Network


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

Hotspot


Dive into the research topics where Jishnu Das is active.

Publication


Featured researches published by Jishnu Das.


Nature Biotechnology | 2012

Three-dimensional reconstruction of protein networks provides insight into human genetic disease

Xiujuan Wang; Xiaomu Wei; Bram Thijssen; Jishnu Das; Steven M. Lipkin; Haiyuan Yu

To better understand the molecular mechanisms and genetic basis of human disease, we systematically examine relationships between 3,949 genes, 62,663 mutations and 3,453 associated disorders by generating a three-dimensional, structurally resolved human interactome. This network consists of 4,222 high-quality binary protein-protein interactions with their atomic-resolution interfaces. We find that in-frame mutations (missense point mutations and in-frame insertions and deletions) are enriched on the interaction interfaces of proteins associated with the corresponding disorders, and that the disease specificity for different mutations of the same gene can be explained by their location within an interface. We also predict 292 candidate genes for 694 unknown disease-to-gene associations with proposed molecular mechanism hypotheses. This work indicates that knowledge of how in-frame disease mutations alter specific interactions is critical to understanding pathogenesis. Structurally resolved interaction networks should be valuable tools for interpreting the wealth of data being generated by large-scale structural genomics and disease association studies.


Science | 2013

Integrative annotation of variants from 1092 humans: application to cancer genomics.

Ekta Khurana; Yao Fu; Vincenza Colonna; Xinmeng Jasmine Mu; Hyun Min Kang; Tuuli Lappalainen; Andrea Sboner; Lucas Lochovsky; Jieming Chen; Arif Harmanci; Jishnu Das; Alexej Abyzov; Suganthi Balasubramanian; Kathryn Beal; Dimple Chakravarty; Daniel Challis; Yuan Chen; Declan Clarke; Laura Clarke; Fiona Cunningham; Uday S. Evani; Paul Flicek; Robert Fragoza; Erik Garrison; Richard A. Gibbs; Zeynep H. Gümüş; Javier Herrero; Naoki Kitabayashi; Yong Kong; Kasper Lage

Introduction Plummeting sequencing costs have led to a great increase in the number of personal genomes. Interpreting the large number of variants in them, particularly in noncoding regions, is a current challenge. This is especially the case for somatic variants in cancer genomes, a large proportion of which are noncoding. Prioritization of candidate noncoding cancer drivers based on patterns of selection. (Step 1) Filter somatic variants to exclude 1000 Genomes polymorphisms; (2) retain variants in noncoding annotations; (3) retain those in “sensitive” regions; (4) prioritize those disrupting a transcription-factor binding motif and (5) residing near the center of a biological network; (6) prioritize ones in annotation blocks mutated in multiple cancer samples. Methods We investigated patterns of selection in DNA elements from the ENCODE project using the full spectrum of variants from 1092 individuals in the 1000 Genomes Project (Phase 1), including single-nucleotide variants (SNVs), short insertions and deletions (indels), and structural variants (SVs). Although we analyzed broad functional annotations, such as all transcription-factor binding sites, we focused more on highly specific categories such as distal binding sites of factor ZNF274. The greater statistical power of the Phase 1 data set compared with earlier ones allowed us to differentiate the selective constraints on these categories. We also used connectivity information between elements from protein-protein-interaction and regulatory networks. We integrated all the information on selection to develop a workflow (FunSeq) to prioritize personal-genome variants on the basis of their deleterious impact. As a proof of principle, we experimentally validated and characterized a few candidate variants. Results We identified a specific subgroup of noncoding categories with almost as much selective constraint as coding genes: “ultrasensitive” regions. We also uncovered a number of clear patterns of selection. Elements more consistently active across tissues and both maternal and paternal alleles (in terms of allele-specific activity) are under stronger selection. Variants disruptive because of mechanistic effects on transcription-factor binding (i.e. “motif-breakers”) are selected against. Higher network connectivity (i.e. for hubs) is associated with higher constraint. Additionally, many hub promoters and regulatory elements show evidence of recent positive selection. Overall, indels and SVs follow the same pattern as SNVs; however, there are notable exceptions. For instance, enhancers are enriched for SVs formed by nonallelic homologous recombination. We integrated these patterns of selection into the FunSeq prioritization workflow and applied it to cancer variants, because they present a strong contrast to inherited polymorphisms. In particular, application to ~90 cancer genomes (breast, prostate and medulloblastoma) reveals nearly a hundred candidate noncoding drivers. Discussion Our approach can be readily used to prioritize variants in cancer and is immediately applicable in a precision-medicine context. It can be further improved by incorporation of larger-scale population sequencing, better annotations, and expression data from large cohorts. Identifying Important Identifiers Each of us has millions of sequence variations in our genomes. Signatures of purifying or negative selection should help identify which of those variations is functionally important. Khurana et al. (1235587) used sequence polymorphisms from 1092 humans across 14 populations to identify patterns of selection, especially in noncoding regulatory regions. Noncoding regions under very strong negative selection included binding sites of some chromatin and general transcription factors (TFs) and core motifs of some important TF families. Positive selection in TF binding sites tended to occur in network hub promoters. Many recurrent somatic cancer variants occurred in noncoding regulatory regions and thus might indicate mutations that drive cancer. Regions under strong selection in the human genome identify noncoding regulatory elements with possible roles in disease. Interpreting variants, especially noncoding ones, in the increasing number of personal genomes is challenging. We used patterns of polymorphisms in functionally annotated regions in 1092 humans to identify deleterious variants; then we experimentally validated candidates. We analyzed both coding and noncoding regions, with the former corroborating the latter. We found regions particularly sensitive to mutations (“ultrasensitive”) and variants that are disruptive because of mechanistic effects on transcription-factor binding (that is, “motif-breakers”). We also found variants in regions with higher network centrality tend to be deleterious. Insertions and deletions followed a similar pattern to single-nucleotide variants, with some notable exceptions (e.g., certain deletions and enhancers). On the basis of these patterns, we developed a computational tool (FunSeq), whose application to ~90 cancer genomes reveals nearly a hundred candidate noncoding drivers.


