervised Machine Learning ,medicine.drug - Abstract
Genetics play a key role in venous thromboembolism (VTE) risk, however established risk factors in European populations do not translate to individuals of African descent because of the differences in allele frequencies between populations. As part of the fifth iteration of the Critical Assessment of Genome Interpretation, participants were asked to predict VTE status in exome data from African American subjects. Participants were provided with 103 unlabeled exomes from patients treated with warfarin for non-VTE causes or VTE and asked to predict which disease each subject had been treated for. Given the lack of training data, many participants opted to use unsupervised machine learning methods, clustering the exomes by variation in genes known to be associated with VTE. The best performing method using only VTE related genes achieved an area under the ROC curve of 0.65. Here, we discuss the range of methods used in the prediction of VTE from sequence data and explore some of the difficulties of conducting a challenge with known confounders. In addition, we show that an existing genetic risk score for VTE that was developed in European subjects works well in African Americans.
- Published
- 2019
- Full Text
- View/download PDF
31. Working towards precision medicine: predicting phenotypes from exomes in the Critical Assessment of Genome Interpretation (CAGI) challenges
- Author
-
Predrag Radivojac, Yanran Wang, Kunal Kundu, Maggie Haitian Wang, Laksshman Sundaram, Pier Luigi Martelli, Sohela Shah, Steven E. Brenner, Emanuela Leonardi, Yuxiang Jiang, Roxana Daneshjou, Mehdi Pirooznia, Marco Carraro, Rita Casadio, Biao Li, Giulia Babbi, Peter P. Zandi, John Moult, Silvio C. E. Tosatto, Andre Franke, Yanay Ofran, James B. Potash, David T. Jones, Mauno Vihinen, Billy Chang, Sean D. Mooney, Pietro Di Lena, Roger A. Hoskins, Russ B. Altman, David K. Gifford, Rajendra Rana Bhat, Kymberleigh A. Pagel, Carlo Ferrari, Yana Bromberg, Susanna Repo, Britt-Sabina Petersen, Xiaolin Li, Yizhou Yin, Alexander A. Morgan, Teri E. Klein, Lipika R. Pal, Ron Unger, Samuele Bovo, Abhishek Niroula, Richard W. McCombie, Vikas Pejaver, Eran Bachar, Matthew D. Edwards, Alessandra Gasparini, Johnathan Roy Azaria, Manuel Giollo, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Daneshjou, Roxana, Wang, Yanran, Bromberg, Yana, Bovo, Samuele, Martelli, Pier L, Babbi, Giulia, Pietro Di, Lena, Casadio, Rita, Edwards, Matthew, Gifford, David, Jones, David T, Sundaram, Laksshman, Bhat, Rajendra Rana, Xiaolin, Li, Pal, Lipika R., Kundu, Kunal, Yin, Yizhou, Moult, John, Jiang, Yuxiang, Pejaver, Vika, Pagel, Kymberleigh A., Biao, Li, Mooney, Sean D., Radivojac, Predrag, Shah, Sohela, Carraro, Marco, Gasparini, Alessandra, Leonardi, Emanuela, Giollo, Manuel, Ferrari, Carlo, Tosatto, Silvio C E, Bachar, Eran, Azaria, Johnathan R., Ofran, Yanay, Unger, Ron, Niroula, Abhishek, Vihinen, Mauno, Chang, Billy, Wang, Maggie H, Franke, Andre, Petersen, Britt-Sabina, Pirooznia, Mehdi, Zandi, Peter, Mccombie, Richard, Potash, James B., Altman, Russ B., Klein, Teri E., Hoskins, Roger A., Repo, Susanna, Brenner, Steven E., and Morgan, Alexander A.
- Subjects
0301 basic medicine ,Bipolar Disorder ,Pharmacogenomic Variants ,Information Dissemination ,Disease ,Biology ,Bioinformatics ,Genome ,Whole Exome Sequencing ,Article ,03 medical and health sciences ,0302 clinical medicine ,Genetic ,Crohn Disease ,bipolar disorder ,Crohn's disease ,exomes ,machine learning ,phenotype prediction ,warfarin ,Genetics ,Genetics (clinical) ,Databases, Genetic ,Exome Sequencing ,Humans ,Genetic Predisposition to Disease ,Precision Medicine ,Exome ,Exome sequencing ,Interpretation (philosophy) ,Computational Biology ,Precision medicine ,Data science ,Phenotype ,030104 developmental biology ,Pharmacogenomic Variant ,Warfarin ,exome ,030217 neurology & neurosurgery ,Human - Abstract
Precision medicine aims to predict a patient's disease risk and best therapeutic options by using that individual's genetic sequencing data. The Critical Assessment of Genome Interpretation (CAGI) is a community experiment consisting of genotypeâphenotype prediction challenges; participants build models, undergo assessment, and share key findings. For CAGI 4, three challenges involved using exome-sequencing data: Crohn's disease, bipolar disorder, and warfarin dosing. Previous CAGI challenges included prior versions of the Crohn's disease challenge. Here, we discuss the range of techniques used for phenotype prediction as well as the methods used for assessing predictive models. Additionally, we outline some of the difficulties associated with making predictions and evaluating them. The lessons learned from the exome challenges can be applied to both research and clinical efforts to improve phenotype prediction from genotype. In addition, these challenges serve as a vehicle for sharing clinical and research exome data in a secure manner with scientists who have a broad range of expertise, contributing to a collaborative effort to advance our understanding of genotypeâphenotype relationships.