BMC Systems Biology | 2012

HINT: High-quality protein interactomes and their applications in understanding human disease

Jishnu Das; Haiyuan Yu

BackgroundA global map of protein-protein interactions in cellular systems provides key insights into the workings of an organism. A repository of well-validated high-quality protein-protein interactions can be used in both large- and small-scale studies to generate and validate a wide range of functional hypotheses.ResultsWe develop HINT (http://hint.yulab.org) - a database of high-quality protein-protein interactomes for human, Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Oryza sativa. These were collected from several databases and filtered both systematically and manually to remove low-quality/erroneous interactions. The resulting datasets are classified by type (binary physical interactions vs. co-complex associations) and data source (high-throughput systematic setups vs. literature-curated small-scale experiments). We find strong sociological sampling biases in literature-curated datasets of small-scale interactions. An interactome without such sampling biases was used to understand network properties of human disease-genes - hubs are unlikely to cause disease, but if they do, they usually cause multiple disorders.ConclusionsHINT is of significant interest to researchers in all fields of biology as it addresses the ubiquitous need of having a repository of high-quality protein-protein interactions. These datasets can be utilized to generate specific hypotheses about specific proteins and/or pathways, as well as analyzing global properties of cellular networks. HINT will be regularly updated and all versions will be tracked.


Bioinformatics | 2013

INstruct: a database of high-quality 3D structurally resolved protein interactome networks

Michael J. Meyer; Jishnu Das; Xiujuan Wang; Haiyuan Yu

UNLABELLED INstruct is a database of high-quality, 3D, structurally resolved protein interactome networks in human and six model organisms. INstruct combines the scale of available high-quality binary protein interaction data with the specificity of atomic-resolution structural information derived from co-crystal evidence using a tested interaction interface inference method. Its web interface is designed to allow for flexible search based on standard and organism-specific protein and gene-naming conventions, visualization of protein architecture highlighting interaction interfaces and viewing and downloading custom 3D structurally resolved interactome datasets. AVAILABILITY INstruct is freely available on the web at http://instruct.yulab.org with all major browsers supported.


Bioinformatics | 2012

Genome-scale analysis of interaction dynamics reveals organization of biological networks

Jishnu Das; Jaaved Mohammed; Haiyuan Yu

Analyzing large-scale interaction networks has generated numerous insights in systems biology. However, such studies have primarily been focused on highly co-expressed, stable interactions. Most transient interactions that carry out equally important functions, especially in signal transduction pathways, are yet to be elucidated and are often wrongly discarded as false positives. Here, we revisit a previously described Smith-Waterman-like dynamic programming algorithm and use it to distinguish stable and transient interactions on a genomic scale in human and yeast. We find that in biological networks, transient interactions are key links topologically connecting tightly regulated functional modules formed by stable interactions and are essential to maintaining the integrity of cellular networks. We also perform a systematic analysis of interaction dynamics across different technologies and find that high-throughput yeast two-hybrid is the only available technology for detecting transient interactions on a large scale.


Cell | 2016

A Proteome-wide Fission Yeast Interactome Reveals Network Evolution Principles from Yeasts to Human.

Tommy V. Vo; Jishnu Das; Michael J. Meyer; Nicolas A. Cordero; Nurten Akturk; Xiaomu Wei; Benjamin Jung Fair; Andrew G. Degatano; Robert Fragoza; Lisa G. Liu; Akihisa Matsuyama; Michelle Trickey; Sachi Horibata; Andrew Grimson; Hiroyuki Yamano; Minoru Yoshida; Frederick P. Roth; Jeffrey A. Pleiss; Yu Xia; Haiyuan Yu

Here, we present FissionNet, a proteome-wide binary protein interactome for S. pombe, comprising 2,278 high-quality interactions, of which ∼ 50% were previously not reported in any species. FissionNet unravels previously unreported interactions implicated in processes such as gene silencing and pre-mRNA splicing. We developed a rigorous network comparison framework that accounts for assay sensitivity and specificity, revealing extensive species-specific network rewiring between fission yeast, budding yeast, and human. Surprisingly, although genes are better conserved between the yeasts, S. pombe interactions are significantly better conserved in human than in S. cerevisiae. Our framework also reveals that different modes of gene duplication influence the extent to which paralogous proteins are functionally repurposed. Finally, cross-species interactome mapping demonstrates that coevolution of interacting proteins is remarkably prevalent, a result with important implications for studying human disease in model organisms. Overall, FissionNet is a valuable resource for understanding protein functions and their evolution.


Human Mutation | 2014

Elucidating common structural features of human pathogenic variations using large-scale atomic-resolution protein networks

Jishnu Das; Hao Ran Lee; Adithya Sagar; Robert Fragoza; Jin Liang; Xiaomu Wei; Xiujuan Wang; Matthew Mort; Peter D. Stenson; David Neil Cooper; Haiyuan Yu

With the rapid growth of structural genomics, numerous protein crystal structures have become available. However, the parallel increase in knowledge of the functional principles underlying biological processes, and more specifically the underlying molecular mechanisms of disease, has been less dramatic. This notwithstanding, the study of complex cellular networks has made possible the inference of protein functions on a large scale. Here, we combine the scale of network systems biology with the resolution of traditional structural biology to generate a large‐scale atomic‐resolution interactome‐network comprising 3,398 interactions between 2,890 proteins with a well‐defined interaction interface and interface residues for each interaction. Within the framework of this atomic‐resolution network, we have explored the structural principles underlying variations causing human‐inherited disease. We find that in‐frame pathogenic variations are enriched at both the interface and in the interacting domain, suggesting that variations not only at interface “hot‐spots,” but in the entire interacting domain can result in alterations of interactions. Further, the sites of pathogenic variations are closely related to the biophysical strength of the interactions they perturb. Finally, we show that biochemical alterations consequent to these variations are considerably more disruptive than evolutionary changes, with the most significant alterations at the protein interaction interface.


Human Mutation | 2016

mutation3D: cancer gene prediction through atomic clustering of coding variants in the structural proteome

Michael J. Meyer; Ryan Lapcevic; Alfonso Romero; Mark Yoon; Jishnu Das; Juan Felipe Beltran; Matthew Mort; Peter D. Stenson; David Neil Cooper; Alberto Paccanaro; Haiyuan Yu

A new algorithm and Web server, mutation3D (http://mutation3d.org), proposes driver genes in cancer by identifying clusters of amino acid substitutions within tertiary protein structures. We demonstrate the feasibility of using a 3D clustering approach to implicate proteins in cancer based on explorations of single proteins using the mutation3D Web interface. On a large scale, we show that clustering with mutation3D is able to separate functional from nonfunctional mutations by analyzing a combination of 8,869 known inherited disease mutations and 2,004 SNPs overlaid together upon the same sets of crystal structures and homology models. Further, we present a systematic analysis of whole‐genome and whole‐exome cancer datasets to demonstrate that mutation3D identifies many known cancer genes as well as previously underexplored target genes. The mutation3D Web interface allows users to analyze their own mutation data in a variety of popular formats and provides seamless access to explore mutation clusters derived from over 975,000 somatic mutations reported by 6,811 cancer sequencing studies. The mutation3D Web interface is freely available with all major browsers supported.