- Published
- 2017
32. Performance of in silico tools for the evaluation of p16INK4a (CDKN2A) variants in CAGI
- Author
-
Steven E. Brenner, Marco Carraro, Rita Casadio, Giovanni Minervini, Roland L. Dunbrack, Lisa Elefanti, Mauno Vihinen, Maria Chiara Scaini, Yizhou Yin, P. Fariselli, Chiara Menin, Yana Bromberg, Qiong Wei, Silvio C. E. Tosatto, Panagiotis Katsonis, Susanna Repo, John Moult, Yuedong Yang, Pier Luigi Martelli, Emidio Capriotti, Carlo Ferrari, Olivier Lichtarge, Qifang Xu, Lipika R. Pal, Emanuela Leonardi, Huiying Zhao, Jan Zaucha, Abhishek Niroula, Manuel Giollo, Yaoqi Zhou, Julian Gough, Carraro, Marco, Minervini, Giovanni, Giollo, Manuel, Bromberg, Yana, Capriotti, Emidio, Casadio, Rita, Dunbrack, Roland, Elefanti, Lisa, Fariselli, Pietro, Ferrari, Carlo, Gough, Julian, Katsonis, Panagioti, Leonardi, Emanuela, Lichtarge, Olivier, Menin, Chiara, Martelli, Pier Luigi, Niroula, Abhishek, Pal, Lipika R, Repo, Susanna, Scaini, Maria Chiara, Vihinen, Mauno, Wei, Qiong, Xu, Qifang, Yang, Yuedong, Yin, Yizhou, Zaucha, Jan, Zhao, Huiying, Zhou, Yaoqi, Brenner, Steven E, Moult, John, and Tosatto, Silvio C E
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Bioinformatics tools ,Pathogenicity predictors ,In silico ,Context (language use) ,Computational biology ,Biology ,Genome ,Article ,Machine Learning ,03 medical and health sciences ,CDKN2A ,Cell Line, Tumor ,Databases, Genetic ,Genetics ,medicine ,CAGI experiment ,cancer ,Cyclin-Dependent Kinase Inhibitor p18 ,Humans ,Computer Simulation ,Genetic Predisposition to Disease ,Genetics (clinical) ,Reliability (statistics) ,Variant interpretation ,Cyclin-Dependent Kinase Inhibitor p16 ,Cancer ,Cell Proliferation ,Bioinformatics and Systems Biology ,Protein Stability ,pathogenicity predictor ,variant interpretation ,Computational Biology ,Genetic Variation ,bioinformatics tools ,pathogenicity predictors ,Variety (cybernetics) ,030104 developmental biology ,Ranking ,bioinformatics tool ,Medical genetics ,Medical Genetics - Abstract
Correct phenotypic interpretation of variants of unknown significance for cancer-associated genes is a diagnostic challenge as genetic screenings gain in popularity in the next-generation sequencing era. The Critical Assessment of Genome Interpretation (CAGI) experiment aims to test and define the state of the art of genotype-phenotype interpretation. Here, we present the assessment of the CAGI p16INK4a challenge. Participants were asked to predict the effect on cellular proliferation of ten variants for the p16INK4a tumor suppressor, a cyclin-dependent kinase inhibitor encoded by the CDKN2A gene. Twenty-two pathogenicity predictors were assessed with a variety of accuracy measures for reliability in a medical context. Different assessment measures were combined in an overall ranking to provide more robust results. The R scripts used for assessment are publicly available from a GitHub repository for future use in similar assessment exercises. Despite a limited test-set size, our findings show a variety of results, with some methods performing significantly better. Methods combining different strategies frequently outperform simpler approaches. The best predictor, Yang&Zhou lab, uses a machine learning method combining an empirical energy function measuring protein stability with an evolutionary conservation term. The p16INK4a challenge highlights how subtle structural effects can neutralize otherwise deleterious variants. This article is protected by copyright. All rights reserved.
- Published
- 2017
33. Temporal genomic analysis of melanoma rejection identifies regulators of tumor immune evasion.
- Author
-
Cohen Shvefel S, Pai JA, Cao Y, Pal LR, Levy R, Yao W, Cheng K, Zemanek M, Bartok O, Weller C, Yin Y, Du PP, Yakubovich E, Orr I, Ben-Dor S, Oren R, Fellus-Alyagor L, Golani O, Goliand I, Ranmar D, Savchenko I, Ketrarou N, Schäffer AA, Ruppin E, Satpathy AT, and Samuels Y
- Abstract
Decreased intra-tumor heterogeneity (ITH) correlates with increased patient survival and immunotherapy response. However, even highly homogenous tumors may display variability in their aggressiveness, and how immunologic-factors impinge on their aggressiveness remains understudied. Here we studied the mechanisms responsible for the immune-escape of murine tumors with low ITH. We compared the temporal growth of homogeneous, genetically-similar single-cell clones that are rejected vs. those that are not-rejected after transplantation in-vivo using single-cell RNA sequencing and immunophenotyping. Non-rejected clones showed high infiltration of tumor-associated-macrophages (TAMs), lower T-cell infiltration, and increased T-cell exhaustion compared to rejected clones. Comparative analysis of rejection-associated gene expression programs, combined with in-vivo CRISPR knockout screens of candidate mediators, identified Mif (macrophage migration inhibitory factor) as a regulator of immune rejection. Mif knockout led to smaller tumors and reversed non-rejection-associated immune composition, particularly, leading to the reduction of immunosuppressive macrophage infiltration. Finally, we validated these results in melanoma patient data., Competing Interests: Conflict of interest: A.T.S. is a founder of Immunai and Cartography Biosciences and receives research funding from Astellas and Merck Research Laboratories. E.R. is a co-founder of MedAware Ltd and a co-founder (divested) and non-paid scientific consultant of Pangea Biomed. The other authors declare that they have no potential conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
34. Synthetic lethality-based prediction of anti-SARS-CoV-2 targets.
- Author
-
Pal LR, Cheng K, Nair NU, Martin-Sancho L, Sinha S, Pu Y, Riva L, Yin X, Schischlik F, Lee JS, Chanda SK, and Ruppin E
- Abstract
Novel strategies are needed to identify drug targets and treatments for the COVID-19 pandemic. The altered gene expression of virus-infected host cells provides an opportunity to specifically inhibit viral propagation via targeting the synthetic lethal (SL) partners of such altered host genes. Pursuing this antiviral strategy, here we comprehensively analyzed multiple in vitro and in vivo bulk and single-cell RNA-sequencing datasets of SARS-CoV-2 infection to predict clinically relevant candidate antiviral targets that are SL with altered host genes. The predicted SL-based targets are highly enriched for infected cell inhibiting genes reported in four SARS-CoV-2 CRISPR-Cas9 genome-wide genetic screens. Integrating our predictions with the results of these screens, we further selected a focused subset of 26 genes that we experimentally tested in a targeted siRNA screen using human Caco-2 cells. Notably, as predicted, knocking down these targets reduced viral replication and cell viability only under the infected condition without harming non-infected cells. Our results are made publicly available, to facilitate their in vivo testing and further validation.
- Published
- 2021
- Full Text
- View/download PDF
35. Genome-scale metabolic modeling reveals SARS-CoV-2-induced metabolic changes and antiviral targets.
- Author
-
Cheng K, Martin-Sancho L, Pal LR, Pu Y, Riva L, Yin X, Sinha S, Nair NU, Chanda SK, and Ruppin E
- Abstract
Tremendous progress has been made to control the COVID-19 pandemic caused by the SARS-CoV-2 virus. However, effective therapeutic options are still rare. Drug repurposing and combination represent practical strategies to address this urgent unmet medical need. Viruses, including coronaviruses, are known to hijack host metabolism to facilitate viral proliferation, making targeting host metabolism a promising antiviral approach. Here, we describe an integrated analysis of 12 published in vitro and human patient gene expression datasets on SARS-CoV-2 infection using genome-scale metabolic modeling (GEM), revealing complicated host metabolism reprogramming during SARS-CoV-2 infection. We next applied the GEM-based metabolic transformation algorithm to predict anti-SARS-CoV-2 targets that counteract the virus-induced metabolic changes. We successfully validated these targets using published drug and genetic screen data and by performing an siRNA assay in Caco-2 cells. Further generating and analyzing RNA-sequencing data of remdesivir-treated Vero E6 cell samples, we predicted metabolic targets acting in combination with remdesivir, an approved anti-SARS-CoV-2 drug. Our study provides clinical data-supported candidate anti-SARS-CoV-2 targets for future evaluation, demonstrating host metabolism-targeting as a promising antiviral strategy., Competing Interests: Conflict of interest The authors declare no competing interests.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.