PLOS Genetics | 2014

A Massively Parallel Pipeline to Clone DNA Variants and Examine Molecular Phenotypes of Human Disease Mutations

Xiaomu Wei; Jishnu Das; Robert Fragoza; Jin Liang; Francisco M. Bastos de Oliveira; Hao Ran Lee; Xiujuan Wang; Matthew Mort; Peter D. Stenson; David Neil Cooper; Steven M. Lipkin; Marcus B. Smolka; Haiyuan Yu

Understanding the functional relevance of DNA variants is essential for all exome and genome sequencing projects. However, current mutagenesis cloning protocols require Sanger sequencing, and thus are prohibitively costly and labor-intensive. We describe a massively-parallel site-directed mutagenesis approach, “Clone-seq”, leveraging next-generation sequencing to rapidly and cost-effectively generate a large number of mutant alleles. Using Clone-seq, we further develop a comparative interactome-scanning pipeline integrating high-throughput GFP, yeast two-hybrid (Y2H), and mass spectrometry assays to systematically evaluate the functional impact of mutations on protein stability and interactions. We use this pipeline to show that disease mutations on protein-protein interaction interfaces are significantly more likely than those away from interfaces to disrupt corresponding interactions. We also find that mutation pairs with similar molecular phenotypes in terms of both protein stability and interactions are significantly more likely to cause the same disease than those with different molecular phenotypes, validating the in vivo biological relevance of our high-throughput GFP and Y2H assays, and indicating that both assays can be used to determine candidate disease mutations in the future. The general scheme of our experimental pipeline can be readily expanded to other types of interactome-mapping methods to comprehensively evaluate the functional relevance of all DNA variants, including those in non-coding regions.


Science Signaling | 2013

Cross-Species Protein Interactome Mapping Reveals Species-Specific Wiring of Stress Response Pathways

Jishnu Das; Tommy V. Vo; Xiaomu Wei; Joseph C. Mellor; Virginia Tong; Andrew G. Degatano; Xiujuan Wang; Lihua Wang; Nicolas A. Cordero; Nathan Kruer-Zerhusen; Akihisa Matsuyama; Jeffrey A. Pleiss; Steven M. Lipkin; Minoru Yoshida; Frederick P. Roth; Haiyuan Yu

Comparing protein-interaction networks of two species of yeast suggests that network rewiring may enable environmental adaptation. Network Rewiring Protein-protein interaction networks represent one important aspect of cellular regulation. Although genomic conservation may indicate that networks should also be conserved, comparison of the stress response signaling networks in two different species of yeast suggests that the networks exhibit substantial rewiring of the interactions. Das et al. compared the interaction networks of proteins, classified as part of the stress response or involved in signal transduction, constructed from yeast two-hybrid data and found that the most orthologous proteins exhibited differences in their interacting partners. This network rewiring may be a mechanism by which different organisms adapt to diverse environments. The fission yeast Schizosaccharomyces pombe has more metazoan-like features than the budding yeast Saccharomyces cerevisiae, yet it has similarly facile genetics. We present a large-scale verified binary protein-protein interactome network, “StressNet,” based on high-throughput yeast two-hybrid screens of interacting proteins classified as part of stress response and signal transduction pathways in S. pombe. We performed systematic, cross-species interactome mapping using StressNet and a protein interactome network of orthologous proteins in S. cerevisiae. With cross-species comparative network studies, we detected a previously unidentified component (Snr1) of the S. pombe mitogen-activated protein kinase Sty1 pathway. Coimmunoprecipitation experiments showed that Snr1 interacted with Sty1 and that deletion of snr1 increased the sensitivity of S. pombe cells to stress. Comparison of StressNet with the interactome network of orthologous proteins in S. cerevisiae showed that most of the interactions among these stress response and signaling proteins are not conserved between species but are “rewired”; orthologous proteins have different binding partners in both species. In particular, transient interactions connecting proteins in different functional modules were more likely to be rewired than conserved. By directly testing interactions between proteins in one yeast species and their corresponding binding partners in the other yeast species with yeast two-hybrid assays, we found that about half of the interactions that are traditionally considered “conserved” form modified interaction interfaces that may potentially accommodate novel functions.

Collaboration


Dive into the Jishnu Das'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

Douglas A. Lauffenburger

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge